Sunteți pe pagina 1din 657

H O M E O S TAT IC C O N T R O L

O F B R A I N F U NC T I O N
H O M E O S TAT IC C O N T R OL
O F B R A I N F U NC T IO N

EDITED BY

D E T L E V B O I S O N Ph D
Director of Basic and Translational Research
Legacy Research Institute
Portland, OR

SUSA NA.M ASINO


Professor, Neuroscience and Psychology
Trinity College
Hartford, CT

1
1
Oxford University Press is a department of the Universityof
Oxford. It furthers the Universitys objective of excellence in research,
scholarship, and education by publishing worldwide.

OxfordNewYork
Auckland Cape Town Dar es Salaam Hong Kong Karachi
Kuala LumpurMadridMelbourneMexico CityNairobi
New DelhiShanghaiTaipeiToronto

With officesin
ArgentinaAustriaBrazilChileCzech RepublicFranceGreece
GuatemalaHungaryItalyJapanPolandPortugalSingapore
South KoreaSwitzerlandThailandTurkeyUkraineVietnam

Oxford is a registered trademark of Oxford UniversityPress


in the UK and certain other countries.

Published in the United States of Americaby


Oxford UniversityPress
198 Madison Avenue, NewYork, NY10016

Oxford University Press2016

All rights reserved. No part of this publication may be reproduced, storedin


a retrieval system, or transmitted, in any form or by any means, without theprior
permission in writing of Oxford University Press, or as expressly permitted bylaw,
by license, or under terms agreed with the appropriate reproduction rights organization.
Inquiries concerning reproduction outside the scope of the above should be senttothe
Rights Department, Oxford University Press, at the addressabove.

You must not circulate this work in any otherform


and you must impose this same condition on any acquirer.

Library of Congress Cataloging-in-PublicationData


Homeostatic control of brain function/edited by Detlev Boison and Susan Masino.
p.;cm.
Includes bibliographical references andindex.
ISBN 9780199322299 (alk.paper)
I. Boison, Detlev, editor. II. Masino, Susan, editor.
[DNLM:1.Brainphysiology. 2.Homeostasisphysiology. 3.Brain Chemistry. WL300]
QP376
612.82-dc23
2015011860

This material is not intended to be, and should not be considered, a substitute for medical or other professional advice.
Treatment for the conditions described in this material is highly dependent on the individual circumstances. And,
while this material is designed to offer accurate information with respect to the subject matter covered and to be current
as of the time it was written, research and knowledge about medical and health issues is constantly evolving and dose
schedules for medications are being revised continually, with new side effects recognized and accounted for regularly.
Readers must therefore always check the product information and clinical procedures with the most up-to-date
published product information and data sheets provided by the manufacturers and the most recent codes of conduct and
safety regulation. The publisher and the authors make no representations or warranties to readers, express or implied, as
to the accuracy or completeness of this material. Without limiting the foregoing, the publisher and the authors make no
representations or warranties as to the accuracy or efficacy of the drug dosages mentioned in the material. The authors
and the publisher do not accept, and expressly disclaim, any responsibility for any liability, loss or risk that may be
claimed or incurred as a consequence of the use and/or application of any of the contents of this material.

987654321
Printed in the United States of America
on acid-freepaper
CONTENTS

Contributors vii 7. Homeostatic Synaptic Scaling


Prologue xi at Central Synapses 108
Niraj S.Desai and
Abbreviations xiii
Elisabeth C.Walcott
8. Homeostatic Role of Heterosynaptic
PART I: Homeostatic Regulators: Molecules
Plasticity 124
andIons
Nicholas M.Bannon,
1. Microdynamics of Water and Ion Marina Chistiakova,
Homeostasis in the Brain:Role and Maxim Volgushev
of Aquaporins and Ion Channels of
Astroglial Cells 3 9. The Blood-Brain Barrier 143
Valentina Benfenati Yonatan Serlin, Alon Friedman,
and Stefano Ferroni and Uwe Heinemann

2. Homeostatic Control of Adenosine 10. Inflammation and Immunomodulation


Levels and Functions in the Brain 31 in Epilepsy and Its Comorbidities 155
Xuesong Chen, Mahmoud L.Soliman, Andrey Mazarati and
Liang Hui, and Jonathan D.Geiger Annamaria Vezzani
3. Glutamate Homeostasis as a Regulator 11. Neuroplasticity 175
of Neurotransmitter Recycling and HiroyukiOkuno
Synaptic Function 44
12. Epigenetics 187
Mary Ellen Kelly
Katja Kobow and Ingmar Blmcke
and Douglas a. Coulter
4. Homeostasis of Neuronal Excitability 13. Adult Neural Stem Cells and Brain
Via Synaptic and Intrinsic Inhibitory Homeostasis 202
Mechanisms 51 Ashok K.Shetty
Jochen Meier, Marcus Semtner,
and Jakob Wolfart
PART III: Homeostatic Manipulators:
Preventative and Restorative Opportunities
PART II: Homeostatic Control:Systems
andCells
14. Systems (Network) Pharmacology
for Brain Functionality Restoration 231
5. Role of Astrocytes in Sleep and Epilepsy 75 Doru Georg Margineanu
Jerome Clasadonte
and Philip G.Haydon 15. Ketogenic Diets for Neurological
Disorders 248
6. AstrocyteNeuron Interactions 98 Lindsey B.Gano, Manisha Patel,
Angus M. Brown and Bruce R.Ransom and JongM.Rho
vi Contents

16. Dietary Manipulations 271 25. Adenosine and Alzheimers


Athanasios Evangeliou and Disease:APossible Epigenetic Link 453
Martha Spilioti David Blum, Ursula S.Sandau,
Olivier Bousiges, Luisa V.Lopes,
17. Exercise 298
Vanessa Flaten, Emilie Faivre,
MarkusDworak Luc Bue, Anne-Laurence Boutillier,
18. Sleep 314 and DetlevBoison
Kristina Simeone, Chaz Johnson, 26. Brain Homeostasis and
Kaeli Samson, Harrison Roundtree, Parkinsons Disease 481
Tim Simeone, and LeilaTarokh DeeptiLall
19. Botanicals 333 27. Brain Homeostasis and Addiction 509
Dana Ekstein and Letisha R.Wyatt
Steven C.Schachter
28. Anxiety and Stress Disorders 535
20. The Role of Acupuncture in Nociception
Eva Mara Marco and
Homeostasis 341
Mara-Paz Viveros
Nanna Goldman, Takahiro Takano,
Benjamin T.Kress, and 29. Malignant Brain Cancer Management
Maiken Nedergaard with Metabolic Therapy 553
Thomas N.Seyfried and
21. Meditation 353
Purna Mukherjee
Galle Desbordes
30. Obesity and Diabetes:Nature,
22. Neurotrophic Regulation in Nurture, and Beyond 570
Neurorestoration of the Brain 371
CharlesMobbs
Caixin Su, Michel Rathbone,
and ShucuiJiang 31. Autism Spectrum Disorder
and Homeostasis 586
Susan A.Masino, Jessica A.Fortin,
PART IV: Homeostatic Therapies Michelle I.Murphy, Lisa Saa,
for Disease and Dysfunction and David N.Ruskin
23. Epilepsy 401
Index 611
DetlevBoison
24. Traumatic Brain Injury 420
Raj G.Kumar and Amy K.Wagner
C O N T R I BU T O R S

Nicholas M.Bannon LucBue


Department of Psychology Inserm
University of Connecticut Lille,France
Storrs,CT Universit deLille
Valentina Benfenati Jean-Pierre Aubert ResearchCentre
Consiglio Nazionale delle Ricerche Institut de Mdecine Prdictive
Istituto per la Sintesi Organica e la et de Recherche Thrapeutique
Fotoreattivit (ISOF) CHRU-Lille
Bologna, Italy Lille,France
DavidBlum XuesongChen
Inserm Department of Basic Biomedical Sciences
Universit deLille University of North Dakota School
Jean-Pierre Aubert Research Centre of Medicine and Health Sciences
Institut de Mdecine Prdictive Grand Forks,ND
et de Recherche Thrapeutique Marina Chistiakova
CHRU-Lille Department of Psychology
Lille,France University of Connecticut
Storrs,CT
Ingmar Blmcke
Department of Neuropathology Jerome Clasadonte
University Hospital Erlangen Department of Neuroscience
Erlangen, Germany Tufts University School of Medicine
Boston,MA
Olivier Bousiges
Laboratoire de Neurosciences Douglas A.Coulter
Cognitives et Adaptatives(LNCA) Departments of Pediatrics and
Universit de Strasbourg Neuroscience
Strasbourg,France Perleman School of Medicine
University of Pennsylvania and the
Anne-Laurence Boutillier
Division of Neurology
Laboratoire de Neurosciences
Childrens Hospital of Philadelphia
Cognitives et Adaptatives(LNCA)
Philadelphia,PA
Universit de Strasbourg
Strasbourg,France Niraj S.Desai
Center for Learning andMemory
Angus M.Brown The University of Texas atAustin
School of Life Sciences Austin,TX
University of Nottingham
Nottingham, England
viii Contributors

Galle Desbordes Lindsey B.Gano


Massachusetts General Hospital & Department of Pharmaceutical Sciences
Harvard MedicalSchool School of Pharmacy
Boston, MA University of Colorado
Denver,CO
MarkusDworak
Institute for Neuroscience Jonathan D.Geiger
German Sport University Cologne Department of Basic Biomedical Sciences
Cologne, Germany University of North Dakota School of Medicine
and Health Sciences
Dana Ekstein Grand Forks,ND
EpilepsyCenter
Department of Neurology Nanna Goldman
Hadassah-Hebrew University Medical Center Center for Translational Neuromedicine
Jerusalem,Israel University of Rochester MedicalCenter
Rochester,NY
Athanasios Evangeliou
Philip G.Haydon
Aristotle University of Thessaloniki
Department of Neuroscience
Department of PediatricsIV
Tufts University School of Medicine
Thessaloniki, Greece
Boston,MA
EmilieFaivre Uwe Heinemann
Universit deLille Institute of Neurophysiology and Neurocure
Jean-Pierre Aubert ResearchCentre ResearchCenter
Institut de Mdecine Prdictive et de Recherche Charit Universittsmedizin
Thrapeutique Berlin, Germany
CHRU-Lille
Lille,France LiangHui
Department of Basic Biomedical Sciences
Stefano Ferroni University of North Dakota School of Medicine
Department of Pharmacy and Biotechnology and Health Sciences
University of Bologna Grand Forks,ND
Bologna,Italy
ShucuiJiang
VanessaFlaten Department of Surgery (Neurosurgery,
Inserm Neurobiology)
Lille,France Hamilton NeuroRestorative Group(NRG)
Universit deLille McMaster University
Jean-Pierre Aubert ResearchCentre Health SciencesCentre
Institut de Mdecine Prdictive et de Recherche Hamilton, ON,Canada
Thrapeutique Chaz Johnson
CHRU-Lille Creighton University School of Medicine
Lille,France Department of Pharmacology
Jessica A.Fortin Omaha, NE
Neuroscience Program Mary EllenKelly
Trinity College Departments of Pediatrics and
Hartford,CT Neuroscience
Alon Friedman Perleman School of Medicine
Department of Physiology and Cell Biology University of Pennsylvania and the
Zlotowski Center for Neuroscience Division of Neurology
Ben-Gurion University of theNegev Childrens Hospital of Philadelphia
Beer-Sheva,Israel Philadelphia,PA
Department of Cognitive and Brain Sciences KatjaKobow
Zlotowski Center for Neuroscience Department of Neuropathology
Ben-Gurion University of theNegev University Hospital Erlangen
Beer-Sheva,Israel Erlangen, Germany
contributors ix

BenjaminKress Maiken Nedergaard


Center for Translational Neuromedicine Center for Translational Neuromedicine
University of Rochester MedicalCenter University of Rochester MedicalCenter
Rochester,NY Rochester,NY
Raj G.Kumar HiroyukiOkuno
Physical Medicine and Rehabilitation Medical InnovationCenter
University of Pittsburgh Graduate School of Medicine
Pittsburgh, PA Kyoto University
DeeptiLall Kyoto,Japan
Board of Governors Regenerative Medicine ManishaPatel
Institute Department of Pharmaceutical Sciences
Cedars-Sinai MedicalCenter School of Pharmacy
Lost Angeles,CA University of Colorado
Luisa V.Lopes Denver,CO
Instituto de Medicina Molecular Bruce R.Ransom
Lisbon, Portugal Department of Neurology
Eva MaraMarco University of Washington
Departamento de Fisiologa (Fisiologa Seattle,WA
AnimalII) Michel Rathbone
Facultad de Biologa Department of Medicine (Neurology,
Universidad Complutense Neuroscience)
Ciudad Universitaria Hamilton NeuroRestorative Group(NRG)
Madrid,Spain McMaster University
Doru Georg Margineanu Health SciencesCentre
Department of Neurosciences Hamilton, ON,Canada
Medicine and Pharmacy JongM.Rho
University ofMons Departments of Pediatrics and Clinical
Mons, Belgium Neurosciences
Andrey Mazarati Alberta Childrens Hospital Research Institute
Department of Pediatrics for Child and MaternalHealth
Childrens Discovery and Innovation Institute University of Calgary
David Geffen School of Medicine Calgary, Alberta,Canada
University of California Harrison Roundtree
Los Angeles,CA Creighton University School of Medicine
JochenMeier Department of Pharmacology
Technical University Braunschweig Omaha, NE
Zoological Institute David N.Ruskin
Division Cell Physiology Neuroscience Program
Braunschweig, Germany Psychology Department
CharlesMobbs Trinity College
Neuroscience, Endocrinology, and Geriatrics Hartford,CT
Mount Sinai School of Medicine LisaSaa
NewYork,NY Neuroscience Program
Purna Mukherjee Trinity College
Biology Department Hartford,CT
Boston College KaeliSamson
Chestnut Hill,MA UrsulaSandau
Michelle I.Murphy Legacy Research Institute
Neuroscience Program Portland,OR
Trinity College
Hartford,CT
x Contributors

Steven C.Schachter TakahiroTakano


Marcus Semtner Center for Translational Neuromedicine
Max Delbrck Center for Molecular Medicine University of Rochester MedicalCenter
Berlin, Germany Rochester,NY
YonatanSerlin LeilaTarokh
Department of Physiology and Cell Biology Creighton University School of Medicine
Zlotowski Center for Neuroscience Department of Pharmacology
Ben-Gurion University of theNegev Omaha, NE
Beer-Sheva,Israel Annamaria Vezzani
Thomas N.Seyfried IRCCSIstituto di Ricerche Farmacologiche
Biology Department MarioNegri
Boston College Milano,Italy
Chestnut Hill,MA Mara-Paz Viveros
Ashok K.Shetty Departamento de Fisiologa (Fisiologa
Institute for Regenerative Medicine AnimalII)
Texas A&M Health Science Center College of Facultad de Biologa
Medicine at Scott andWhite Universidad Complutense
Temple,TX Ciudad Universitaria
Research Service Madrid,Spain
Olin E.Teague Veterans MedicalCenter Maxim Volgushev
Central Texas Veterans Health careSystem Department of Psychology
Temple,TX University of Connecticut
Department of Molecular and Cellular Medicine Storrs,CT
Texas A&M Health Science Center College of
Medicine Amy K.Wagner
College Station,TX Physical Medicine and Rehabilitation
University of Pittsburgh
Kristina Simeone Pittsburgh,PA
Creighton University School of Medicine
Department of Pharmacology Elisabeth C.Walcott
Omaha, NE Department of Psychological Sciences
University of SanDiego
Tim Simeone San Diego,CA
Creighton University School of Medicine
Department of Pharmacology Jakob Wolfart
Omaha, NE Oscar Langendorff Institute of Physiology
University of Rostock
Mahmoud L.Soliman Rostock, Mecklenburg-Vorpommern, Germany
Department of Basic Biomedical Sciences
University of North Dakota School of Medicine Letisha R.Wyatt
and Health Sciences R.S. Dow Neurobiology Laboratories
Grand Forks,ND Legacy Research Institute
Portland,OR
Martha Spilioti
Aristotle University of Thessaloniki
Department of PediatricsIV
Thessaloniki, Greece
CaixinSu
Department of Surgery (Neurosurgery,
Neurobiology)
Hamilton NeuroRestorative Group(NRG)
McMaster University
Health SciencesCentre
Hamilton, ON,Canada
PROLOGUE

The brain is a complex and metabolically- brains significantly depends on the ways we live
demanding organ. The loss of homeostatic con- and what we eat or drink. In an age of an increas-
trol of brain function is expressed as a diverse ing personal and social burden of brain diseases
array of neurological disorders. When current such as Alzheimers or Parkinsons, the realiza-
therapies are inadequate or ineffective, restor- tion of homeostatic mechanisms that could be
ing or maintaining homeostatic functions can exploited to promote brain health is an emerging
improve brain health, regardless of the disorder. and timely topic of broad significance.
There is also evidence that targeting homeostatic The volume targets scientists, clinicians and
regulatory systems may delay or avert dysfunc- researchers at all levels as well as undergraduate,
tion. This book offers a broad overview of brain graduate and medical students, and a sophisti-
health and treatments for brain disease via home- cated public audience. The volume covers a very
ostatic control systems such as mitochondria, the broad topic which applies to a wide variety of
immune system, and epigenetic changes as well disciplines.
as via regulatory molecules such as ions, neuro- Understanding and promoting homeostatic
peptides, and neuromodulators. We also high- regulation of brain health is an urgent and
light emerging research on non-pharmaceutical emerging topic. At this time, highlighting this in
approaches such as botanicals, meditation, diet a specific volume which includes basic research
and exerciseall of which have been shown to and clinical content will help to coalesce this
impact brain function. Altogether we provide concept for therapeutic potential and incorpo-
high-quality science from molecules to disor- rate new ideas into existing paradigms. With this
ders, alongside emerging practical informa- concept in mind, the volume is unique in its col-
tion for improving homeostasis. This book will lection of chapters and sets the book apart from
convey the main message that the health of our otherworks.
A B B R E V I AT I O N S

5-HT 5-hydroxytryptamine (serotonin) IFN interferon


5-HTT serotonin transporter iGluRs ionotropic glutamate receptors
ACh acetylcholine IL interleukin
AD Alzheimers disease iNOS nitric oxide synthase
ADK adenosinekinase LB Lewybody
AMPA alpha-amino-3-hydroxy-5- L-DOPA L-3,4-dihydroxyphenylalanine
methyl-4-isoxazolepropionicacid LID levodopa-induced dyskinesia
AQP4 aquaporin-4 LRRK2 leucine-rich repeat kinase2
BBB blood-brain barrier LSD lysergic acid diethylamide
BDNF brain-derived neurotrophicfactor LTP long-term potentiation
CNS central nervoussystem MAO-B
COX-2 cyclooxygenase-2 inhibitors monoamine oxidase inhibitors
CPA conditioned place avoidance MDMA 3,4-methylenedioxy
CPP conditioned place preference methamphetamine
CPu caudate-putamen mGluR metabotropic glutamate receptor
CTA conditioned taste aversion MPTP 1-methyl-4-phenyl-1,2,3,6-
DA dopamine tetrahydropyridine
DAergic dopaminergic MR muscarinic receptor
DAT dopamine transporter NAC N-acetylcysteine
DHEAS dehydroepiandrosterone sulfate NAc nucleus accumbens
DR dopamine receptors nAch neuronal acetylcholine
EAAT excitatory amino acid transporters nAChR nicotinic receptors
EE environmental enrichment NE norepinephrine
EGF epidermal growthfactor NF-kB nuclear factor
EIF4G1 eukaryotic translation initiation kappa-light-chain-enhancer of
factor 4 gamma1 activated Bcells
ERK extracellular signal-regulated NGF nerve growthfactor
proteinkinase NIDA National Institute on DrugAbuse
FBXO7 F-box only protein7gene NMDA N-methyl-D-asparticacid
FDA Food and Drug Administration NTF neurotrophicfactor
GABA gamma aminobutyricacid OHDA 6-hydroxydopamine
GBA glucocerebrosidase PD Parkinsons disease
GDNF glial cell line-derived PF11 psuedogensenoside-F11
neurotrophicfactor PFC prefrontalcortex
GFAP glial fibrillary acidic protein PI-3K phosphatidylinositol-3-kinase
GLT-1 glutamate transporter-1 PINK1 phosphatase and tensin homolog
GPi globus pallidus (PTEN)-induced putative kinase1
HPA hypothalamic-pituitary-adrenalaxis PLA2G6 phospholipase A2, groupVI
ICSS intracranial self-stimulation PLC phospholipaseC
xiv Abbreviations

PPN pedunculopontine nucleus Tfam mitochondrial transcription factorA


PTZ pentylenetetrazol TGF transforming growth factor-alpha
ROS reactive oxygen species THC tetrahydrocannabinol
SE status epilepticus TLR Toll-like receptor
SN substantianigra TNF tumor necrosisfactor
SNCA alpha-synuclein TNF- tumor necrosis factor-alpha
SNpc substantia nigra pars compacta UPS ubiquitin-proteasomesystem
SPNs spiny projection neurons VPS35 vacuolar protein sorting35
SSRI selective serotonin reuptake inhibitor VTA ventral tegmentalarea
PARTI

Homeostatic Regulators

Molecules andIons
1
Microdynamics ofWater and Ion
Homeostasis intheBrain
Role of Aquaporins and Ion Channels of AstroglialCells

VA L E N T I N A B E N F E N AT I A N D S T E FA N O F E R R O N I

ASTROCYTES AND space (Abbott et al., 2006). The processes that


B R A I N H O M E O S TA S I S we define here as microdynamics, are mediated
Maintaining constancy in the internal medium by the so-called neurovascular unit, a modular
is also essential for the proper functioning of the dynamic ensemble formed by neurons, astroglial
central nervous system (CNS), whose activi- cells and vascular endothelia of the BBB. It is now
ties rely on the unequal distribution of solutes well known that the neurovascular unit is essen-
(organic molecules, ions, and neurotransmitters) tial for CNS physiology by providing and regu-
and water between the intra- and extracellular lating local blood supply and integrating CNS
compartments (Simard and Nedergaard, 2004). function (Iadecola and Nedergaard, 2007; Filosa
The brain is constrained by the rigidity of the and Iddings, 2013). In this scenario, astrocytes
skull, which can lead to a deleterious increase in are central players as several pieces of in vivo
intracranial pressure when brain swelling, initi- and in vitro evidence have established their piv-
ated by a pathological redistribution of water and otal function in homeostatic regulation of water
electrolytes in the brain parenckyma, occurs. The and ion concentrations, as well as in the control
mechanisms underlying the ion and water trans- of neurotransmitter dynamics in the interstitial
port across plasma-membrane of the cells that fluid (Anderson and Swanson, 2000; Kofuji and
face and delimit fluid-filled compartments of Newman, 2004; Simard and Nedergaard,2004).
the brain have a central role in brain physiology The term astrocyte (astroglial cell) was first
and pathophysiology and are becoming attrac- proposed by Michael von Lenhossek describing
tive pharmacological targets to counteract the cells with cylindrical processes departing from a
neuronal damage caused by the dysregulation defined round cell body, presenting a star-like
of brain homeostatic processes (Kimelberg and morphological phenotype (Somjen, 1988). The
Nedergaard,2010). complexity and heterogeneity of astrocytes were
Three physical barriers delimit and regulate then defined by their location in the white versus
the exchange between the microvasculature and grey matter, classifying them into fibrous and
the fluid compartments of the brain. Two of these protoplasmatic astroglial subtypes (Freeman,
are epithelia that separate the blood from the ven- 2010). Fibrous astrocytes are abundant in the
tricular cerebrospinal fluid (the choroid plexus), white matter. They display numerous filaments
and from the subarachnoid cerebrospinal fluid that stain with the glial fibrillary acidic protein,
(arachnoid epithelium). The third one (the brain which is the typical glial marker. Protoplasmic
blood barrier, BBB) is formed by the endothelial astrocytes populate the grey matter and have
cells lining brain microvessels and demarcates more irregular processes and few glial fila-
the boundary between the blood and the intersti- ments (Nedergaard et al., 2003). They contact
tial fluid of the cerebral tissue. The BBB enables and cover synapses by extending thousands of
a selective, dynamic bidirectional communica- thin processes, with typically only one or two
tion between the brain microenvironment and in contact with blood vessels or CNS bounda-
the blood, thereby tightly controlling the water ries (Bushong, 2002; Filosa and Iddings, 2013).
and osmolyte concentrations in the interstitial Two types of specialized astroglial cells are also
4 Part I: Homeostatic Regulators

recognized in the CNS: Bergmann glia and regulators of the brain function. The homeo-
Mller cells that are present primarily in the cer- static control is performed by astrocytes in a
ebellum and the retina, respectively. Bergmann stratified and ordered manner with dynamics
glia cell bodies lie within the Purkinje cell layer, involving cellular and molecular events (micro-
and their processes extend into the granule cell dynamics) with both processes occurring
layer to end at the pial surface (Hoogland and throughout the life of the brain, from its neu-
Kuhn, 2010). Retinal Mller cells are oriented rogenesis to development and aging (Molofsky
radially, spanning from the photoreceptor layer etal., 2012; Nagelhus etal., 2013). Astroglia are,
to the inner retinal surface (Reichenbach and in fact, intimately involved in CNS formation
Bringmann, 2013). as both stem cell elements and regulators of
Over recent years, glioanatomy studies based neurogenesis. During development, astrocytes
on electron microscopy, confocal imaging, and define the pathways regulating migration of
3D computational reconstruction, as well as neural cells and control synaptogenesis/synap-
two photon imaging analyses on enhanced tic pruning, thereby shaping microarchitecture
green-fluorescent protein-expressing astrocytes of the grey matter (structural homeostasis)
(Bushong et al., 2002; Nedergaard et al., 2003; (Nedergaard et al., 2003; Bernardinelli et al.,
Mathiisen et al., 2010), have provided indica- 2014). Astrocytes store glycogen and deliver
tion against the view of a random distribution glucose and metabolic substrates such as lactate
of astrocytes throughout the adult CNS. It is (metabolic homeostasis) to neurons (Pellerin
remarkable that the morphology of astrocytes etal., 2007; Stobart and Anderson, 2013). They
resembles more a polyhedron rather than a also control and maintain local ion and water
star-like shape, with major processes that ramify homeostasis, and provide clearance of cat-
into thinner ones and are uniformly distributed abolites and neurotransmitters/neuromodu-
in leaflet-like appendages through which each lators released during synaptic transmission,
astrocyte, filling a specific and limited volume which are sequestered through active uptake
and space, defines its own anatomical (functional) (microdynamic homeostasis) (Simard and
domain. Each protoplasmic astrocyte endfoot Nedergaard, 2004; Benfenati and Ferroni,
occupies its own district and covers all sur- 2010). Moreover, astrocytes regulate systemic
rounding neuronal elements, parcelling the grey homeostatic responses as they are also able to
matter into specific subdomains (Bushong etal., perceive systemic fluctuations in CO2 and pH
2002). Therefore, this highly and orderly organ- and variations in sodium (Na+) and water con-
ized astrocyte cytoarchitecture (Figure 1.1A) tents (osmolarity) and then elaborate the appro-
defines the precise modular geometry of the priate responses at molecular and organ levels
entire neuropil of the grey matter (Nedergaard (Simard and Nedergaard, 2004; Kimelberg and
etal., 2003; Kettenmann and Verkhratsky, 2008). Nedergaard,2010).
This picture is even more relevant considering To fully understand the homeostatic role of
that a loss of such a compartmentalized organi- astrocytes, one should refer to their compart-
zation is now believed to be involved in a variety mentalized and polyhedral morphology and
of brain pathologies (Seifert etal., 2006; Chvtal envision them as multifunctional units regu-
et al., 2008; Takano et al., 2009; Steinhauser lating separated fluid compartments through
et al., 2012). Astroglial microdomains are also various functional microdomains located in
highly organized and compartmentalized at the their process endfeet (Simard and Nedergaard,
interface with the blood vessels, with each astro- 2004; Benfenati and Ferroni, 2010). At the
cyte endfoot enveloping the endothelial cells in a neuronastrocyte interface the astroglial micro-
specific district, thus providing a contiguous but domain take ups and secretes ions, organic mol-
nonoverlapping sheath around the capillary bor- ecules, and water (Simard and Nedergaard, 2004;
dering (Simard etal.,2003). Kimelberg and Nedergaard, 2010). To ensure ion
The evidence on astroglial cytoarchitecture and water homeostasis of the interstitial fluid,
and distribution have supported for years the different processes occur at the endfeet facing the
plausible hypothesis of a nutritional and trophic vasculature and at those of astroglia contacting
role of astrocytes, as it was first proposed by the cells lining other fluid-filled compartments
Golgi (Somjen, 1988). Over the past decades, (Amiry-Moghaddam and Ottersen, 2003; Kofuji
however, this view has radically changed as and Newman, 2004; Simard and Nedergaard,
numerous studies have clearly shown that astro- 2004). Aprerequisite for their homeostatic func-
cytes also serve as metabolic and homeostatic tion is the abundant interastrocytic connection
Microdynamics of Water and Ion Homeostasis in the Brain 5

(a) (b)

FIGURE 1.1: Structural and functional relationships of astroglia in the regulation of brain
homeostasis. (A) Spatial interaction between astrocytes and neurons in the rat cortex. Neurons are labeled
with antibody to microtubule-associated protein 2 (MAP-2), whereas astrocytes are the cells expressing enhanced
green-fluorescent protein (eGFP). eGFP-positive astrocytic endfeet are abutting blood vessel. (B) Hypothetical
diagram of the microdynamics involved in astrocyte-mediated extracellular homeostasis. Astrocyte morphology
is characterized by an irregularly shaped cell body from which depart many leaflet-like processes. The endfeet of
the processes contact the neuronal soma (arrow a) and cover most synapses (arrow b) defining nonoverlapping
districts. In a given astrocyte other endfeet make contact with the surface of the brain or abut the blood vessels,
thereby forming a perivascular sheet (arrow c). Astrocytes are coupled by gap junctions (arrow d) that allow
synchronization of distal cell responses in the astroglial syncytium. The astroglial architecture is functional to
the homeostatic role of astrocytes, which is crucial for the maintenance of physiological neuronal activity. As a
consequence of the propagation of an action potential (1), neurotransmitters and ions are released in the inter-
synaptic space and perineuronal milieu (2). Astrocytes, by forming the neuro-glial unit, recover and maintain
the extracellular potassium (K+) and glutamate (Glu-) concentrations at optimal levels to ensure proper function
of the neuronal network (3):Astroglial Na+-coupled Glu- transporter (GLT-1) is responsible (in conjunction with
GLAST transporter) for Glu- uptake. Among the large variety of K+ channels expressed by astrocytes, the inwardly
rectifying K+ channel (Kir4.1) is crucially involved in local K+ uptake from the extracellular space. (4)Excess of
intracellular K+ is redistributed toward the vascular system, and gap junctions shunt K+ from perineuronal space
to distal vascular districts. (5)Finally, K+ is secreted at gliovascular interface mainly via Kir4.1, even though out-
wardly rectifying K+ channels (Kv) and calcium (Ca 2+)-activated K+ channel (BKCa) could also play a role. During
the spatial buffering of K+, the transient osmotic gradients created are rapidly counteracted by water flux. An
exclusive role for osmotic water movement is mediated by AQP4, which is largely distributed at astroglial endfeet
surrounding blood vessels and contacting the pial surface. The transport of chloride (Cl) ions is also critical for
the regulation of astroglial functions. The Na+-K+-2Cl- cotransporter and the Cl-/HCO3 anion exchanger (not
shown in the scheme) play a major role for pumping Cl into astrocyte. The role of Cl channels of the ClC fam-
ily that have been described in astrocytes is still unclear. Volume-regulated anion channels (VRACs) have been
identified in vitro and might contribute to cell volume homeostasis. However, whether they are also expressed and
operative in vivo is still unclear. The expression of the Ca 2+ permeable channel TRPV4 has been reported both in
situ and in vitro. For clarity, pumps and transporters that contribute to the transmembrane movement of ions have
not been included in the scheme.
Panel Amodified from Nedergaard etal. [2003], panel B modified from Benfenati and Ferroni [2010].

and coupling that allow astrocytes to act as a turn, the efficacy of the homeostatic processes
functional syncytium (Nagy and Rash, 2000). (Scemes and Giaume,2006).
This enables rapid and coordinated communi- It is widely accepted that astrocytes regu-
cation within the astroglial tissue increasing, in late the extracellular fluid homeostasis by using
6 Part I: Homeostatic Regulators

a variety of transmembrane proteins that drive pathological conditions such as spreading depres-
passive and active transport of ions, organic sion (Somjen, 2001). The principle mechanisms
osmolytes, and osmotically obliged water responsible for balancing [K+]o during neuronal
(Pasantes-Morales and Cruz-Rangel, 2010). activity are called K+ uptake and spatial K+ buff-
Envisaging the astrocyte as a multifunctional ering (Figure 1.2) (Odette and Newman,1988).
unit regulating separated fluid compartments, In the K+ uptake mechanism, the excess of
it is not surprising that ion and water channels [K ]o is cleared by the coordinated action
+

display a polarized distribution (Figure 1.1B) of Na+-K+-2Cl cotransporter (NKCC) and


and that, in order to mediate specific functional Na+/K+-ATPase pump (Walz and Hertz, 1982;
processes, they co-localize in cellular domains Walz and Hinks, 1985). Whereas both are likely to
(Benfenati and Ferroni, 2010). In the follow- contribute significantly to K+ uptake under physi-
ing sections, microdynamics regulating ion and ological [K+]o variations, when [K+]o increases
water homeostasis are described, addressing the above ceiling levels, as it occurs in ischemia and
specific ion and water channels involved and trauma, the NKCC would become predominant
the interplay of microdomains engaged in these (Walz and Hertz, 1984). In this context, it was
homeostatic processes. shown that the NKCC inhibitor bumetanide was
effective in limiting edema in ischemic condi-
MICRODY NAMICS tions in vivo (Yan etal., 2003), a result that was
R E G U L AT I N G also observed in NKCC1 knock-out (KO) mice
EXTR ACELLULAR (Chen et al., 2005). Simulated ischemic condi-
P O TA S S I U M tions in vitro also caused astrocyte swelling,
H O M E O S TA S I S which was blocked by bumetanide (Su et al.,
A fundamental homeostatic function of astro- 2002). Moreover, in vitro as well in vivo studies
cytes is the maintenance of the low extracellular on rat optical nerve indicated that the rise in [K+]o
concentration of potassium ion ([K+]o). In neu- induced astrocyte swelling, which was reversibly
rons the relative concentrations of K+ and Na+ depressed by furosemide that also inhibits NKCC
inside and outside the cell define the resulting (MacVicar et al., 2002). In contrast, available
chemical gradients across the plasma membrane, information suggests that Na+/K+-ATPase activ-
which are crucial to the genesis and propagation ity could play a prominent role in the clearance
of the action potentials. Since information pro- of the stimulus-evoked increment in [K+]o when
cessing through action potential firing is medi- its level is 1012 mM, as determined by the obser-
ated by Na+ influx and K+ efflux through the cell vation that under conditions of physiological
membrane, it follows that after neuronal activ- neuronal activity pharmacological inhibition of
ity, increments in [K+]o do occur and should be Na+/K+-ATPase caused a strong delay in K+
considered significant, if correlated to the lim- clearance (Ransom et al., 2000; DAmbrosio
ited volume of the extracellular space in the etal.,2002).
CNS (Nicholson and Sykov, 1998). If a rise in Another mechanism that appears to play a
[K+]o remained unbalanced, the neuronal rest- relevant role in [K+]o homeostasis under physi-
ing membrane potential would be altered and ological conditions is the so-called spatial K+
the activity of plasma membrane proteins form- buffering. This is a mechanism involved in
ing channels, receptors, and transporters would local [K+]o uptake and K+ dissipation through
be affected with consequent modifications of a the astroglial tissue, which is regulated by the
variety of neuronal processes, including synap- K+ equilibrium potential and the gradient in
tic transmission and electrogenic transport of syncytial membrane potential (Kofuji and
neurotransmitters, which would lead to a com- Newman, 2004). The first evidence supporting
promission of the functionality and integrity of the K+ spatial buffering was provided by Orkand
the CNS (Seifert etal., 2006; Takano etal., 2009; etal. (1966), demonstrating that stimulation of
Steinhauser etal., 2012). In the brain the [K+]o is the amphibian optic nerve led to slow depolari-
tightly controlled and regulated to be kept close zation and repolarization of the glial cells sur-
to the physiological value of 3 mM. However, in rounding the nonmyelinated axons. According
vivo studies have demonstrated that under physi- to the Orkand model (Orkand, 1986), at the
ological conditions characterized by high neu- neuronal site, where [K+]o is locally increased
ronal activity, [K+]o can transiently augment to by neuronal activity, the resting membrane
ceiling levels of 1012 mM (Kofuji and Newman, potential of astrocytes is negative with respect
2004)and reach values as high as 60 mM during to the Nernst equilibrium potential for K+.
Microdynamics of Water and Ion Homeostasis in the Brain 7

1 EK = Vm = 90 mV; [K+]o = 3 mM

2 3
[K+]i = 108 mM Spatial
Net
Uptake Buffer

[K+]o = 12 mM [K+]o = 4 mM
EK = Vm = 56 mV; [K+] o = 12 mM Na+
K+

K+
K+ [K+]i = 110 mM [K+]i = 108 mM
K+
Na+ K+
K+ Cl Na+
50 50 EK
EK = Vm = 56 mV K+ in
mV 70 mV 70 Vm K+ out

90 90
Distance Distance

FIGURE 1.2: Mechanisms of extracellular potassium homeostasis by the astroglial syncytium.


(1) Under conditions in which [K+]o is 3 mM the astroglial syncytium has a resting membrane potential (Vm)
close to the Nernst equilibrium potential for K+ (EK -90 mV). (2)K+ uptake mechanism:when [K+]o is increased
up to 12 mM as a consequence of neuronal activity, astrocytes accumulate [K+]o prevalently by the activity of a
Na+/K+ATPase and, to a lesser extent, by the Na+-K+-2Cl- cotransporter. Under these conditions the membrane
potential in the astroglial syncytium is estimated to be spatially uniform at a value close to that of EK of 56 mV.
(3)K+ spatial buffering mechanism:local increases of [K+]o produce a glial depolarization that spreads through the
glial syncytium. The local difference between the astrocyte Vm and EK causes the K+ inflow in regions of elevated
[K+]o and K+ outward efflux at distant regions. The result is a net flux of K+ away from the region where it is accu-
mulated extracellularly.The graphs show the distribution of EK and Vm as a function of distance along the astroglial
syncytium for the two mechanisms.
From Orkand,1986.

This difference drives the inflow of K+ into astro- K+ channels, gap junctions, and their interplay
cytes compartments exposed to the transient with the water channels aquaporins (AQPs)
[K+]o elevation, causing a local astrocytes depo- (Benfenati and Ferroni,2010).
larization that propagates through electrically
coupled astrocytes and allowing the movement Potassium Channels Involved
of K+ toward regions and districts where [K+]o inAstrocyte-Mediated
is lower. In certain CNS regions like the retina, Homeostasis ofExtracellular
K+ currents flow within single Mller glial cells Potassium
rather than through a cell network by a mecha- The use of tools and technologies in the field
nism called K+ siphoning (Newman etal., 1984). of molecular biology (cloning, expression, and
To satisfy the Orkand model, two conditions are mutagenesis), genetics (full and conditional
essential: (a) astroglial cells should be highly KO mice with cell-specific gene deletion), elec-
permeable to K+; and (b) astrocytes should be trophysiology (path-clamp), and microscopy
electrically coupled to form a syncytium in (immunogold electron microscopy, laser scan-
which K+ currents can traverse relatively long ning confocal microscopy) have enabled the
distances. Moreover, as recently pointed out, the acquisition of functional and molecular insights
overall mechanism implies osmotically driven into the channel proteins that play a role in astro-
water movement across the plasma membrane glial K+ homeostasis through the mechanism of
(Nagelhus et al., 2004). Several lines of in vivo spatial buffering. Astrocytes are highly permeable
evidence in the past decades have indicated to K+ owing to a variety of K+ channels expressed
that spatial buffering and siphoning depend on in their plasma membrane (Verkhratsky
proper distribution and function of astrocytic and Steinhauser, 2000). Inward rectifying
8 Part I: Homeostatic Regulators

K+ channels (Kir) underpin the dominant astro- expressed by hippocampal astroglia (Hibino etal.,
glial K+ conductance and they are the only K+ 2004). In Mller cells, expression of Kir2.1 was
channels to date for which clear physiological and shown to accompany that of Kir4.1 (Kofuji et al.,
pathological roles have been established (Horio, 2002). The functional relevance of this differential
2001; Olsen and Sontheimer, 2008). After being channel compositions is unknown.
cloned and studied in a heterologous expression The pivotal role of Kir4.1 in spatial buffer-
system for the first time (Kubo etal., 1993), over ing has been confirmed in experiments per-
20 genes are currently known to encode various formed in mice lacking the gene encoding for
Kir channel subunits (Hibino etal., 2010). Among Kir4.1 (KCNJ10). Genetic inactivation or condi-
the various subfamilies cloned at least five of tional deletion of Kir4.1 was shown to produce
these (Kir 2.1, 2.2, 2.3, 4.1, 5.1) have been iden- a strong depolarization of the resting mem-
tified in astrocytes (Higashi etal., 2001; Hibino brane potential of Mller cells (Kofuji et al.,
etal., 2004). Apeculiar biophysical feature of Kir 2000)and hippocampal astrocytes (Djukic etal.,
channels is their rectification property, which 2007; Chever et al., 2010). In Mller cells this
implies that at any voltage applied, K+ inward flow effect was accompanied by a 10-fold increase in
is greater than outward flux caused by an oppo- the membrane input resistance and an altered
site driving force (Nichols and Lopatin, 1997). response to light-evoked electro-retinogram,
This behavior depends on channel blockade by which is dependent on K+ influx (Kofuji et al.,
cytoplasmic magnesium (Mg2+) and polyamines, 2000). In hippocampal astrocytes the lack of
which limits K+ outflow (Matsuda, 1991; Lopatin Kir4.1 impaired the K+ and glutamate uptake
et al., 1994). Another typical feature of the Kir and increased short-term synaptic potentiation
channels is their dependence of gating on [K+]o, (Djukic et al., 2007). The role of Kir4.1 in [K+]o
which results in an increase in membrane K+ per- and glutamate homeostasis by astrocytes was
meability upon augmentation of [K+]o (Nichols also investigated in an in vitro study showing
and Lopatin, 1997). Astroglial Kir channels are that the knockdown of Kir4.1 protein by RNA
characterized by a high sensitivity to extracellu- interfering caused a decrease in Kir4.1 expres-
lar exposure to submillimolar barium (Ba2+) and sion that was paralleled by a diminution in glu-
caesium (Cs+) (Ransom and Sontheimer,1995). tamate uptake capability (Kucheryavykh et al.,
Numerous in vitro and in situ molecular and 2007). Kir4.1 deletion was also shown to alter the
functional evidence have shown that Kir4.1 under- [K+]o homeostasis in the respiratory network but
lies the majority of the astrocytic Kir conductance did not affect the network activity (Neusch etal.,
(Li et al., 2001; Olsen et al., 2006; Butt and Kalsi, 2006). Mutations causing loss of function of the
2006). The Kir4.1 drives bidirectional movement KCNJ10 gene result in a patient phenotype with
of K+ depending on the transmembrane K+ gradi- multisymptomatic syndrome, called SeSAME
ent because it is weakly rectifying. This property is syndrome, characterized by epilepsy, sensorineu-
essential for the K+ buffering and siphoning occur- ral deafness, ataxia, and electrolyte imbalance
ring in specific astroglial districts (Kofuji and (Bockenhauer etal., 2009; Scholl etal.,2009).
Newman, 2004; Butt and Kalsi, 2006). The contri- A major issue in the evaluation of the Kir4.1
bution of Kir to the astrocytic K+ conductance has role in astrocytes in vitro is that its molecular
been addressed in in vitro and in situ biophysical and functional expression is rare without phar-
and pharmacological analyses, showing that a cur- macological manipulation (Ferroni et al., 1995;
rent inhibited by relatively low concentrations of Guadagno and Moukhles, 2004; Benfenati etal.,
Ba2+ was expressed in cultured astrocytes and in 2006; Noell etal., 2007). The setup of experimen-
Mller cells (Barres etal., 1990; Ferroni etal., 1995; tal models in which astrocytes in vitro resemble
Ransom and Sontheimer, 1995; Olsen etal., 2006; more the functional and molecular phenotypes
Benfenati etal., 2006). Molecular analyses studies in vivo remains challenging, and it is currently
have shown that whereas Kir2.1 is expressed mainly the target of novel emerging tools and technolo-
in neurons, the Kir4.1 subtype is depicted almost gies (Benfenati etal., 2010; Benfenati etal., 2013;
exclusively in different subtypes of macroglial cells Benfenati etal.,2014).
(Butt and Kalsi, 2006; Olsen and Sontheimer, 2008), The process of spatial redistribution of K+ in
reinforcing the hypothesis of its primary role in the astroglial syncytium mediated by Kir chan-
homeostatic astroglial function. Notably, in mouse nels assumes that these channels are compart-
neocortex Kir4.1 was reported to form a heterote- mentalized in specific astrocytic membrane
tramer with Kir5.1 whereas homomeric Kir4.1 was regions facing liquid compartments where
Microdynamics of Water and Ion Homeostasis in the Brain 9

oscillations in [K+]o occur. Kir4.1 was shown studies have shown that Kir4.1 is developmen-
to be enriched at the endfeet of astrocytic pro- tally regulated as Kir4.1 transcript and protein
cesses enwrapping synapses and facing blood rise up during the first postnatal days, thereby
vessels and the pia mater (Higashi et al., 2001; leading to a large increase in astrocyte inward
Hibino et al., 2004; Nagelhus et al., 2004) or K+ current density underlying the passive cur-
in perivascular processes of Mller glial cells rent pattern of hippocampal astrocytes (Seifert
(Nagelhus et al., 1999; Kofuji et al., 2002). The etal.,2009).
pattern of Kir4.1 expression suggests that the It should be pointed out that, even though
microdynamics regulating [K+]o homeostasis Kir4.1 has a major role in [K+]o homeostasis,
could be mediated by plasma-membrane pro- other K+ channels are emerging as possible can-
teins arranged in a microdomain forming mul- didates contributing to this process. Of potential
timolecular functional complexes. The analysis relevance could be the members belonging to
of Kir4.1 indicates that its C-terminus harbors the two-pore domain K+ (K 2P) channels family
a PDZ-binding motif for the interaction with (Patel and Honor, 2001). The TREK and TWIK
proteins containing PDZ domains (Connors subfamilies have been recently characterized in
et al., 2004). Kir4.1 was found to colocalize astrocytes in vitro (Gnatenco etal., 2002; Ferroni
with syntrophins, a group of proteins belonging etal., 2003)and in situ (Seifert etal., 2009; Zhou
to the dystrophin-associated protein complex etal., 2009; Benesova etal., 2012). These channels
(DAPC), and to form a macromolecular assem- are open in a large range of membrane poten-
bly with dystrophin (Dp71), -dystroglycan, tials, providing a substantial contribution to the
-dystroglycan and -syntrophin both in high astrocyte passive K+ conductance in situ
Mller cells and astrocytes (Ishii et al., 1997; (Zhou et al., 2009). A strong molecular interac-
Guadagno and Moukhles, 2004; Hibino et al., tion mediating TWIK-1/TREK-1 heterodimers
2004; Nol et al., 2005; Connors and Kofuji, was shown to underlie astrocytic passive con-
2006). Alpha-syntrophin was shown to be cru- ductance and cannabinoid-induced glutamate
cial for DAPCKir4.1 interaction in brain cortex release from astrocytes (Hwang et al., 2014).
and retina, depicting a critical role of scaffold- Immunofluorescence studies showing that K 2P
ing proteins in [K+]o homeostasis (Connors etal., channels are particularly enriched at the astro-
2004; Connors and Kofuji, 2006). Mice deficient cytes endfeet support the view of their poten-
of the syntrophin gene display Kir4.1 dis- tial role in [K+]o homeostasis (Zhou etal., 2009;
tribution comparable to that of wild-type ani- Seifert et al., 2009). In this context, K 2P chan-
mals. However, the absence of the syntrophin nels were suggested to be involved in K+ uptake
causes a delay in [K+]o clearance elicited by in the hippocampus (Psler et al., 2007). It also
sustained neuronal activity due to mislocaliza- remains to be defined whether outward K+ cur-
tion of the water channel aquaporin-4 (AQP4) rents mediated by voltage-gated (Kv) and cal-
(Amiry-Moghaddam etal., 2003). Moreover, two cium (Ca 2+)-activated K+ channels that have been
other anchoring proteins, PSD95 and SAP97, described in astrocytes both in vitro and in situ
were reported also to colocalize with Kir4.1 in have a role in [K+]o homeostasis (Tse etal., 1992;
Mller cells, thereby providing indirect evidence Bordey and Sontheimer, 1999; Bekar etal., 2005;
of their involvement in channel compartmental- Filosa etal., 2006). Interestingly, it was reported
ization (Horio etal., 1997). The crucial role of the that Ca2+-activated K+ channels (rSlo) and
patterned distribution of Kir4.1 in [K+]o homeo- the voltage-gated K+ channel Kv1.5 are highly
stasis was also confirmed by works showing expressed at astroglial endfeet enwrapping
that its deletion caused an epileptic phenotype microvasculature which are also rich in AQP4
(Haj-Yasein etal., 2011). Notably, in the sclerotic (Roy etal., 1996; Price etal., 2002). Another issue
hippocampus of patients with mesial temporal that remains to be addressed in the context of
lobe epilepsy, a loss of perivascular Kir4.1 expres- [K+]o homeostasis mediated by the spatial buff-
sion was associated with loss of dystrophin and ering mechanism is the fact that the expression
-syntrophin but not with that of -dystroglycan of K+ channels is dynamically regulated, and
(Heuser et al., 2012). These data suggest that changes in K+ conductance have been reported
the perturbation of the dystrophin-associated in various experimental paradigms of pathologi-
protein complex, by altering Kir4.1 expression, cal conditions in situ and in vitro (MacFarlane
could modify [K+]o buffering and clearance, thus and Sontheimer, 1997; Anderov et al., 2004;
contributing to the epileptiform activity. Other Pivonkova etal.,2010).
10 Part I: Homeostatic Regulators

Role ofAquaporins in astrocytes prominently in the CA1 stratum


inAstroglial Potassium lacunosum-moleculare and the molecular layer
Homeostasis of the dentate gyrus (Hsu et al., 2011). AQP4
There is experimental evidence that in the ret- can assemble in supramolecular assemblies
ina, as well as in other regions of the CNS, the in the plasma membrane, called orthogonal
increase in [K+]o caused by neuronal activity pro- arrays of particles (OAPs). Immunogold stain-
duces a diminution in extracellular space owing ing by anti-AQP4 specific antibodies and analy-
to a transient increase in volume of glial endfeet sis by freeze-fracture electron microscopy have
mediated by the high rate of fluid accumulation allowed the earliest description of OAP visu-
accompanying K+ uptake (Holthoff and Witte, alization in brain astrocytes (Rash etal., 1998),
2000; stby etal., 2009). Notably, AQP4, which a result confirmed by live imaging performed
is the predominant water channel in astrocytes, by using super-resolution microscopy (Rossi
has been proposed as the molecular partner of etal., 2012). While the physiological role of such
Kir4.1 in spatial buffering of K+ by facilitating highly hierarchically organized supramolecular
the osmotic water movement through the plasma structure is still unclear, a crucial role for AQP4
membrane (Nagelhus et al., 1999; Nagelhus OAPs has been demonstrated in the pathophysi-
etal.,2004). ology of neuromyelitis optica (NMO). NMO
AQP4 is one of the members of the large patients are characterized by the presence of IgG
family of water channels called aquaporins, autoantibody (NMO-IgG), which binds to AQP4
which in mammals is composed of 13 members (Jarius et al., 2008). This antibody recognizes
(Papadopoulos and Verkman, 2013). The first AQP4 when it is aggregated into OAP square
member of the family, called AQP1, was identi- arrays (Nicchia etal., 2009). Aspecific nontoxic
fied by Agre and coworkers who discovered that inhibitor for AQP4 is still lacking, but experi-
the protein CHIP28 is a bona fide water chan- ments performed on different KO mice models
nel when expressed in Xenopus oocytes (Preston have provided clear evidence on the crucial role
et al., 1992). The functional subunit of AQP is of AQP4 in brain water microdynamics, astro-
a monomer composed of six transmembrane cyte migration, neurodevelopment, neuron exci-
domains in which two short helical segments tation, and synaptic plasticity (Papadopoulos
that surround cytoplasmic and extracellular ves- and Verkman,2013).
tibules, connected by a narrow aqueous pore, can AQP4 has been shown to be associated
be identified (Ho et al., 2009). The pore allows with DAPC by interacting with -syntrophin
the transport of only 1 ion for 109 water mol- (Amiry-Moghaddam et al., 2004), and in
ecules (Pohl, 2004). The functional channel is a vivo evidence using different pathophysi-
tetrameric assembly, implying that four conduit ological models supports the view that the
pathways are present in each AQP protein (Walz molecular interplay between AQP4-Kir4.1
etal., 1994; Cheng etal.,1997). and -syntrophin-mediated binding to DAPC
Expression pattern of AQPs in the mamma- is essential to K+ clearance in pathohysiologi-
lian brain is characterized by a dominant pres- cal conditions (Amiry-Moghaddham et al.,
ence of AQP4 and AQP1 (Amiry-Moghaddam 2003). Data regarding the interactions of
and Ottersen, 2003; Papadopoulos and Verkman, AQP4 with Kir4.1 in the physiological con-
2013). The expression of AQP9 has also been text are more controversial. Brain astrocytes
reported (Badaut et al., 2004), but its localiza- in which Kir4.1 was knocked down by siRNA
tion is still controversial (Amiry-Moghaddam did not display alteration in water perme-
et al., 2005). In rodent AQP1 is expressed in ability (Zhang and Verkman, 2008). Genetic
the epithelium of the choroid plexus facing the ablation of AQP4 did not alter Kir4.1 distribu-
cerebrospinal fluid, whereas AQP4 is primar- tion and functional properties in Mller cells
ily expressed in the astrocytic processes of the (Ruiz-Ederra etal., 2007), but caused a slowed
glia limitans facing the brain surface and in dynamics in [K+]o homeostasis in rodent brain
the endfeet of perivascular astrocytes (Badaut (Binder et al., 2006; Strohschein et al., 2011).
etal., 2002). AQP4 is also expressed in ependy- Electrophysiological studies in hippocampal
mal cells of the glia limitans and in perivascular slices from AQP4 KO mice evidenced a marked
processes of Mller cells (Hamann etal., 1998). alteration of stimulus-evoked K+ uptake due to
In the mouse hippocampus, AQP4 was shown the impairment of the astroglial Na+/K+-ATPase
to be developmentally regulated and expressed (Strohschein etal., 2011), which was also shown
Microdynamics of Water and Ion Homeostasis in the Brain 11

to interact with AQP4 (Illarionova etal., 2010). underpinning [K+]o homeostasis (Scemes and
Dysregulated water and [K+]o homeostasis have Spray, 2012). High [K+]o was shown to increase
been linked to epilepsy (Binder and Steinhuser, coupling between cultured spinal cord astro-
2006; Binder et al., 2012). A pronounced cytes (Enkvist etal., 1994). The major evidence
decrease in AQP4 expression was observed of gap junction implication in [K+]o homeo-
in the early phases of epilepsy in a model of stasis has been provided recently by use of
kainate-induced seizures (Lee etal., 2012)and K+-selective microelectrodes in double-KO mice
immunogold analysis showed that in human for astroglial Cx30 and Cx43 (Wallraff et al.,
epileptic tissue the impaired buffering of [K+]o 2006). It was shown that in the hippocampus,
was associated with a loss of AQP4 in perivas- gap junctional communication contributed to
cular astrocyte endfeet (Eid etal., 2005). Based the radial dissipation of [K+]o in the stratum
on these results, it could be envisaged that the lacunosum-moleculare but not in hippocampal
characterization of microdomains serving the astrocytes located in the stratum radiatum. The
process of water homeostasis would be useful mechanism underpinning the role of Cx and gap
to unravel the pathophysiology of unbalanced junction communication in [K+]o homeostasis is
[K+]o microdynamics. not clearly understood. It has been shown that
changes in phosphorylation state of Cx43 are
Role ofConnexins and likely to be involved in cell-coupling increase
Pannexins inAstroglial following a challenge with high [K+]o (De
Potassium Homeostasis Pina-Benabou et al., 2001; Scemes and Spray,
A prerequisite for the homeostatic function of 2012). Several pieces of in vivo evidence have
astroglia is the abundant interconnection and indicated that astroglial Cx expression could
coupling to work as a functional syncytium. be modified in many diseases and pathological
Gap junction protein subunits, called connex- conditions characterized by alteration in [K+]o
ins (Cxs), are the membrane channels distrib- homeostasis (Eugenin et al., 2012; Steinhauser
uted along the astrocytic processes that provide etal., 2012). However, mainly because of differ-
unique direct conduit between astrocytes. Gap ences in animal models, types of experimen-
junctions play a crucial role in enabling the tal paradigm used, and parameters analyzed,
synchronization of cellular processes in astro- inconsistencies and contradictions about Cx
cytes, such as intercellular Ca 2+ waves propa- role in neuronal excitability have been reported.
gation and metabolite trafficking and delivery Some studies have explored the effect of phar-
to neurons (Giaume et al., 1997) but also for macological disruption of gap junction commu-
signaling at the gliovascular interface (Simard nication on seizure activity (Medina-Ceja et al.,
et al., 2003). They mediate the passage of ions 2008; Voss et al., 2009). Notably, most of these
(K+), amino acids, glucose, glutathione, ATP, researches investigated the effect of Cx modula-
and small signaling molecules (Theis et al., tors such as carbenoxolone, which, however, is
2005). The main gap junction protein isoforms now known to affect also other plasma mem-
expressed by astrocytes are Cx30 and Cx43 brane ion channels at least in cultured astro-
(Nagy and Rash, 2000; Theis etal., 2005). Cx43/ cytes (Ye et al., 2009; Benfenati etal.,2009).
Cx30 double-KO mice have only minimal gap Concerning the mechanisms of [K+]o homeo-
junction communication between astrocytes stasis, it was reported that AQP4 knockdown
(Wallraff et al., 2006; Rouach et al., 2008), by siRNA induced the downregulation of Cx43
suggesting that both proteins are the main protein expression and junctional coupling in
components of functional astroglial gap junc- primary cortical astrocytes, suggesting that a
tion channels. There is evidence that astrocyte functional interaction exists between AQP4 and
Cxs could also form hemichannels that open Cx43 in astrocytes in vitro (Nicchia etal., 2005).
between the cell interior and the extracellular However, recent evidence has revealed improved
space both in physiological and pathological K+ spatial buffering occurring in AQP4-deficient
conditions, allowing exchange of small mol- mice that might result from the increase in gap
ecules between the cytoplasm and the extracel- junction coupling (Strohschein et al., 2011).
lular environment (Bennet etal.,2003). Further studies will elucidate whether a molecu-
Several findings indicate that gap junc- lar or functional interplay occurs between AQP4
tions play a major role in the coordination and Cxs to serve the microdynamic regulation of
and synchronization of astroglial signaling [K+]o homeostasis.
12 Part I: Homeostatic Regulators

Pannexin 1 (panx1) is a protein that func- The cytotoxic swelling in isotonic conditions
tions as a large-pore membrane channel per- causes the movement of ions and water through
meable to relatively large molecules including brain capillaries, which leads to the formation of
ATP (MacVicar and Thompson, 2010). In the ionic edema. If these dynamic changes develop
CNS it is expressed in neurons as well as in with the rupture of the BBB, fluid, proteins
astrocytes (Thompson and MacVicar, 2008). and cells enter the brain parenchyma, promot-
In astrocytes-neuron cocultures it was shown ing the accumulation of fluid and the develop-
that high [K+]o activates panx1 channels by ment of vasogenic edema (Simard etal., 2007a).
voltage-independent mechanisms (Scemes and Vasogenic edema occurs at late stages of various
Spray, 2012). In a mouse model of epilepsy, panx1 neurological insults in which cytotoxic edema
blockade was reported to reduce seizure activity, are observed, such as ischemic insults, trau-
suggesting that it may contribute to its develop- matic injuries, and brain tumors (Marmarou,
ment (Santiago et al., 2011). However, the pre- 2007) but can also develop as a result of men-
cise role of panx1 in [K+]o homeostasis remains ingitis (Nau and Brck, 2002). Transient vari-
largely unknown. ations in ESV also occur under physiological
conditions in response to high neuronal activ-
MICRODY NAMICS ity (MacVicar and Hochman, 1991; Holthoff
OFCELL VOLUME and Witte, 1996). Measurements of changes in
H O M E O S TA S I S cellular volume obtained from optical and dif-
Brain homeostasis is also characterized by the fusion properties in the brain suggest that they
ability of brain cells to maintain their volume were caused by alterations of astrocytic vol-
in response to physiological stimuli. This is ume (Holthoff and Witte, 1996). Even though
possible because they can constantly counter- it was reported that in situ transient swelling of
act the increase in cell volume induced by the astrocytes promoted the outflow of glutamate,
formation of transient and spatially-defined which modulated neuronal excitability (Takano
osmotic gradients with a process called regula- etal., 2005), the functional consequences of this
tory volume decrease (RVD). Since the osmotic physiological volume increase remain to be fully
gradients are generated by the release and elucidated.
uptake of osmolites at the neuron-astrocyte and
astrocyte-endothelium interfaces, it results that Plasma Membrane Channels
astrocytes are central players in brain volume Involved inCell Volume
homeostasis. It is also well known that under Regulation ofAstroglia
several acute and chronic pathological condi- In the past decade, significant efforts have been
tions larger and more sustained cell volume made to uncover the molecular underpinnings
increments are observed, which lead to dynamic that contribute to cell volume regulation in
changes in the extracellular space volume (ESV). physiological conditions and whose dysfunction
The astrocyte volume increase promoted by the underlies the development of cytotoxic edema. In
redistribution of water and osmolytes between this context, plasma-membrane transporters, ion
intra- and extracellular compartments is called channels, and water proteins have been reported
cytotoxic edema (Liang etal., 2007), Cytotoxic to be key effectors of physiological and patho-
swelling affects ESV and geometry, by creating physiological stimuli that promote astroglial
diffusion barriers for signaling molecules that swelling (Pasantes-Morales and Vzquez-Jurez,
contribute to neuron-glia communication and 2012). The role of astrocytic transporters in cell
hence can alter the excitability of the neuronal volume regulation is well known (Kahle et al.,
tissue (Sykov and Nicholson, 2008). It devel- 2009), and therefore we review here only the pos-
ops as consequence of hyponatremia, ischemia, sible relevance of ion and water channels, some
and hepatic encephalopathy but also as result of of which have been characterized just recently
head trauma and epilepsy (Liang et al., 2007). (Figure 1.3). The functional modulation of these
Accumulation of [K+]o, glutamate and ammo- proteins, as well as alterations in their expression
nia, as well as an augmentation of products of and distribution in astrocyte microdomains,
oxidative stress, have been shown to regulate have been linked to the development of cyto-
the cellular processes controlling the cell vol- toxic edema and therefore could become inter-
ume homeostasis and to have a pathogenetic esting therapeutic candidates to counteract the
role in the development of cytotoxic edema cell swelling that occurs in various neurological
(Kimelberg, 2005; Hussinger and Grg, 2010). disorders.
Microdynamics of Water and Ion Homeostasis in the Brain 13

H2O (AQP4) 5 Ca2+(TRPV4) (?)


Glu Na+ 1
3
H2O Na+ (?)

2
3 [Ca2+]
H2O 1 [osm]
4

1 K+ Cl, Tau, EAA


5 (VRAC)

H2O 4
K+

1) Rise in intracellular osmolyte concentration


2) Water influx and osmotic swelling
3) Volume sensing and osmotransduction
4) Inorganic and organic osmolyte efflux
5) Osmotic water efflux and volume recovery

FIGURE 1.3 Astroglial swelling and possible routes for cell volume recovery. Under physiological
conditions astrocyte volume remains stable owing to the osmotic equilibrium between intra- and extracellu-
lar compartments. However, cell volume homeostasis can be locally compromised by the transient increment of
intracellular osmolarity. As consequence of the neuronal activity K+, Na+, and Glu- uptake occurs at the endfeet
of astroglial cells (1). This increase in intracellular osmolytes is accompanied by osmotically driven water influx
through diffusion or by means of AQP4 (2). The mechanism underlying astroglial volume sensing, which initi-
ates the series of events leading to the process of cell volume recovery, called regulatory volume decrease (RVD),
remains to be determined unequivocally. Swelling-induced, intracellular Ca 2+ elevation ([Ca2+]i) might play a criti-
cal role at least in vitro (3). In addition to Ca 2+ release from intracellular stores, members of the transient receptor
potential (TRP) channel superfamily are plausible molecular candidates for osmotransduction. The osmosensitive
Ca 2+ channel TRPV4 is expressed in astrocytes in vitro and in situ and may be involved. The ensuing process of
RVD is based on the extrusion of intracellular solutes paralleled by obliged water efflux. The volume-regulated
anion channel (VRAC), by permeating inorganic (Cl) and organic osmolytes such as taurine and excitatory
amino acids could be an effector of RVD in vitro and perhaps also in vivo. Acontribution of K+ efflux is also likely,
but which are the specific volume-sensitive K+ channels involved is still unclear (4). The osmolyte outflow creates
the gradient for water efflux through diffusion and AQP4, and promotes RVD to recover the initial volume (5).
The diagram highlights the role of ion channels and does not consider the relevant role of various transporters.
Modified from Benfenati and Ferroni [2010]

Water Channels produced an augmentation of the rate of cyto-


It is now widely accepted that AQPs are involved toxic swelling, which was associated with an
in pathological brain swelling and have been increase in intracranial pressure (Yang et al.,
postulated to be relevant pharmacological tar- 2008). Results confirming the role of AQP4 in
gets (Pasantes-Morales and Cruz-Rangel, 2010; cytotoxic edema were obtained in dystrophin-
Badaut et al., 2011; Verkman, 2012). The role and -syntrophin KO mice, in which the dis-
of AQP4 in cytotoxic edema has been extrapo- ruption of the macromolecular complex with
lated by studies in AQP4-KO mice subjected to AQP4 caused a mislocalization of astroglial
water intoxication and ischemic stroke (Manley AQP4 and delayed the development of brain
et al., 2000). In these mice, toxic hypotonic edema (Vajda etal., 2002; Amiry-Moghaddam
challenge caused a reduced swelling of perivas- et al., 2004). The mislocalization of AQP4 at
cular endfeet and displayed a less severe neu- astrocytic endfeet has been proposed to be of
rological phenotype and minor mortality. By possible therapeutic value to counteract the
contrast, in mice in which AQP4 was overex- early formation of cytotoxic edema in ischemic
pressed in astrocytes, acute water intoxication mice (Steiner etal.,2012).
14 Part I: Homeostatic Regulators

Variations in functional assembly of AQP4 in Potassium Channels


target astrocytic regions could also be involved As already stated, a close correlation exists
in cell volume regulation by affecting the efficacy between astrocytic volume regulation and the
of the process of [K+]o homeostasis. High [K+]o ability of astrocyte to remove extracellularly
was reported to induce astrocytic swelling in situ accumulated K+, thereby suggesting the presence
(Anderov etal., 2001), and, in the human brain, of closely coupled mechanisms. As described
high [K+]o correlates with an increase in intrac- in the previous section, Kir4.1 channels are the
ranial pressure in patients with severe traumatic predominant pathway of K+ uptake in astroglial
brain injury (Reinert et al., 2000). AQP4 is also cells. The co-expression of Kir channels and
upregulated in pathological conditions associ- AQP4 in astrocyte endfeet suggests that K+ and
ated with the development of cytotoxic edema water could be cotransported through the syn-
(Vizuete et al., 1999; Taniguchi et al., 2000; cytium and contribute to the activity-dependent
Saadoun etal., 2002; Aoki etal., 2003; Aoki etal., volume changes of the extracellular space
2005), and reduction of AQP4 expression was (Nagelhus et al., 2004). Upregulation of both
shown to be protective (Zeng etal., 2010; Fukuda proteins has been observed in human brain
etal., 2013; Rama Rao etal.,2014). astrocytes following several pathological states
It must be pointed out that the role of AQP4 (Saadoun etal., 2003). The K+ flux through Kir4.1
in the regulation of brain volume homeostasis was reported to regulate swelling of astroglial
seems completely different in vasogenic edema. processes in experimental spinal cord edema
Since AQP4 permits the bidirectional movement and in other regions of the CNS challenged
of water, in vasogenic edema it could facilitate with an hypotonic stimulus (Dibaj et al., 2007;
the removal of water from brain parenchyma. Hirrlinger etal.,2008).
To support this view it was shown that in Recent studies in situ have shown that in slices
AQP4-deficent mice, infusion of an isotonic subjected to oxygen glucose deprivation, astro-
solution augmented the intracranial pressure cytic swelling is modulated by putative inhibi-
owing to fluid accumulation (Papadopoulos tors of K2P channels, suggesting that they might
etal., 2004). The same result, accompanied by a play a pivotal role in cell volume responses dur-
worsening of neurological scores, was obtained ing ischemia (Benesova etal., 2012). Some mem-
upon genetic ablation of AQP4 in a model of bers of the TREK subfamily of K 2P contribute to
tumor-induced vasogenic edema (Papadopoulos the K+ conductance in hippocampal astrocytes in
et al., 2004), upon induction of brain abscesses situ and are particularly enriched at the process
(Bloch etal., 2005), and as a consequence of sub- endfeet (Zhou etal., 2009; Seifert etal., 2009). In
arachnoid hemorrhage (Tait et al., 2010). In the cultured astrocytes, two members, TREK-1 and
context of brain volume homeostasis, the abil- TREK-2, have been identified and functionally
ity of AQP4 to remove the excess of water from characterized (Gnatenco etal., 2002; Ferroni etal.,
brain compartments could also influence the 2003). Simulated ischemia in vitro was shown
development of hydrocephalus. Hydrocephalus to promote an increment in expression of both
is the result of the imbalance between produc- channels (Kucheryavykh etal., 2009; Wang etal.,
tion and absorption of cerebrospinal fluid that 2012; Wu etal., 2013). However, only studies per-
leads to fluid accumulation in the ventricular sys- formed in KO animals will provide a clear answer
tem and an increase in intracrananial pressure. about their putative role in cell volume regulation.
Obstructive hydrocephalus, which can be caused In epithelial cells, Ca2+-activated K+ channels of
by trauma, tumor, hemorrhage, and infection, is intermediate/small conductance and large con-
determined by a diminution of the cerebrospinal ductance have been associated with cell volume
fluid draining into the subarachnoid space. In an regulation (Barfod et al., 2007). In astrocytes in
experimental mice model of progressive obstruc- situ, large-conductance Ca2+-activated K+ chan-
tive hydrocephalus, the deletion of AQP4 pro- nels of the rSLo subtype have been reported to be
duced a strong increase in intracranial pressure regionally distributed in AQP4-expressing astro-
and an enlargement of lateral ventricles (Bloch cyte endfeet that abut blood vessels (Price etal.,
etal., 2006). This finding again supports the tenet 2002). Although the activity of these K+ channels
that the modulation of AQP4 expression and/or has been linked to neuron-mediated regulation of
function could be exploited for favoring water vascular tone (Filosa etal., 2006; Girouard etal.,
removal from brain compartments in conditions 2010), a role in cell volume regulation, especially
of fluid accumulation. at the endfeet, cannot be ruledout.
Microdynamics of Water and Ion Homeostasis in the Brain 15

Chloride Channels inhibition but to the diminution of excitotoxic


In virtually all mammalian cells volume control amino acids release through VRAC (Abdullaev
is achieved through tightly regulated activation et al., 2006). Another Cl conductance, the
of chloride (Cl) and cation channels and the con- hyperpolarization-activated, inward rectifier Cl
tribution of transport mechanisms (Hoffmann current (IClh), has been described both in vitro
et al., 2009). Whereas in cell swelling and the and in situ (Ferroni et al., 1995, Ferroni et al.,
ensuing RVD the role of different types of cat- 1997, Makara etal., 2003). There is evidence that
ion channels depends on their relative expres- IClh is mediated by ClC-2 (Makara et al., 2003),
sion levels and on their gating process, Cl fluxes which is a member of the growing family of the
through a specific volume-sensitive ion channel Cl channels and transporters called ClC (Jentsch
can contribute to both cell swelling and volume etal., 2002). ClC-2 is ubiquitously expressed and
recovery owing to the dynamics of the electro- abundantly present in the brain (Thiemann etal.,
chemical driving force. In neurons, prolonged 1992). In astrocytes in situ ClC-2 was found in the
glutamate challenge was reported to generate CA1 and CA3 regions of the hippocampus and
the formation of varicosities with the contri- in brain slices (Sk et al., 2000; Benesova et al.,
bution of Cl influx through volume-sensitive 2012). A detailed analysis showed that ClC-2
anion channels. The same channels were also is expressed at the endfeet of astrocytes facing
involved in its extrusion, leading to volume blood vessels (Blanz etal., 2007). The fact that IClh
compensation upon glutamate washout (Inoue favours the outflow of Cl at membrane poten-
and Okada, 2007). Astrocytes in vitro display a tials negative to the equilibrium potential for Cl
very low background Cl permeability (Walz, makes this conductance an actractive candidate
2002), and previous studies have shown that as one of the RVD effectors. Notably, recombi-
Cl actively accumulates in astrocytes reaching nant ClC-2 activity was shown to be upregulated
an equilibrium potential more positive than the by hypotonic challenge (Grnder etal., 1992)and
resting membrane potential (Kettenmann etal., in various cell types it was reported to contrib-
1987; MacVicar et al., 1989). Cultured astro- ute to cell volume regulation (Furukawa et al.,
cytes respond to a hypotonic challenge with an 1998, Xiong et al., 1999). Deletion of ClC-2
augmentation of Cl conductance promoted by causes white matter spongiform vacuolation in
the activation of volume-regulated anion chan- mice brain and spinal cord, similar to that seen
nels (VRACs) (Parkerson and Sontheimer 2004; in humans with leukoencephalopathies (Blanz
Benfenati et al., 2007a; Benfenati et al., 2009). et al., 2007). Recent work has evidenced that in
Asimilar current was also reported to be stimu- patients with leukoencephalopathies character-
lated by changes in astrocyte morphology and ized by white matter edema, ClC-2 is mutated
biochemical alteration of the actin cytoskeleton (Depienne et al., 2013). The IClh is highly sensi-
(Lascola et al., 1998). The molecular identity of tive to mild extra- and intracellular acidification
VRAC, or part of it, has recently been unveiled (Ferroni etal., 2000), which are conditions asso-
(Qiu etal., 2014; Voss etal., 2014)but it remains ciated with the development of brain ischemia.
to be determined whether it also underlies In cultured astrocytes there is no correlation
astrocyticVRAC. between ClC-2 expression and functional activ-
VRAC stimulation in cultured astrocytes ity (Parkerson and Sontheimer, 2004), and IClh is
has been associated with RVD (Parkerson and stimulated only upon astrocyte morphological
Sontheimer, 2003). Interestingly, a downregula- differentiation with a long-term treatment with
tion of AQP4 in cultured cortical astrocytes led cAMP analogs (Ferroni etal., 1995). Even though
to a strong diminution in VRAC activity, adding other isoforms of the ClC family have been found
further support to its role in cell volume regula- in astrocytes, their relevance to cell volume regu-
tion (Benfenati et al., 2007a). Pharmacological lation is still questionable. Immunostaining anal-
analysis showed that a putative inhibitor of VRAC, ysis has shown that in cultured astrocytes other
tamoxifen, was neuroprotective in a rat model of members of the ClC family, namely ClC-3, ClC-5,
focal ischemia (Kimelberg etal., 2003). The same and ClC-7, were also expressed at the cell surface
effect was reported with another specific VRAC but were not functionally identified (Parkerson
blocker, DCPIB, in a model of ischemia induced and Sontheimer, 2004). Moreover, astrocytic iso-
by middle cerebral artery occlusion (Zhang etal., forms of the skeletal muscle ClC-1 channels have
2008). However, neuroprotection was not attrib- also been described but not yet characterized at
uted to cell volume modulation through VRAC a functional level (Zhang et al., 2004). Finally,
16 Part I: Homeostatic Regulators

bestrophin-1, a gene-encoding calcium-activated necessary to keep Na+/K+-ATPase operative


Cl channel, was identified in astrocytes in vitro and therefore could influence [K+]o homeosta-
and in situ (Park et al., 2009). Interestingly, the sis (Sontheimer etal., 1994), but their relevance
fruit fly (Drosophila melanogaster) ortholog of in cell volume regulation has not been demon-
bestropin-1 was reported to be independently strated. It must be emphasized that Na+ could
activated by intracellular Ca2+ elevation and also permeate into astrocytes through nonse-
extracellular hypotonicity, therefore suggesting lective cation channels. One channel that has
that it might play a role in cell volume homeo- recently attracted great interest is the NCCa-ATP,
stasis (Chien and Hartzell, 2007). However, its which was identified in freshly dissociated adult
volume sensitivity in mammalian cells is still astrocytes (Chen and Simard, 2001). This chan-
controversial (Fischmeister and Hartzell, 2005), nel is permeable to inorganic monovalent cati-
and also the involvement of bestrophin-1 in the ons but not to Ca 2+ and Mg 2+. Importantly, its
regulation of astrocytic volume remains to be opening is dependent on the presence of intra-
addressed. cellular nanomolar free Ca 2+ and the absence
of ATP, therefore suggesting that it could be
Cation Channels operative in conditions of metabolic failure.
It is now widely accepted that cationic channels The NCCa-ATP channel is made of a pore-forming
play an important role in volume regulation in subunit and a regulatory subunit, the sulfonylu-
mammalian cells (Hoffmann et al., 2009). It is rea receptor 1 (SUR1), which is also the regula-
also well known that cation entry in astrocytes, tory subunit of the K ATP channels in pancreatic
by promoting osmotically driven water influx, beta cells (Chen etal., 2003). The pore subunit is
are critically involved in triggering cytotoxic formed by the transient receptor potential mel-
swelling (Liang et al., 2007). Na+ and Ca 2+ have astin 4 (Woo etal., 2013). The NCCa-ATP activity
favorable electrochemical gradients for moving is downregulated by sulfonylurea compounds
into astrocytes when cation channels and tras- such as tolbutamide and glibenclamide which
porters are activated. Whereas the massive entry inhibit the SUR1-regulated K+ channel (K ATP).
of Na+ into the cells creates the osmotic gradient Interestingly, this channel is not constitutively
that drives water influx, the cytosolic rise of the expressed, but it becomes apparent following
second messenger Ca 2+ could influence cell vol- hypoxia or injury (Chen and Simard, 2001),
ume through more complex biochemical path- causing astrocyte depolarization and the devel-
ways (OConnor and Kimelberg, 1993; Akita and opment of cytotoxic edema. Its blockage in vivo
Okada,2011). by glibenclamide promotes a strong reduction in
The presence of different subtypes of the formation of cerebral edema (Simard etal.,
voltage-gated Na+ channels has been reported 2006). Other ion channels permeable to cations
in cultured cortical and spinal cord astrocytes that are likely to play a role in cell volume home-
(Sontheimer et al., 1991) as well as in slices of ostasis include those belonging to the transient
different CNS regions (Bordey and Sontheimer receptor potential (TRP) superfamily (Montell,
2000; Chvtal et al., 1995). The expression of 2001). The first TRP channel was described in
voltage-gated Na+ channels is heterogeneous in the photoreceptor of Drosophila melanogaster
terms of sensitivity to the specific blocker tetro- (Montell and Rubin, 1989). More than 20 TRP
dotoxin and biophysical properties (Sontheimer genes have been identified in mammals (Nilius
et al., 1991; Thio and Sontheimer, 1993), and and Owsianik, 2011). From protein homol-
it is also developmentally regulated (Kressin ogy, members of the TRP channel family are
et al., 1995; Bordey and Sontheimer, 1997). grouped into seven subfamilies (Wu etal., 2010).
Differences in expression level have also been Few members are selectively permeable to Ca 2+,
observed in gliotic astrocytes in situ and in vitro whereas most are non-selective cation channels.
(Jabs et al., 1997; MacFarlane and Sontheimer, Their activation mechanisms are very different,
1998). The estimated densities and the biophysi- and, interestingly, some of them are stimulated
cal properties of the channel subtype expressed by conditions which are relevant to ischemia
provides some clues to their functions that and lead to the development of brain edema
are not related to the genesis of action poten- such as pH acidification, osmotic changes, and
tials (Thio and Sontheimer, 1993). It has been membrane stretching (Simard et al., 2007b).
reported that voltage-gated Na+ channels cre- The expression and functional characterization
ate the route for the small, sustained Na+ influx of the astrocytic TRP subtypes involved in cell
Microdynamics of Water and Ion Homeostasis in the Brain 17

volume homeostasis is still incomplete. Cultured in cultured astrocytes in which it was shown that
astrocytes express mRNAs and proteins for stimulation with the neurotransmitter glutamate
various subtypes of TRPC subfamily (C stands or application of a mechanical stimulus caused
for canonical) (Pizzo etal., 2001; Beskina etal., transient elevations in [Ca 2+]i that propagated as
2007) which appear to be developmentally [Ca2+]i waves through the astrocyte monolayer
regulated (Malarkey et al., 2008). TRPCs have (Cornell-Bell et al., 1990; Charles et al., 1991).
variable cation permeability ratios and have Importantly, [Ca 2+]i waves were reported also
been implicated in mediating store-operated in vivo, a result that corroborates the view that
Ca 2+ entry in astroglial cells (Malarkey et al., they are physiologically relevant (Kuga et al.,
2008; Akita and Okada, 2011). Of note, TRPC1, 2011). In addition to Ca2+ permeation through
which is permeable to both Ca 2+ and Na+, was ion channels, [Ca2+]i rise in vitro and in situ can
shown to be activated by mechanical stress be triggered by stimulation of various receptors
(Hua et al., 2004; Malarkey et al., 2008; Reyes that allow Ca2+ entry or its release from intra-
et al., 2013), a stimulus that triggers RVD in cellular stores (Verkhratsky et al., 2012). There
astrocytes (Hua et al., 2010). A role of TRPC1 are indications that [Ca2+]i signaling modu-
in cell volume regulation, therefore, cannot be lates neuronal excitability (Araque et al. 2001;
ruled out but deserves further investigation. Volterra and Steinhuser, 2004), even though
Additional TRP candidates that could play a this view has recently been questioned (Agulhon
role in cell volume regulation are those of the etal., 2010). Astrocytic [Ca 2+]i dynamics are also
TRPV subfamily (V stands for vanilloid). This involved in the regulation of cerebral vascular
subfamily is composed of 6 members which tone (Filosa and Idding, 2013). It has been dem-
are activated by various chemical and physi- onstrated that the [Ca2+]i elevation can promote
cal stimuli and display large Ca 2+ over mono- the astroglial release of signaling molecules (gli-
valent cations permeability ratios (Owsianik otransmitters) through vesicular mechanisms
etal., 2006). TRPV4 was shown to be involved in in vitro and in situ (Parpura and Zorec, 2010),
osmoregulation and hence could function as an but its physiological relevance is still unclear
osmosensor (Mizuno etal., 2003; Liedtke etal., (Hamilton and Attwell, 2010). In cultured astro-
2003). Cortical and hippocampal rat astrocytes cytes it was reported that hypotonic challenge
in vitro and in situ express functional TRPV4 caused an increase in [Ca 2+]i, which was neces-
(Benfenati et al., 2007b; Bai and Lipski, 2010; sary for RVD (OConnor and Kimelberg, 1993),
Butenko etal., 2012). High-resolution immuno- and therefore a role of [Ca 2+]i dynamics in cell
gold analysis showed that TRPV4 expression in volume homeostasis in vivo cannot be ruled out.
situ was largely confined to astrocytic endfeet As second messenger molecule, [Ca2+]i signaling
enriched in AQP4 and abutting blood capillaries could modulate astrocytic functions through
and the pial surface (Benfenati etal., 2007b). It different mechanisms (Verkhratsky etal., 1988).
is noteworthy that in cortical astrocytes TRPV4 However, which are those involved in the modu-
was reported to interact physically with AQP4 lation of ion channels that regulate cell volume
and the macromolecular complex was demon- in vitro is still unclear. Previous work has shown
strated to be necessary for the regulation of cell that in cultured astrocytes extracellular ATP
volume because manipulation of the expression released through multidrug resistance protein
of TRPV4 and AQP4 led to a dramatic altera- could modulate volume-sensitive Cl current in
tion in RVD following hypotonicity challenge an autocrine fashion via activation of purinergic
(Benfenati etal.,2011). metabotropic receptors coupled with intracellular
Ca 2+ mobilization (Darby etal., 2003). The hypo-
ROLE OFINTR ACELLULAR tonically activated Cl current was also abolished
CALCIUM AND SODIUM by broad-spectrum inhibitors of calcium/calmo-
DY NAMICS INASTROCY TE dulin protein kinase (Li etal., 2002; Olson etal.,
R E G U L AT I O N O F B R A I N 2004). Unfortunately, the lack of unequivocal
H O M E O S TA S I S evidence about the presence of Cl channels acti-
It is now widely accepted that in astroglial cells vated by hypotonicity in situ has hampered the
the intracellular Ca2+ concentration ([Ca 2+]i) can possibility of defining the relevance of this in vitro
oscillate with temporal and spatial dynamics that findings. In this context, the bestrophin channel
are critical for brain homeostasis. The first evi- could deserve closer inspection. The astrocytic
dence of [Ca2+]i dynamics came from experiments bestrophin-1 channel is, in fact, activated by a rise
18 Part I: Homeostatic Regulators

in [Ca2+]i mediated by activation of Gq-coupled 2008). The relevance of [Na+]i transients in the
receptors (Park et al., 2009) and is expressed in context of astrocyte regulation of brain homeo-
CA1 hippocampal astrocytes in situ. However, it stasis in physiological conditions is linked, to
remains to be investigated whether it is also pre- a large extent, to the capacity of accumulated
sent in astrocytes of other brain regions and what Na+ to be exported through the Na+/K+-ATPase,
is its localization in specific cell microdomains. hence contributing to the uptake of extracel-
In cultured astrocytes two types of Ca2+-activated lular K+ (Sontheimer et al., 1994; DAmbrosio
K+ channels have been reported to be stimulated etal., 2002). Less clear are the functional conse-
by metabotropic glutamate receptors coupled to quences of pathological [Na+]i increases such as
Ca2+ mobilization (Gebremedhin etal., 2003)and those occurring during energetic failure (Silver
in situ the presence in the endfeet of intermedi- etal., 1997). An increase in [Na+]i mediated by
ate and large-conductance Ca 2+-activated K+ the NKCC1 transporter was shown to contrib-
channels has been described (Filosa etal., 2006; ute to the development of cerebral edema fol-
Longden etal., 2011). Their relevance in cell vol- lowing ischemia (Chen and Sun, 2005; Kahle
ume regulation in these spatially restricted cell et al., 2009). Clearly, since the unravelment of
regions is still unknown and warrants further [Na+]i signaling in astrocytes is relatively recent,
investigations. more work is warranted to uncover the molecu-
Another ion that has recently been associ- lar mechanisms through which [Na+]i dynamics
ated with intracellular signaling is Na+. It has can regulate astrocyte homeostatic processes,
been reported that in vitro and in situ tran- especially in pathological settings. In this con-
sient changes in cytosolic concentration of Na+ text, it was recently reported that in cultured
([Na+]i) occur as a result of synaptic stimulation astrocytes the activity of VRAC was negatively
(Kirischuk etal., 2007; Langer and Rose, 2009), regulated by pathological elevation of [Na+]i
and [Na+]i signaling can propagate as Na+ waves (Minieri etal., 2015).
through the astroglial syncytium (Rose and
Ransom, 1997; Langer and Rose, 2009). Chemical
and mechanical stimulations can also promote CONCLUDING REMARKS
astrocytic [Na+]i transients in situ and in vitro There is clear evidence that alterations of the
(Rose and Karus, 2013). These [Na+]i dynamics mechanisms regulating ion and volume homeo-
are mediated by Na+ influx through ion channels stasis are involved in the cytotoxic edema of the
or Na+-coupled secondary transporters for gluta- astrocytic syncytium that develops upon vari-
mate and gamma-aminobutyric acid (Rose and ous brain injuries (Liang etal., 2007). Astrocyte
Karus, 2013). It is also worth noting that the Na+ swelling occurs as result of a variety of biochemi-
and Ca2+ dynamics are tightly linked because of cal and molecular changes that affect the activity
the presence of plasmalemmal Na+/Ca2+ exchang- of the ion channels and transporters which regu-
ers and ion channels that are permeable to both late the movements through the plasma mem-
Na+ and Ca2+, such as TRP channels and several brane of organic and inorganic osmolytes as well
ionotropic receptors. as water (Pasantes-Morales and Vzquez-Jurez,
The amplitude and spatial dynamics of 2012). Since cytotoxic edema has a causal role in
[Na+]i transients in astrocytes are governed the secondary injury of neuronal tissue, the pre-
by the cellular distribution of the diverse Na+ vention of astrocyte swelling should be a primary
influx and efflux pathways. For example, the goal that novel pharmacological approaches shall
Na+-dependent glutamate transporters are more address for the development of effective therapeu-
expressed at the endfeet facing the synapses tic strategies (Kimelberg and Nedergaard, 2010).
than those abutting blood vessels (Chaudhry However, the pharmacological tools available so
et al., 1995; Lehre et al., 1995). An important far are still relatively poor in terms of specificity
factor that influences [Na+]i dynamics is also and knowledge of the underlying action mecha-
the fact that accumulated Na+ does not appear nisms. Previous work has shown that tamoxifen,
to be readily buffered, and there are no known a known inhibitor of VRAC channels (Voets
specific intracellular binding proteins. This cer- et al., 1995), was neuroprotective in a model of
tainly helps explain the general observation that focal ischemia (Kimelberg etal., 2000; Kimelberg
[Na+]i dynamics develop with a much slower etal., 2003), but tamoxifen has a large spectrum
time course as compared to [Ca 2+]i oscillations of known targets, including other ion channels
(Paemeleire and Leybaert, 2000; Langer et al., (Rivera-Guevara and Camacho, 2011). The puta-
2012)and can last tens of seconds (Bennay etal., tive, more specific inhibitor of VRAC channels,
Microdynamics of Water and Ion Homeostasis in the Brain 19

DCPIB, was shown to reduce infarct size in the of astroglial cells and new avenues to manipulat-
ischemic cortical penumbra in a model of mid- ing their physiology.
dle cerebral artery occlusion (Zhang etal., 2008),
but recently the same molecule was shown to References
modify the activity of other targets that are likely Abbott, N.J., Rnnbck, L., Hansson, E. (2006).
to play a role in cell volume regulation (Minieri Astrocyte-endothelial interactions at the blood-
et al., 2013; Bowens et al., 2013). Perhaps the brain barrier. Nat Rev Neurosci. 7:4153.
more attractive pharmacological candidate so Abdullaev, I.F., Rudkouskaya, A., Schools, G.P.,
far is the SUR1 inhibitor glibenclamide that Kimelberg, H.K., Mongin, A.A. (2006).
blocks the SUR1-regulated NCCa-ATP channel and Pharmaco logical comparison of swelling-
ameliorates edema formation and tissue dam- activated excitatory amino acid release and Cl
age (Simard et al., 2012). The pharmacology currents in cultured rat astrocytes. J Physiol.
issue is further complicated by the fact that it is 572:677689.
not known whether the changes in cell volume Agulhon, C., Fiacco, T.A., McCarthy, K.D. (2010).
regulation observed in pathological conditions Hippocampal short- and long-term plasticity
could also be due to a dysfunction of ancillary or are not modulated by astrocyte Ca 2+ signaling.
regulatory subunits that interact with the known Science 327:12501254.
targets. In this view, albeit in another context, it Akita, T., Okada, Y. (2011). Regulation of
was shown that the aberrant astrocytic volume bradykinin-induced activation of volume-
control involved in megalencephalic leukoen- sensitive outwardly rectifying anion channels by
cephalopathy with subcortical cysts, a genetic Ca2+ nanodomains in mouse astrocytes. J Physiol.
disease characterized by chronic white matter 589:39093927.
edema (van der Knaap et al., 2012), was due to Amiry-Moghaddam, M., Lindland, H., Zelenin, S.,
mutations of a protein called GlialCAM, which Roberg, B.A., Gundersen, B.B., Petersen, P.,
interacts with the Cl channel ClC-2 (Jeworutzki Rinvik, E., Torgner, I.A., Ottersen, O.P. (2005).
etal.,2012). Brain mitochondria contain aquaporin water
channels:Evidence for the expression of a short
Even though the amount of information now
AQP9 isoform in the inner mitochondrial mem-
available on the physiology and pathophysiol-
brane. FASEB J. 19:14591467.
ogy of astrocytes concerning water and ion
Amiry-Moghaddam, M., Ottersen, O.P. (2003). The
homeostasis is substantial, much work remains
molecular basis of water transport in the brain.
to be done to obtain a more exhaustive picture
Nat Rev Neurosci. 4:9911001.
of the underlying mechanisms. In this view, the
Amiry-Moghaddam, M., Williamson, A., Palomba, M.,
possibility of exploiting complementary inter- Eid, T., de Lanerolle, N.C., Nagelhus, E.A., Adams,
disciplinary approaches could be essential to M.E., Froehner, S.C., Agre, P., Ottersen, O.P.
identify new perspectives for addressing these (2003). Delayed K+ clearance associated with
homeostatic processes. In the past two decades aquaporin-4 mislocalization:Phenotypic defects
the development of technologies concerning cell in brains of alpha-syntrophin-null mice. Proc
isolation and manipulation, coupled with the Natl Acad Sci USA 100:1361513620.
discovery and implementation of electrophysio- Amiry-Moghaddam, M., Xue, R., Haug, F.M.,
logical, molecular biology, and cell biology tools, Neely, J.D., Bhardwaj, A., Agre, P., Adams, M.E.,
has enabled functional and molecular identifi- Froehner, S.C., Mori, S., Ottersen, O.P. (2004).
cations of plasma-membrane proteins forming Alpha-syntrophin deletion removes the perivas-
receptors and voltage-dependent ion channels cular but not endothelial pool of aquaporin-4 at
in astroglia that were thought to be uniquely the blood-brain barrier and delays the develop-
expressed in neurons (Barres, 1990; Verkhratsky ment of brain edema in an experimental model
and Steinhuser, 2000). Furthermore, the appli- of acute hyponatremia. FASEB J. 18:542544.
cation of optical technologies for monitoring Anderov, M., Antonova, T., Petrk, D., Neprasov, H.,
ion dynamics in vitro and in situ has provided Chvtal, A., Sykov, E. (2004). Voltage-
new insights into astrocytic functions (Maschio dependent potassium currents in hypertrophied
etal., 2012). It can be envisaged that in the near rat astrocytes after a cortical stab wound. Glia
future the emerging interface between biology, 48:311326.
nano-materials, and bioelectronic technologies Anderov, M., Kubinov, S., Mazel, T., Chvtal, A.,
(Benfenati etal., 2010; Benfenati etal., 2013)will Eliasson, C., Pekny, M., Sykov, E. (2001). Effect
provide novel tools to discover unexpected roles of elevated K(+), hypotonic stress, and cortical
20 Part I: Homeostatic Regulators

spreading depression on astrocyte swelling in chloride channels contributes to the diverse vol-
GFAP-deficient mice. Glia 35:189203. ume regulation in cortical astrocytes of GFAP/
Anderson, C.M., Swanson, R.A. (2000). Astrocyte EGFP mice. PLoS One 7:e29725.
glutamate transport:Review of properties, regu- Benfenati, V., Amiry-Moghaddam, M., Caprini, M.,
lation, and physiological functions. Glia 32:114. Mylonakou, M.N., Rapisarda, C., Ottersen, O.P.,
Aoki, K., Uchihara, T., Duyckaerts, C., Nakamura, A., Ferroni, S. (2007b). Expression and functional
Hauw, J.J., Wakayama, Y. (2005). Enhanced characterization of transient receptor potential
expression of aquaporin 4 in human brain vanilloid-related channel 4 (TRPV4) in rat corti-
with inflammatory diseases. Acta Neuropathol. cal astrocytes. Neuroscience 148:876892.
110:281288. Benfenati, V., Caprini, M., Dovizio, M.,
Aoki, K., Uchihara, T., Tsuchiya, K., Nakamura, A., Mylonakou, M.N., Ferroni, S., Ottersen, O.P.,
Ikeda, K., Wakayama, Y. (2003). Enhanced Amiry-Moghaddam M. (2011). An aquaporin-4/
expression of aquaporin 4 in human brain with transient receptor potential vanilloid 4 (AQP4/
infarction. Acta Neuropathol. 106:121124. TRPV4) complex is essential for cell-volume
Araque, A., Carmignoto, G., Haydon, P.G. (2001). control in astrocytes. Proc Natl Acad Sci USA
Dynamic signaling between astrocytes and neu- 108:25632568.
rons. Annu Rev Physiol. 63:795813. Benfenati, V., Caprini, M., Nicchia, G.P., Rossi, A.,
Badaut, J., Ashwal, S., Obenaus, A. (2011). Dovizio, M., Cervetto, C., Nobile, M., Ferroni, S.
Aquaporins in cerebrovascular disease:Atarget (2009). Carbenoxolone inhibits volume-regulated
for treatment of brain edema? Cerebrovasc Dis. anion conductance in cultured rat cortical astro-
31:521531. glia. Channels 3:323336.
Badaut J, Lasbennes F, Magistretti PJ, Regli L (2002). Benfenati, V., Caprini, M., Nobile, M., Rapisarda, C.,
Aquaporins in brain: distribution, physiology, Ferroni, S. (2006). Guanosine promotes the
and pathophysiology. J Cereb Blood Flow Metab. up-regulation of inward rectifier potassium cur-
22:367378. rent mediated by Kir4.1 in cultured rat cortical
Badaut, J., Petit, J.M., Brunet, J.F., Magistretti, P.J., astrocytes. J Neurochem. 98:430345.
Charriaut-Marlangue, C., Regli, L. (2004). Benfenati, V., Ferroni, S. (2010). Water transport
Distribution of Aquaporin 9 in the adult rat between CNS compartments: Functional and
brain: Preferential expression in catecholamin- molecular interactions between aquaporins and
ergic neurons and in glial cells. Neuroscience ion channels. Neuroscience 168:926940.
128:2738. Benfenati, V., Martino, N., Antognazza, M.R.,
Bai, J.Z., Lipski, J. (2010). Differential expression of Pistone, A., Toffanin, S., Ferroni, S., Lanzani, G.,
TRPM2 and TRPV4 channels and their poten- Muccini, M. (2014). Photostimulation of whole-
tial role in oxidative stress-induced cell death cell conductance in primary rat neocortical
in organotypic hippocampal culture. Neurotox. astrocytes mediated by organic semiconducting
31:204214. thin films. Adv Healthc Mater. 3:392399.
Barfod, E.T., Moore, A.L., Roe, M.W., Lidofsky, S.D. Benfenati, V., Nicchia, G.P., Svelto, M., Rapisarda, C.,
(2007). Ca 2+-activated IK1 channels associate Frigeri, A., Ferroni, S. (2007a). Functional down-
with lipid rafts upon cell swelling and mediate regulation of volume-regulated anion channels
volume recovery. J Biol Chem. 282:89848993. in AQP4 knockdown cultured rat cortical astro-
Barres, B.A., Koroshetz, W.J., Chun, L.L., Corey, D.P. cytes. J Neurochem. 100:87104.
(1990). Ion channel expression by white matter Benfenati, V, Toffanin, S., Bonetti, S., Turatti, G.,
glia:The type-1 astrocyte. Neuron 5:527544. Pistone, A., Chiappalone, M., Sagnella, A.,
Bekar, L.K., Loewen, M.E., Cao, K., Sun, X., Leis, J., Stefani, A., Generali, G., Ruani, G., Saguatti, D.,
Wang, R., Forsyth, G.W., Walz, W. (2005). Zamboni, R., Muccini, M. (2013). A transpar-
Complex expression and localization of inac- ent organic transistor structure for bidirectional
tivating Kv channels in cultured hippocampal stimulation and recording of primary neurons.
astrocytes. J Neurophysiol. 93:16991709. Nat Mater. 12:672680.
Benesova, J., Hock, M., Butenko, O., Prajerova, I., Benfenati, V., Toffanin, S., Capelli, R., Camassa,
Anderova, M., Chvtal, A. (2009). Quantification of L.M., Ferroni, S., Kaplan, D.L., Omenetto, F.G.,
astrocyte volume changes during ischemia in situ Muccini, M., Zamboni, R. (2010). A silk platform
reveals two populations of astrocytes in the cortex that enables electrophysiology and targeted drug
of GFAP/EGFP mice. J Neurosci Res. 87:96111. delivery in brain astroglial cells. Biomaterials
Benesova, J., Rusnakova, V., Honsa, P., Pivonkova, H., 31:78837891.
Dzamba, D., Kubista, M., Anderova, M. (2012). Bennett, M.V., Contreras, J.E., Bukauskas, F.F., Sez,
Distinct expression/function of potassium and J.C. (2003). New roles for astrocytes: gap junction
Microdynamics of Water and Ion Homeostasis in the Brain 21

hemichannels have something to communicate. Bordey, A., Sontheimer, H. (2000). Ion channel
Trends Neurosci. 26:610617. expression by astrocytes in situ:Comparison of
Bernardinelli, Y., Muller, D., Nikonenko, I. (2014). different CNS regions. Glia 30:2738.
Astrocyte-synapse structural plasticity. Neural Bowens, N.H., Dohare, P., Kuo, Y.H., Mongin, A.A.
Plast. 2014:232105. (2013). DCPIB, the proposed selective blocker of
Bennay, M., Langer, J., Meier, S.D., Kafitz, K.W., volume-regulated anion channels, inhibits sev-
Rose, C.R. (2008). Sodium signals in cerebel- eral glutamate transport pathways in glial cells.
lar Purkinje neurons and Bergmann glial cells Mol Pharmacol. 83:2232.
evoked by glutamatergic synaptic transmission. Bushong, E.A. (2002). Protoplasmic astrocytes in
Glia 56:11381149. CA1 stratum radiatum occupy separate anatom-
Beskina, O., Miller, A., Mazzocco-Spezzia, A., ical domains. J Neurosci. 22:183192.
Pulina, M.V., Golovina, V.A. (2007). Mechanisms Butenko, O., Dzamba, D., Benesova, J., Honsa, P.,
of interleukin-1beta-induced Ca 2+ signals in Benfenati, V., Rusnakova, V., Ferroni, S.,
mouse cortical astrocytes: Roles of store- and Anderova, M. (2012).The increased activity of
receptor-operated Ca 2+ entry. Am J Physiol Cell TRPV4 channel in the astrocytes of the adult rat
Physiol. 293:11031111. hippocampus after cerebral hypoxia/ischemia.
Binder, D.K., Nagelhus, E.A., Ottersen, O.P. (2012). PLoS One 7:e39959.
Aquaporin-4 and epilepsy. Glia 60:12031214. Butt, A.M., Kalsi, A. (2006). Inwardly rectifying
Binder, D.K., Steinhuser, C. (2006). Functional potassium channels (Kir) in central nervous sys-
changes in astroglial cells in epilepsy. Glia tem glia:Aspecial role for Kir4.1 in glial func-
54:358368. tions. J Cell Mol Med. 10:3344.
Binder, D.K., Yao, X., Zador, Z., Sick, T.J., Verkman, A.S., Charles, A.C., Merrill, J.E., Dirksen, E.R., Sanderson,
Manley, G.T. (2006). Increased seizure duration M.J. (1991). Intercellular signaling in glial
and slowed potassium kinetics in mice lacking cells:Calcium waves and oscillations in response
aquaporin-4 water channels. Glia 53:631636. to mechanical stimulation and glutamate.
Blanz, J., Schweizer, M., Auberson, M., Maier, H., Neuron 6:983992.
Muenscher, A., Hbner, C.A., Jentsch, T.J. Chaudhry, F.A., Lehre, K.P., van Lookeren
(2007). Leukoencephalopathy upon disrup- Campagne, M., Ottersen, O.P., Danbolt, N.C.,
tion of the chloride channel ClC-2. J Neurosci. Storm-Mathisen, J. (1995). Glutamate transport-
27:65816589. ers in glial plasma membranes: Highly differ-
Bloch, O., Auguste, K.I. Manley, G.T., Verkman, A.S. entiated localizations revealed by quantitative
(2006). Accelerated progression of kaolin-induced ultrastructural immunocytochemistry. Neuron
hydrocephalus in aquaporin-4-deficient mice. J 15:711720.
Cereb Blood Flow Metab. 26:15271537. Chen, H., Sun, D. (2005). The role of Na-K-Cl
Bloch, O., Papadopoulos, M.C., Manley, G.T., Verkman, co-transporter in cerebral ischemia. Neurol Res.
A.S. (2005). Aquaporin-4 gene deletion in mice 27:280286.
increases focal edema associated with staphylococ- Chen, M., Dong, Y., Simard, J.M. (2003). Functional
cal brain abscess. J Neurochem. 95:254262. coupling between sulfonylurea receptor type 1 and
Bockenhauer, D., Feather, S., Stanescu, H.C., anonselective cation channel in reactive astrocytes
Bandulik, S., Zdebik, A.A., Reichold, M., Tobin, J., from adult rat brain. J Neurosci. 23:85688577.
Lieberer, E., Sterner, C., Landoure, G., Arora, R., Chen, H., Luo, J., Kintner, D.B., Shull, G.E., Sun, D.
Sirimanna, T., Thompson, D., Cross, J.H., vant (2005). Na(+)-dependent chloride transporter
Hoff, W., Al Masry, O., Tullus, K., Yeung, S., (NKCC1)-null mice exhibit less gray and white
Anikster, Y., Klootwijk, E., Hubank, M., Dillon, matter damage after focal cerebral ischemia. J
M.J., Heitzmann, D., Arcos-Burgos, M., Knepper, Cereb Blood Flow Metab. 25:5466.
M.A., Dobbie, A., Gahl, W.A., Warth, R., Chen, M., Simard, J.M. (2001). Cell swelling and a
Sheridan, E., Kleta, R. (2009). Epilepsy, nonselective cation channel regulated by inter-
ataxia, sensorineural deafness, tubulopa- nal Ca 2+ and ATP in native reactive astrocytes
thy, and KCNJ10 mutations. N Engl J Med. from adult rat brain. J Neurosci. 21:65126521.
360:19601970. Cheng, A., van Hoek, A.N., Yeager, M., Verkman,
Bordey, A., Sontheimer, H. (1997). Postnatal devel- A.S., Mitra, A.K. (1997). Three-dimensional
opment of ionic currents in rat hippocampal organization of a human water channel. Nature
astrocytes in situ. J Neurophysiol. 78:461477. 387:627630.
Bordey, A., Sontheimer, H. (1999). Differential Chever, O., Djukic, B., McCarthy, K.D., Amzica, F.
inhibition of glial K(+) currents by 4-AP. J (2010). Implication of Kir4.1 channel in excess
Neurophysiol. 82:34763487. potassium clearance: An in vivo study on
22 Part I: Homeostatic Regulators

anesthetized glial-conditional Kir4.1 knock-out of Kir4.1 leads to glial membrane depolarization,


mice. J Neurosci. 30:1576915777. inhibition of potassium and glutamate uptake,
Chien, L.T., Hartzell, H.C. (2007). Drosophila and enhanced short-term synaptic potentiation.
bestrophin-1 chloride current is dually regu- J Neurosci. 27:1135411365.
lated by calcium and cell volume. J Gen Physiol. Eid, T., Lee T.S., Thomas, M.J., Amiry-Moghaddam,
130:513524. M., Bjrnsen, L.P., Spencer, D.D., Agre, P.,
Chvtal, A., Anderov, M., Neprasov, H., Prajerov, I., Ottersen, O.P., de Lanerolle, N.C. (2005). Loss of
Benesov, J., Butenko, O., Verkhratsky, A. (2008). perivascular aquaporin 4 may underlie deficient
Pathological potential of astroglia. Physiol Res. water and K+ homeostasis in the human epilep-
57:S101S110. togenic hippocampus. Proc Natl Acad Sci USA.
Chvtal, A., Pastor, A., Mauch, M., Sykov, E., 102:11931198.
Kettenmann, H. (1995). Distinct populations of Enkvist, M.O., McCarthy, K.D. (1994). Astroglial gap
identified glial cells in the developing rat spinal junction communication is increased by treat-
cord slice:Ion channel properties and cell mor- ment with either glutamate or high K+ concen-
phology. Eur J Neurosci. 7:129142. tration. J Neurochem. 62:489495.
Connors, N.C., Adams, M.E., Froehner, S.C., Kofuji, P. Eugenin, E.A., Basilio, D., Sez, J.C., Orellana, J.A.,
(2004). The potassium channel Kir4.1 associ- Raine, C.S., Bukauskas, F., Bennett, M.V.,
ates with the dystrophin-glycoprotein com- Berman, J.W. (2012). The role of gap junction
plex via alpha-syntrophin in glia. J Biol Chem. channels during physiologic and pathologic con-
279:2838728392. ditions of the human central nervous system. J
Connors, N.C., Kofuji, P. (2006). Potassium channel Neuroimmune Pharmacol. 7:499518.
Kir 4.1 macromolecular complex in retinal glial Ferroni, S., Marchini, C., Nobile, M., Rapisarda, C.
cells. Glia 53:124131. (1997). Characterization of an inwardly rectify-
Cornell-Bell, A.H., Finkbeiner, S.M., Cooper, M.S., ing chloride conductance expressed by cultured
Smith, S.J. (1990). Glutamate induces calcium rat cortical astrocytes. Glia 21:217227.
waves in cultured astrocytes: Long-range glial Ferroni, S., Marchini, C., Schubert, P., Rapisarda,
signaling. Science 247:470473. C. (1995). Two distinct inwardly rectify-
DAmbrosio, R., Gordon, D.S., Winn, H.R. (2002). ing conductances are expressed in long term
Differential role of KIR channel and Na(+)/K(+)- dibutyryl-cyclic-AMP treated rat cultured corti-
pump in the regulation of extracellular K(+) in cal astrocytes. FEBS Lett. 367:319325.
rat hippocampus. J Neurophysiol. 87:87102. Ferroni, S., Nobile, M., Caprini, M., Rapisarda, C.
Darby, M., Kuzmiski, J.B., Panenka, W., Feighan, D., (2000). pH modulation of an inward rectifier
MacVicar, B.A. (2003). ATP released from astro- chloride current in cultured rat cortical astro-
cytes during swelling activates chloride chan- cytes. Neuroscience 100:431438.
nels. J Neurophysiol. 89:18701877. Ferroni, S., Valente, P., Caprini, M., Nobile, M.,
De Pina-Benabou, M.H. (2001). Calmodulin kinase Schubert, P., Rapisarda, C. (2003). Arachidonic
pathway mediates the K+-induced increase in acid activates an open rectifier potassium chan-
gap junctional communication between mouse nel in cultured rat cortical astrocytes. J Neurosci
spinal cord astrocytes. J Neurosci. 21:66356643. Res. 72:363372.
Depienne, C., Bugiani, M., Dupuits, C., Galanaud, Filosa, J.A., Bonev, A.D., Straub, S.V., Meredith, A.L.,
D., Touitou, V., Postma, N., van Berkel, C., Wilkerson, M.K., Aldrich, R.W., Nelson, M.T.
Polder, E., Tollard, E., Darios, F., Brice, A., de (2006). Local potassium signaling couples neu-
Die-Smulders, C.E., Vles, J.S., Vanderver, A., ronal activity to vasodilation in the brain. Nat
Uziel, G., Yalcinkaya, C., Frints, S.G., Kalscheuer, Neurosci. 9:13971403.
V.M., Klooster, J., Kamermans, M., Abbink, T.E., Filosa, J.A., Iddings, J.A. (2013). Astrocyte regulation
Wolf, N.I., Sedel, F., van der Knaap, M.S. (2013). of cerebral vascular tone. Am J Physiol Heart Circ
Brain white matter oedema due to ClC-2 chlo- Physiol. 305:H609H619.
ride channel deficiency: An observational ana- Fischmeister, R., Hartzell, H.C. (2005). Volume
lytical study. Lancet Neurol. 12:659668. sensitivity of the bestrophin family of chloride
Dibaj, P., Kaiser, M., Hirrlinger, J., Kirchhoff, F., channels. J Physiol. 562:477491.
Neusch, C. (2007). Kir4.1 channels regulate Freeman, M.R. (2010). Specification and morpho-
swelling of astroglial processes in experi- genesis of astrocytes. Science 330:774778.
mental spinal cord edema. J Neurochem. Fukuda, A.M., Adami, A., Pop, V., Bellone, J.A., Coats,
103:26202688. J.S., Hartman, R.E., Ashwal, S., Obenaus, A.,
Djukic, B., Casper, K.B., Philpot, B.D., Chin, L.S., Badaut, J. (2013). Posttraumatic reduction of
McCarthy, K.D. (2007). Conditional knock-out edema with aquaporin-4 RNA interference
Microdynamics of Water and Ion Homeostasis in the Brain 23

improves acute and chronic functional recovery. Kir4.1 potassium channels in the sclerotic
J Cereb Blood Flow Metab. 33:16211632. hippocampus of patients with mesial tempo-
Furukawa, T., Ogura, T., Katayama, Y., Hiraoka, M. ral lobe epilepsy. J Neuropathol Exp Neurol.
(1998). Characteristics of rabbit ClC-2 cur- 71:814825.
rent expressed in Xenopus oocytes and its con- Hibino, H., Fujita, A., Iwai, K., Yamada, M.,
tribution to volume regulation. Am J Physiol. Kurachi, Y. (2004). Differential assembly of
274:C500C512. inwardly rectifying K+ channel subunits, Kir4.1
Gebremedhin, D., Yamaura, K., Zhang, C., Bylund, J., and Kir5.1, in brain astrocytes. J Biol Chem.
Koehler, R.C., Harder, D.R. (2003). Metabotropic 279:4406544073.
glutamate receptor activation enhances the Hibino, H., Inanobe, A., Furutani, K., Murakami, S.,
activities of two types of Ca 2+-activated K+ chan- Findlay, I., Kurachi, Y. (2010). Inwardly rec-
nels in rat hippocampal astrocytes. J Neurosci. tifying potassium channels: Their structure,
23:16781687. function, and physiological roles. Physiol Rev.
Giaume, C., Tabernero, A., Medina, J.M. (1997). 90:291366.
Metabolic trafficking through astrocytic gap Higashi, K., Fujita, A., Inanobe, A., Tanemoto, M.,
junctions. Glia 21:114123. Doi, K., Kubo, T., Kurachi, Y. (2001). An inwardly
Girouard, H., Bonev, A.D., Hannah, R.M., Meredith, A., rectifying K(+) channel, Kir4.1, expressed in
Aldrich, R.W., Nelson, M.T. (2010). Astrocytic astrocytes surrounds synapses and blood vessels
endfoot Ca2+ and BK channels determine both in brain. Am J Physiol Cell Physiol. 281:922931.
arteriolar dilation and constriction. Proc Natl Hirrlinger, P.G., Wurm, A., Hirrlinger, J., Bringmann,
Acad Sci USA 107:38113816. A., Reichenbach, A. (2008). Osmotic swelling
Gnatenco, C., Han, J., Snyder, A.K., Kim, D. (2002). characteristics of glial cells in the murine hip-
Functional expression of TREK-2 K+ chan- pocampus, cerebellum, and retina in situ. J
nel in cultured rat brain astrocytes. Brain Res. Neurochem. 105:14051417.
931:5667. Ho, J.D., Yeh, R., Sandstrom, A., Chorny, I., Harries,
Grnder, S., Thiemann, A., Pusch, M., Jentsch, T.J. W.E., Robbins, R.A., Miercke, L.J., Stroud, R.M.
(1992). Regions involved in the opening of CIC-2 (2009). Crystal structure of human aquaporin 4
chloride channel by voltage and cell volume. at 1.8 Aand its mechanism of conductance. Proc
Nature 360:759762. Natl Acad Sci USA 106:74377442.
Guadagno, E., Moukhles, H. (2004). Laminin-induced Hoffmann, E.K., Lambert, I.H., Pedersen, S,F.
aggregation of the inwardly rectifying potassium (2009). Physiology of cell volume regulation in
channel, Kir4.1, and the water permeable chan- vertebrates. Physiol Rev. 89:193277.
nel, AQP4, via a dystroglycan-containing com- Holthoff, H., Witte, O.W. (2000). Directed spatial
plex in astrocytes. Glia 47:138149. potassium redistribution in rat neocortex. Glia
Haj-Yasei, N.N., Jensen, V., Vindedal, G.F., 29:288292.
Gundersen, G.A., Klungland, A., Ottersen, O.P., Holthoff, K., Witte, O. W. (1996). Intrinsic opti-
Hvalby, O., Nagelhus, E.A. (2011). Evidence that cal signals in rat neocortical slices measured
compromised K+ spatial buffering contributes with near-infrared dark-field microscopy reveal
to the epileptogenic effect of mutations in the changes in extracellular space. J Neurosci.
human Kir4.1 gene (KCNJ10). Glia 59:16351642. 16:27402749.
Hamilton, N.B., Attwell, D. (2010). Do astrocytes Hoogland, T.M., Kuhn, B. (2010). Recent develop-
really exocytose neurotransmitters? Nat Rev ments in the understanding of astrocyte function
Neurosci. 11:227238. in the cerebellum in vivo. Cerebellum 9:264271.
Hamann, S., Zeuthen, T., La Cour, M., Nagelhus, Horio, Y. (2001). Potassium channels of glial
E.A., Ottersen, O.P., Agre, P., Nielsen, S. (1998). cells:Distribution and function. Jpn J Pharmacol.
Aquaporins in complex tissues: distribution 87:16.
of aquaporins 15 in human and rat eye. Am J Horio, Y., Hibino, H., Inanobe, A. (1997). Clustering
Physiol. 274:C13321345. and enhanced activity of an inwardly rectify-
Hussinger, D., Grg, B. (2010). Interaction of oxi- ing potassium channel, Kir4.1, by an anchor-
dative stress, astrocyte swelling and cerebral ing protein, PSD-95/SAP90. J Biol Chem.
ammonia toxicity. Curr Opin Clin Nutr Metab 272:1288512888.
Care 13:8792. Hsu, M.S., Seldin, M., Lee, D.J., Seifert, G.,
Heuser, K., Eid, T., Lauritzen, F., Thoren, A.E., Steinhuser, C., Binder, D.K. (2011). Laminar-
Vindedal, G.F., Taubll, E., Gjerstad, L., specific and developmental expression of aqua-
Spencer, D.D., Ottersen, O.P., Nagelhus, E.A., porin-4 in the mouse hippocampus. Neuroscience
de Lanerolle, N.C. (2012). Loss of perivascular 178:2132.
24 Part I: Homeostatic Regulators

Hua, S.Z., Gottlieb, P.A., Heo, J., Sachs, F. (2010). Kahle, K.T., Simard, J.M., Staley, K.J., Nahed, B.V.,
A mechanosensitive ion channel regulat- Jones, P.S., Sun, D. (2009). Molecular mecha-
ing cell volume. Am J Physiol Cell Physiol. nisms of ischemic cerebral edema:Role of elec-
298:14241430. troneutral ion transport. Physiology (Bethesda)
Hua, X., Malarkey, E.B., Sunjara, V., Rosenwald, S.E., 24:257265.
Li, W.H., Parpura, V. (2004). Ca2+-dependent Kettenmann, H., Backus, K.H., Schachner, M. (1987).
glutamate release involves two classes of endo- Gamma-aminobutyric acid opens Cl channels
plasmic reticulum Ca(2+) stores in astrocytes. J in cultured astrocytes. Brain Res. 404:19.
Neurosci Res. 76:8697. Kettenmann, H., Verkhratsky, A. (2008).
Hwang, E.M., Kim, E., Yarishkin, O., Woo, D.H., Neuroglia:The 150years after. Trends Neurosci.
Han, K.S., Park, N., Bae, Y., Woo, J., Kim, D., 31:653659.
Park, M., Lee, C.J., Park, J.Y. (2014). A Kimelberg, H.K. (2005). Astrocytic swelling in cere-
disulphide-linked heterodimer of TWIK-1 and bral ischemia as a possible cause of injury and
TREK-1 mediates passive conductance in astro- target for therapy. Glia 50:389397.
cytes. Nat Commun. 5:3227. Kimelberg, H.K., Feustel, P.J., Jin, Y., Paquette, J.,
Iadecola, C., Nedergaard, M. (2007). Glial regulation Boulos, A., Keller, R.W. Jr., Tranmer, B.I. (2000).
of the cerebral microvasculature. Nat Neurosci. Acute treatment with tamoxifen reduces isch-
10:13691376. emic damage following middle cerebral artery
Illarionova, N.B., Gunnarson, E., Li, Y., Brismar, H., occlusion. Neuroreport 11:26752679.
Bondar, A., Zelenin, S., Aperia, A. (2010). Kimelberg, H.K., Jin, Y., Charniga, C., Feustel, P.J.
Functional and molecular interactions between (2003). Neuroprotective activity of tamoxi-
aquaporins and Na, K-ATPase. Neuroscience fen in permanent focal ischemia. J Neurosurg.
168:915925. 99:138142.
Inoue, H., Okada, Y., (2007). Roles of volume-sensitive Kimelberg, H.K., Nedergaard, M. (2010). Functions
chloride channel in excytotoxic neuronal injury. of astrocytes and their potential as therapeutic
J Neurosci. 27:14451455. targets. Neurotherap. 7:338353.
Ishii, M., Horio, Y., Tada, Y., Hibino, H., Inanobe, A., Kirischuk, S., Kettenmann, H., Verkhratsky, A.
Ito, M., Yamada, M., Gotow, T., Uchiyama, Y., (2007). Membrane currents and cytoplasmic
Kurachi, Y. (1997). Expression and clustered dis- sodium transients generated by glutamate trans-
tribution of an inwardly rectifying potassium port in Bergmann glial cells. Pflugers Arch.
channel, KAB-2/Kir4.1, on mammalian retinal 454:245252.
Mller cell membrane:Their regulation by insu- Kofuji, P., Biedermann, B., Siddharthan, V., Raap, M.,
lin and laminin signals. J Neurosci. 17:77257735. Iandiev, I., Milenkovic, I., Thomzig, A., Veh, R.W.,
Jabs, R., Paterson, I.A., Walz, W. (1997). Bringmann, A., Reichenbach, A. (2002). Kir
Qualitative analysis of membrane currents potassium channel subunit expression in reti-
in glial cells from normal and gliotic tissue nal glial cells:Implications for spatial potassium
in situ: Down-regulation of Na+ current and buffering. Glia 39:292303.
lack of P2 purinergic responses. Neuroscience Kofuji, P., Ceelen, P., Zahs, K.R., Surbeck, L.W.,
81:847860. Lester, H.A., Newman, E.A. (2000). Genetic
Jarius S, Paul F, Franciotta D, Waters P, Zipp F, inactivation of an inwardly rectifying potassium
Hohlfeld R, Vincent A, Wildemann B (2008). channel (Kir4.1 subunit) in mice: Phenotypic
Mechanisms of disease:aquaporin-4 antibodies impact in retina. J Neurosci. 20:57335740.
in neuromyelitis optica. Nat Clin Pract Neurol. Kofuji, P., Newman, E.A. (2004). Potassium buffer-
4:202214. ing in the central nervous system. Neuroscience
Jentsch, T.J., Stein, V., Weinreich, F., Zdebik, A.A. 129:10451056.
(2002). Molecular structure and physiologi- Kressin, K., Kuprijanova, E., Jabs, R., Seifert, G.,
cal function of chloride channels. Physiol Rev. Steinhuser, C. (1995). Developmental regulation
82:503568. of Na+ and K+ conductances in glial cells of mouse
Jeworutzki, E., Lpez-Hernndez, T., hippocampal brain slices. Glia 15:173187.
Capdevila-Nortes, X., Sirisi, S., Bengtsson, L., Kubo, Y., Baldwin, T.J., Jan, Y.N., Jan, L.Y. (1993).
Montolio, M., Zifarelli, G., Arnedo, T., Mller, Primary structure and functional expression
C.S., Schulte, U., Nunes, V., Martnez, A., of a mouse inward rectifier potassium channel.
Jentsch, T.J., Gasull, X., Pusch, M., Estvez, R. Nature 362:127133.
(2012). GlialCAM, a protein defective in a leuko- Kucheryavykh, L.Y., Kucheryavykh, Y.V., Inyushin, M.,
dystrophy, serves as a ClC-2 Cl(-) channel auxil- Shuba, Y.M., Sanabria, P., Cubano, L.A.,
iary subunit. Neuron 73:951961. Skatchkov, S.N., Eaton, M.J. (2009). Ischemia
Microdynamics of Water and Ion Homeostasis in the Brain 25

increases TREK-2 channel expression in astro- Lopatin, A.N., Makhina, E.N., Nichols, C.G. (1994).
cytes: Relevance to glutamate clearance. Open Potassium channel block by cytoplasmic poly-
Neurosci J. 3:4047. amines as the mechanism of intrinsic rectifica-
Kucheryavykh, L.Y., Nichols, C.G., Maldonado, tion. Nature 372:366369.
H.M., Baksi, K., Reichenbach, A., Skatchkov, S.N., MacFarlane, S.N., Sontheimer, H. (1997).
Eaton, M.J. (2007). Downregulation of Kir4.1 Electrophysiological changes that accom-
inward rectifying potassium channel subunits pany reactive gliosis in vitro. J Neurosci.
by RNAi impairs potassium transfer and gluta- 17:73167329.
mate uptake by cultured cortical astrocytes. Glia MacFarlane, S.N., Sontheimer, H. (1998). Spinal cord
55:274281. astrocytes display a switch from TTX-sensitive
Kuga, N., Sasaki, T., Takahara, Y., Matsuki, N., to TTX-resistant sodium currents after
Ikegaya, Y. (2011). Large-scale calcium waves injury-induced gliosis in vitro. J Neurophysiol.
traveling through astrocytic networks in vivo. J 79:22222226.
Neurosci. 31:26072614. MacVicar, B.A., Feighan, D., Brown, A., Ransom, B.
Langer, J., Rose, C.R. (2009). Synaptically induced (2002). Intrinsic optical signals in the rat optic
sodium signals in hippocampal astrocytes in nerve: Role for K(+) uptake via NKCC1 and
situ. J Physiol. 587:58595877. swelling of astrocytes. Glia 37:114123.
Langer, J., Stephan, J., Theis, M., Rose, C.R. (2012). MacVicar, B., Hochman, A.D. (1991). Imaging of
Gap junctions mediate intercellular spread of synaptically evoked intrinsic optical signals in
sodium between hippocampal astrocytes in situ. hippocampal slices. J Neurosci. 11:14581469.
Glia 60:239252. MacVicar, B.A., Thompson, R.J. (2010). Non-junction
Lascola, C.D., Nelson, D.J., Kraig, R.P. (1998). functions of pannexin-1 channels Trends
Cytoskeletal actin gates a Cl channel in neocor- Neurosci. 33:93102.
tical astrocytes. J Neurosci. 18:16791692. MacVicar, B.A., Tse, F.W., Crichton, S.A.,
Lee, D.J., Hsu, M.S., Seldin, M.M., Arellano, J.L., Kettenmann, H. (1989). GABA-activated Cl
Binder, D.K. (2012). Decreased expression of the channels in astrocytes of hippocampal slices. J
glial water channel aquaporin-4 in the intrahip- Neurosci. 9:35773583.
pocampal kainic acid model of epileptogenesis. Makara, J.K., Rappert, A., Matthias, K.,
Exp Neurol. 235:246255. Steinhuser, C., Spt, A., Kettenmann, H.
Lehre, K.P., Levy, L.M., Ottersen, O.P., (2003). Astrocytes from mouse brain slices
Storm-Mathisen, J., Danbolt, N.C. (1995). express ClC-2-mediated Cl currents regulated
Differential expression of two glial glutamate during development and after injury. Mol Cell
transporters in the rat brain: Quantitative and Neurosci. 23:521530.
immunocytochemical observations. J Neurosci. Malarkey, E.B., Ni, Y., Parpura, V. (2008). Ca 2+ entry
15:18351853. through TRPC1 channels contributes to intracel-
Li, G., Liu, Y., Olson, J.E. (2002). Calcium/ lular Ca 2+ dynamics and consequent glutamate
calmodulin-modulated chloride and taurine release from rat astrocytes. Glia 56:821835.
conductances in cultured rat astrocytes. Brain Manley, G.T., Fujimura, M., Ma, T., Noshita, N.,
Res. 925:18. Filiz, F., Bollen, A.W., Chan, P., Verkman, A.S.
Li, L., Head, V., Timpe, L.C. (2001). Identification (2000). Aquaporin-4 deletion in mice reduces
of an inward rectifier potassium channel gene brain edema after acute water intoxication and
expressed in mouse cortical astrocytes. Glia ischemic stroke. Nat Med. 6:159163.
33:5771. Marmarou, A. (2007). A review of progress in under-
Liang, D., Bhatta, S., Gerzanich, V., Simard, J.M. standing the pathophysiology and treatment of
(2007). Cytotoxic edema:Mechanisms of patho- brain edema. Neurosurg Focus 22:E1.
logical cell swelling. Neurosurg Focus 22:E2. Maschio MD, Beltramo R, De Stasi AM, Fellin T.
Liedtke, W., Tobin, D.M., Bargmann, C.I., Friedman, (2012). Two-photon calcium imaging in the
J.M. (2003). Mammalian TRPV4 (VR-OAC) intact brain. Adv Exp Med Biol. 740:83102.
directs behavioral responses to osmotic and Mathiisen, T.M., Lehre, K.P., Danbolt, N.C.,
mechanical stimuli in Caenorhabditis elegans. Ottersen, O.P. (2010). The perivascular astroglial
Proc Natl Acad Sci USA 100:1453114536. sheath provides a complete covering of the brain
Longden, T.A., Dunn, K.M., Draheim, H.J., Nelson, microvessels:An electron microscopic 3D recon-
M.T., Weston, A.H., Edwards, G. (2011). struction. Glia 58:10941103.
Intermediate-conductance calcium-activated Matsuda, H. (1991). Magnesium gating of the
potassium channels participate in neurovascular inwardly rectifying K+ channel. Annu Rev
coupling. Br J Pharmacol. 164:922933. Physiol. 53:289298.
26 Part I: Homeostatic Regulators

Medina-Ceja, L., Cordero-Romero, A., Morales- Nedergaard, M., Ransom, B., Goldman, S.A. (2003).
Villagrn, A. (2008). Antiepileptic effect of New roles for astrocytes: Redefining the func-
carbenoxolone on seizures induced by 4-ami- tional architecture of the brain. Trends Neurosci.
nopyridine: a study in the rat hippocampus and 26:523530.
entorhinal cortex. Brain Res. 1187:7481. Neusch, C., Papadopoulos, N., Mller, M., Maletzki,
Minieri, L., Pivonkova, H., Caprini, M., Harantova, L., I., Winter, S.M., Hirrlinger, J., Handschuh, M.,
Anderova, M., Ferroni, S. (2013). The inhibi- Bhr, M., Richter, D.W., Kirchhoff, F., Hlsmann,
tor of volume-regulated anion channels DCPIB S. (2006). Lack of the Kir4.1 channel subunit
activates TREK potassium channels in cultured abolishes K+ buffering properties of astro-
astrocytes. Br J Pharmacol. 168:12401254. cytes in the ventral respiratory group: impact
Minieri, L., Pivonkova, H., Harantova, L., Anderova, on extracellular K+ regulation. J Neurophysiol.
M., Ferroni. S. (2015). Intracellular Na(+) inhib- 95:18431852.
its volume-regulated anion channel in rat corti- Newman, E.A., Frambach, D.A., Odette, L.L.
cal astrocytes. J Neurochem. 132:286300. (1984). Control of extracellular potassium lev-
Mizuno, A., Matsumoto, N., Imai, M., Suzuki, M. els by retinal glial cell K+ siphoning. Science
(2003). Impaired osmotic sensation in mice 225:11741175.
lacking TRPV4. Am J Physiol Cell Physiol. Nicchia, G.P., Mastrototaro, M., Rossi, A., Pisani, F.,
285:96101. Tortorella, C., Ruggieri, M., Lia, A., Trojano, M.,
Molofsky, A.V., Krencik, R., Ullian, E.M., Tsai, H.H., Frigeri, A., Svelto M. (2009). Aquaporin-4
Deneen, B., Richardson, W.D., Barres, B.A., orthogonal arrays of particles are the target
Rowitch, D.H. (2012). Astrocytes and disease: for neuromyelitis optica autoantibodies. Glia
A neurodevelopmental perspective. Genes Dev. 57:13631373.
26:891907. Nicchia, G.P., Srinivas, M., Li, W., Brosnan, C.F.,
Montell, C. (2001). Physiology, phylogeny, and func- Frigeri, A., Spray, D.C. (2005). New possible roles
tions of the TRP superfamily of cation channels. for aquaporin-4 in astrocytes:Cell cytoskeleton
Sci STKE.90. and functional relationship with connexin43.
Montell, C., Rubin, G.M. (1989). Molecular charac- FASEB J. 19:16741676.
terization of the Drosophila trp locus:Aputative Nichols, C.G., Lopatin, A.N. (1997). Inward rec-
integral membrane protein required for photo- tifier potassium channels. Ann Rev Physiol.
transduction. Neuron 2:13131323. 59:171191.
Nagelhus, E.A., Amiry-Moghaddam, M., Bergersen, Nicholson, C., Sykova, E. (1998). Extracellular space
L.H., Bjaalie, J.G., Eriksson, J., Gundersen, V., structure revealed by diffusion analysis. Trends
Leergaard, T.B., Morth, J.P., Storm-Mathisen, J., Neurosci. 21:207215.
Torp, R., Walhovd, K.B., Tnjum, T. (2013). Nilius, B., Owsianik, G. (2011). The transient recep-
The glia doctrine: Addressing the role of glial tor potential family of ion channels. Genome
cells in healthy brain ageing. Mech Ageing Dev. Biol. 12:218.
134:449459. Nol, G., Belda, M., Guadagno, E., Micoud, J.,
Nagelhus, E.A., Horio, Y., Inanobe, A., Fujita, A., Klcker, N., Moukhles, H. (2005). Dystroglycan
Haug, F.M., Nielsen, S., Kurachi, Y., Ottersen, O.P. and Kir4.1 coclustering in retinal Mller glia
(1999). Immunogold evidence suggests that cou- is regulated by laminin-1 and requires the
pling of K+-siphoning and water transport in rat PDZ-ligand domain of Kir4.1. J Neurochem.
retinal Mller cells is mediated by a coenrich- 94:691702.
ment of Kir4.1 and AQP4 in specific membrane Noell, S., Fallier-Becker, P., Beyer, C., Krger, S.,
domains. Glia 26:4754. Mack, A.F., Wolburg, H. (2007). Effects of agrin
Nagelhus, E.A., Mathiisen, T.M., Ottersen, O.P. on the expression and distribution of the water
(2004). Aquaporin-4 in the central nervous channel protein aquaporin-4 and volume regu-
system: Cellular and subcellular distribution lation in cultured astrocytes. Eur J Neurosci.
and coexpression with Kir4.1. Neuroscience 26:21092118.
129:905913. OConnor, E.R., Kimelberg, H.K. (1993). Role of
Nagy, J.I., Rash, J.E. (2000). Connexins and gap junc- calcium in astrocyte volume regulation and in
tions of astrocytes and oligodendrocytes in the the release of ions and amino acids. J Neurosci.
CNS. Brain Res Brain Res Rev. 32:2944. 13:26382650.
Nau, R., Brck, W. (2002). Neuronal injury in bac- Odette, L.L., Newman, E.A. (1988). Model of potas-
terial meningitis:Mechanisms and implications sium dynamics in the central nervous system.
for therapy. Trends Neurosci. 25:3845. Glia 1:198210.
Microdynamics of Water and Ion Homeostasis in the Brain 27

Olsen, M.L., Higashimori, H., Campbell, S.L., Pasantes-Morales, H., Cruz-Rangel, S. (2010). Brain
Hablitz, J.J., Sontheimer, H. (2006). Functional volume regulation: Osmolytes and aquaporin
expression of Kir4.1 channels in spinal cord perspectives. Neuroscience 168:871884.
astrocytes. Glia 53:516528. Pasantes-Morales, H., Vzquez-Jurez, E. (2012).
Olsen, M.L., Sontheimer, H. (2008). Functional Transporters and channels in cytotoxic astrocyte
implications for Kir4.1 channels in glial biol- swelling. Neurochem Res. 37:23792387.
ogy: From K+ buffering to cell differentiation. J Psler, D., Gabriel, S., Heinemann, U. (2007).
Neurochem. 107:589601. Two-pore-domain potassium channels con-
Olson, J.E., Li, G.Z., Wang, L., Lu, L. (2004). tribute to neuronal potassium release and glial
Volume-regulated anion conductance in cul- potassium buffering in the rat hippocampus.
tured rat cerebral astrocytes requires calmodulin Brain Res. 1173:1426.
activity. Glia 46:391401. Patel, A.J., Honor, E. (2001). Properties and modu-
Orkand, R.K. (1986). Glial-interstitial fluid exchange. lation of mammalian 2P domain K+ channels.
Ann NY Acad Sci. 481:269272. Trends Neurosci. 24:339346.
Orkand, R.K., Nicholls, J.G., Kuffler, S.W. (1966). Pellerin, L., Bouzier-Sore, A. K., Aubert, A., Serres,
Effect of nerve impulses on the membrane poten- S., Merle, M., Costalat, R., Magistretti, P.J. (2007).
tial of glial cells in the central nervous system of Activity-dependent regulation of energy metabo-
amphibia. J Neurophysiol. 29:788806. lism by astrocytes:An update. Glia 55:12511262.
stby, I., yehaug, L., Einevoll, G.T., Nagelhus, E.A., Pivonkova, H., Benesova, J., Butenko, O., Chvtal,
Plahte, E., Zeuthen T., Lloyd, C.M., Ottersen, O.P., A., Anderova, M. (2010). Impact of global cere-
Omholt, S.W. (2009). Astrocytic mechanisms bral ischemia on K+ channel expression and
explaining neural-activity-induced shrink- membrane properties of glial cells in the rat hip-
age of extraneuronal space. PLoS Comput Biol. pocampus. Neurochem Int. 57:783794.
5:e1000272. Pizzo, P., Burgo, A., Pozzan, T., Fasolato, C.
Owsianik, G., Dhoedt, D., Voets, T., Nilius, B. (2001). Role of capacitative calcium entry on
(2006). Structure-function relationship of the glutamate-induced calcium influx in type-I rat
TRP channel superfamily. Rev Physiol Biochem cortical astrocytes. J Neurochem. 79:98109.
Pharmacol. 156:6190. Pohl, P. (2004). Combined transport of water and
Paemeleire, K., Leybaert, L. (2000). Ionic changes ions through membrane channels. Biol Chem.
accompanying astrocytic intercellular calcium 385:921926.
waves triggered by mechanical cell damaging Preston, G.M., Carroll, T.P., Guggino, W.B., Agre,
stimulation. Brain Res. 857:235245. P. (1992). Appearance of water channels in
Papadopoulos, M.C., Manley, G.T., Krishna, S., Xenopus oocytes expressing red cell CHIP28
Verkman, A.S. (2004). Aquaporin-4 facilitates protein. Science 256:385387.
reabsorption of excess fluid in vasogenic brain Price, D.L., Ludwig, J.W., Mi, H., Schwarz, T.L.,
edema. FASEB J. 18:12913. Ellisman, M.H. (2002). Distribution of rSlo
Papadopoulos, M.C., Verkman, A.S. (2013). Ca 2+-activated K+ channels in rat astrocyte peri-
Aquaporin water channels in the nervous sys- vascular endfeet. Brain Res. 956:183193.
tem. Nat Rev Neurosci. 14:265277. Qiu, Z., Dubin, A.E., Mathur, J., Tu, B., Reddy, K.,
Park, H., Oh, S.J., Han, K.S., Woo, D.H., Park, H., Miraglia, L.J., Reinhardt, J., Orth, A.P., Patapoutian,
Mannaioni, G., Traynelis, S.F., Lee, C.J. (2009). A. (2014). SWELL1, a plasma membrane protein, is
Bestrophin-1 encodes for the Ca 2+-activated an essential component of volume-regulated anion
anion channel in hippocampal astrocytes. J channel. Cell 157:447458.
Neurosci. 29:1306313073. Rama Rao, K.V., Verkman, A.S., Curtis, K.M. (2014).
Parkerson, K.A., Sontheimer, H. (2003). Contribution Aquaporin-4 deletion in mice reduces encepha-
of chloride channels to volume regulation of lopathy and brain edema in experimental acute
cortical astrocytes. Am J Physiol Cell Physiol. liver failure. Neurobiol Dis. 63:222228.
284:14601467. Ransom, C.B., Ransom, B.R., Sontheimer, H. (2000).
Parkerson, K.A., Sontheimer H. (2004). Biophysical Activity-dependent extracellular K+ accumula-
and pharmacological characterization of hypo- tion in rat optic nerve:The role of glial and axo-
tonically activated chloride currents in cortical nal Na+ pumps. J Physiol. 522:427442.
astrocytes. Glia 46:419436. Ransom, C.B., Sontheimer, H. (1995). Biophysical
Parpura, V., Zorec, R. (2010). Gliotransmission: and pharmacological characterization of
Exocytotic release from astrocytes. Brain Res inwardly rectifying K+ currents in rat spinal cord
Rev. 63:8392. astrocytes. J Neurophysiol. 73:333346.
28 Part I: Homeostatic Regulators

Rash, J.E., Yasumura, T., Hudson, C.S., Agre, P., Santiago, M.F., Veliskova J., Patel, N.K., Lutz, S.E.,
Nielsen, S. (1998). Direct immunogold label- Caille, D., Charollais, A., Meda, P., Scemes, E.
ling of aquaporin-4 in square arrays of astrocyte (2011). Targeting pannexin1 improves seizure
and ependymocyte plasma membranes in rat outcome. PLoS One 6:e25178.
brain and spinal cord. Proc Natl Acad Sci USA Scemes, E., Giaume, C. (2006). Astrocyte calcium
95:1198111986. waves: What they are and what they do. Glia
Reinert, M., Khaldi, A., Zauner, A., Doppenberg, E., 54:716725.
Choi, S., Bullock, R. (2000). High extracellular Scemes, E., Spray, D.C. (2012). Extracellular K+ and
potassium and its correlates after severe head astrocyte signaling via connexion and pannexin
injury: Relationship to high intracranial pres- channels. Neurochem Res. 37:23102316.
sure. Neurosurg Focus8:e10. Scholl, U.I., Choi, M., Liu, T., Ramaekers, V.T.,
Reichenbach, A., Bringmann, A. (2013). New func- Hausler, M.G., Grimmer, J., Tobe, S.W., Farhi,
tions of Mller cells. Glia 61:651678. A., Nelson-Williams, C., Lifton, R.P. (2009).
Reyes, R.C., Verkhratsky, A., Parpura, V. (2013). Seizures, sensorineural deafness, ataxia, mental
TRPC1-mediated Ca2+ and Na+ signalling in retardation, and electrolyte imbalance (SeSAME
astroglia: differential filtering of extracellular syndrome) caused by mutations in KCNJ10. Proc
cations. Cell Calcium 54:120125. Natl Acad Sci USA 106:58425847.
Rivera-Guevara, C., Camacho, J. (2011). Tamoxifen Seifert, G., Httmann, K., Binder, D.K., Hartmann,
and its new derivatives in cancer research. Recent C., Wyczynski, A., Neusch, C., Steinhuser, C.
Pat Anticancer Drug Discov. 6:237245. (2009). Analysis of astroglial K+ channel expres-
Rose, C.R., Karus, C. (2013). Two sides of the same sion in the developing hippocampus reveals
coin: Sodium homeostasis and signaling in a predominant role of the Kir4.1 subunit. J
astrocytes under physiological and pathophysi- Neurosci. 29:74747488.
ological conditions. Glia 61:11911205. Seifert, G., Schilling, K., Steinhuser, C. (2006).
Rose, C.R., Ransom, B.R. (1997). Gap junctions Astrocyte dysfunction in neurological disor-
equalize intracellular Na+ concentration in ders:a molecular perspective. Nat Rev Neurosci.
astrocytes. Glia 20:299307. 7:194206.
Rossi, A., Moritz, T.J., Ratelade, J., Verkman, A.S. Sk, A., Smith, R.L., Freund, T.F. (2000). Distribution
(2012). Super-resolution imaging of aquaporin-4 of chloride channel-2-immunoreactive neuronal
orthogonal arrays of particles in cell membranes. and astrocytic processes in the hippocampus.
J Cell Sci. 125:44054412. Neuroscience 101:5165.
Rouach, N., Koulakoff, A., Abudara, V., Willecke, K., Silver, I.A., Ereciska, M. (1997). Energetic demands
Giaume, C. (2008). Astroglial metabolic net- of the Na+/K+ ATPase in mammalian astrocytes.
works sustain hippocampal synaptic transmis- Glia 21:3545.
sion. Science 322:15511555. Simard, J.M., Chen, M., Tarasov, K.V., Bhatta, S.,
Roy, M.L., Saal, D., Perney, T., Sontheimer, H., Ivanova, S., Melnitchenko, L., Tsymbalyuk,
Waxman, S.G., Kaczmarek, L.K. (1996). N., West, G.A., Gerzanich, V. (2006). Newly
Manipulation of the delayed rectifier Kv1.5 expressed SUR1-regulated NC(Ca-ATP) channel
potassium channel in glial cells by antisense oli- mediates cerebral edema after ischemic stroke.
godeoxynucleotides. Glia 18:177184. Nat Med. 12:433440.
Ruiz-Ederra, J., Zhang, H., Verkman, A.S. (2007). Simard, J.M., Kent, T.A., Chen, M., Tarasov, K.V.,
Evidence against functional interaction between Gerzanich, V. (2007a). Brain edema in focal isch-
aquaporin-4 water channels and Kir4.1 potas- aemia:Molecular pathophysiology and theoreti-
sium channels in retinal Mller cells. J Biol cal implications. Lancet Neurol. 6:258268.
Chem. 282:2186621872. Simard, J.M., Tarasov, K.V., Gerzanich, V. (2007b)
Saadoun, S., Papadopoulos, M.C., Davies, D.C., Non-selective cation channels, transient receptor
Krishna, S., Bell, B.A. (2002). Aquaporin-4 potential channels and ischemic stroke. Biochim
expression is increased in oedematous human Biophys Acta. 1772:947957.
brain tumours. J Neurol Neurosurg Psych. Simard, J.M., Woo, S.K., Schwartzbauer, G.T.,
72:262265. Gerzanich, V. (2012). Sulfonylurea receptor 1 in
Saadoun, S., Papadopoulos, M.C., Krishna, S. (2003). central nervous system injury:Afocused review.
Water transport becomes uncoupled from K+ J Cereb Blood Flow Metab. 32:16991717.
siphoning in brain contusion, bacterial menin- Simard, M., Arcuino, G., Takano, T., Liu, Q.S.,
gitis, and brain tumours:Immunohistochemical Nedergaard, M. (2003). Signaling at the gliovas-
case review. Clin Pathol. 56:972975. cular interface. J Neurosci. 23:92549262.
Microdynamics of Water and Ion Homeostasis in the Brain 29

Simard, M., Nedergaard, M. (2004). The neuro- Taniguchi, M., Yamashita, T., Kumura, E., Tamatani, M.,
biology of glia in the context of water and ion Kobayashi, A., Yokawa, T., Maruno, M., Kato, A.,
homeostasis. Neuroscience 129:877896. Ohnishi, T., Kohmura, E., Tohyama, M.,
Somjen, G.G. (1988). Nervenkitt: Notes on the his- Yoshimine, T. (2000). Induction of aquaporin-4
tory of the concept of neuroglia. Glia1:29. water channel mRNA after focal cerebral ischemia
Somjen, G.G. (2001). Mechanisms of spreading in rat. Brain Res Mol Brain Res. 78:131137.
depression and hypoxic spreading depression- Theis, M., Shl, G., Eiberger, J., Willecke, K.
like depolarization. Physiol Rev. 81:10651096. (2005). Emerging complexities in identity and
Sontheimer, H., Fernandez-Marques, E., Ullrich, N., function of glial connexins. Trends Neurosci.
Pappas, C.A., Waxman, S.G. (1994). Astrocyte 28:188195.
Na+ channels are required for maintenance Thiemann, A., Grnder, S., Pusch, M., Jentsch, T.J.
of Na+/K(+)-ATPase activity. J Neurosci. (1992). A chloride channel widely expressed
14:24642475. in epithelial and non-epithelial cells. Nature
Sontheimer, H., Ransom, B.R., Cornell-Bell, A.H., 356:5760.
Black, J.A., Waxman, S.G. (1991). Na(+)-current Thio, C.L., Sontheimer, H. (1993). Differential mod-
expression in rat hippocampal astrocytes in vitro: ulation of TTX-sensitive and TTX-resistant
Alterations during development. J Neurophysiol. Na+ channels in spinal cord astrocytes follow-
65:319. ing activation of protein kinase C. J Neurosci.
Steiner, E., Enzmann, G.U., Lin, S., Ghavampour, S., 13:48894897.
Hannocks, M.J., Zuber, B., Regg, M.A., Sorokin, L., Thompson, R.J., MacVicar, B.A. (2008). Connexin
Engelhardt, B. (2012). Loss of astrocyte polar- and pannexin hemichannels of neurons and
ization upon transient focal brain ischemia as a astrocytes. Channels 2:8186.
possible mechanism to counteract early edema Tse, F.W., Fraser, D.D., Duffy, S., MacVicar, B.A.
formation. Glia 60:16461659. (1992). Voltage-activated K+ currents in acutely
Steinhuser, C., Seifert, G., Bedner, P. (2012). isolated hippocampal astrocytes. J Neurosci.
Astrocyte dysfunction in temporal lobe epi- 12:17811788.
lepsy: K+ channels and gap junction coupling. Vajda, Z., Pedersen, M., Fchtbauer, E.M., Wertz, K.,
Glia 60:11921202. Stdkilde-Jrgensen, H., Sulyok, E., Dczi, T.,
Stobart, J.L., Anderson, C.M. (2013). Multifunctional Neely, J.D., Agre, P., Frkiaer, J., Nielsen, S.
role of astrocytes as gatekeepers of neuronal (2002). Delayed onset of brain edema and mis-
energy supply. Front Cell Neurosci.7:38. localization of aquaporin-4 in dystrophin-null
Strohschein, S., Httmann, K., Gabriel, S., Binder, D.K., transgenic mice. Proc Natl Acad Sci USA
Heinemann, U. Steinhuser, C., (2011). Impact of 99:1313113136.
aquaporin-4 on K+ buffering and gap junction cou- van der Knaap, M.S., Boor, I., Estvez, R. (2012).
pling in the hippocampus. Glia 59:973980. Megalencephalic leukoencephalopathy with sub-
Su, G., Kintner, D.B., Sun, D. (2002). Contribution cortical cysts:Chronic white matter oedema due
of Na(+)-K(+)-Cl(-) cotransporter to high- to a defect in brain ion and water homoeostasis.
[K(+)](o)- induced swelling and EAA release Lancet Neurol. 11:973985.
in astrocytes. Am J Physiol Cell Physiol. Verkhratsky, A., Orkand, R.K., Kettenmann, H.
282:11361146. (1988). Glial calcium:Homeostasis and signaling
Sykov, E., Nicholson, C. (2008). Diffusion in brain function. Physiol Rev. 78:99141.
extracellular space. Physiol. Rev. 88:12771340. Verkhratsky, A., Rodrguez, J.J., Parpura, V. (2012).
Tait, M.J., Saadoun, S., Bell, B.A., Verkman, Calcium signalling in astroglia. Mol Cell
A.S., Papadopoulos, M.C. (2010). Increased Endocrinol. 353:4556.
brain edema in aqp4-null mice in an experi- Verkhratsky, A., Steinhuser, C. (2000). Ion channels
mental model of subarachnoid hemorrhage. in glial cells. Brain Res Brain Res Rev. 32:380412.
Neuroscience 167:6067. Verkman, A.S. (2012). Aquaporins in clinical medi-
Takano, T., Kang, J., Jaiswal, J.K., Simon, S.M., Lin, cine. Ann Rev Med. 63:303316.
J.H., Yu, Y., Li, Y., Yang, J., Dienel, G., Zielke, Vizuete, M.L., Venero, J.L., Vargas, C., Ilundin,
H.R., Nedergaard, M. (2005). Receptor-mediated A.A., Echevarra, M., Machado, A., Cano, J.
glutamate release from volume sensitive chan- (1999). Differential upregulation of aquaporin-4
nels in astrocytes. Proc Natl Acad Sci USA mRNA expression in reactive astrocytes after
102:1646616471. brain injury: Potential role in brain edema.
Takano, T., Oberheim, N., Cotrina, M.L., Neurobiol Dis. 6:245258.
Nedergaard, M. (2009). Astrocytes and ischemic Voets, T., Szcs, G., Droogmans, G., Nilius, B. (1995).
injury. Stroke 40:812. Blockers of volume-activated Cl- currents inhibit
30 Part I: Homeostatic Regulators

endothelial cell proliferation. Pflugers Arch. Pharmacology. LXXVI. Current progress in the
431:132134. mammalian TRP ion channel family. Pharmacol
Volterra, A., Steinhuser, C. (2004). Glial modula- Rev. 62:381404.
tion of synaptic transmission in the hippocam- Wu, X., Liu, Y., Chen, X., Sun, Q., Tang, R., Wang, W.,
pus. Glia 47:249257. Yu, Z., Xie, M. (2013). Involvement of TREK-1
Voss, L.J., Jacobson, G., Sleigh, J.W., Steyn-Ross, activity in astrocyte function and neuroprotec-
A., Steyn-Ross, M. (2009). Excitatory effects tion under simulated ischemia conditions. J Mol
of gap junction blockers on cerebral cortex Neurosci. 49:499506.
seizure-like activity in rats and mice. Epilepsia Xiong, H., Li, C., Garami, E., Wang, Y., Ramjeesingh,
50:19711978. M., Galley, K., Bear, C.E. (1999). ClC-2 activation
Voss, F.K., Ullrich, F., Munch, J., Lazarow, K., Lutter, modulates regulatory volume decrease. J Membr
D., Mah, N., Andrade-Navarro, M.A., von Kries, Biol. 167:215221.
J.P., Stauber, T., Jentsch, T.J. (2014). Identification Yan, Y., Dempsey, R. J., Flemmer, A., Forbush, B.,
of LRRC8 heteromers as an essential component Sun, D. (2003). Inhibition of Na(+)-K(+)-Cl(-)
of the volume-regulated anion channel VRAC. cotransporter during focal cerebral ischemia
Science 344:634638. decreases edema and neuronal damage. Brain
Wallraff, A., Kohling, R., Heinemann, U., Theis, Res. 961:2231.
M., Willecke, K., Steinhauser, C. (2006). The Yang, B., Zador, Z., Verkman, A.S. (2008). Glial cell
impact of astrocytic gap junctional coupling on aquaporin-4 overexpression in transgenic mice
potassium buffering in the hippocampus. Glia accelerates cytotoxic brain swelling. J Biol Chem.
47:249257. 283:1528015286.
Walz, W., Hertz, L. (1982). Ouabain-sensitive and Ye, Z.C., Oberheim, N., Kettenmann, H., Ransom,
ouabain-resistant net uptake of potassium into B.R. (2009). Pharmacological cross-inhibition
astrocytes and neurons in primary cultures. J of connexin hemichannels and swelling acti-
Neurochem. 39:7077. vated anion channels. Glia 57:258269.
Walz, W., Hertz, L. (1984). Intense furosemide-sensitive Zeng, H.K., Wang, Q.S., Deng, Y.Y., Fang, M.,
potassium accumulation in astrocytes in the pres- Chen, C.B., Fu, Y.H., Jiang, W.Q., Jiang, X.
ence of pathologically high extracellular potas- (2010). Hypertonic saline ameliorates cerebral
sium levels. J Cereb Blood Flow Metab. 4:301304. edema through downregulation of aquaporin-4
Walz, T., Smith, B.L., Agre, P., Engel, A. (1994). The expression in the astrocytes. Neuroscience
three-dimensional structure of human erythro- 166:878885.
cyte aquaporin CHIP. EMBO J. 13:29852993. Zhang, H., Verkman, A.S. (2008). Aquaporin-4 inde-
Walz, W. (2002). Chloride/anion channels in glial pendent Kir4.1 K+ channel function in brain glial
cell membranes. Glia 40:110. cells. Mol Cell Neurosci. 37:110.
Walz, W., Hinks, E.C. (1985). Carrier-mediated KCl Zhang, X.D., Morishima, S., Ando-Akatsuka, Y.,
accumulation accompanied by water movements Takahashi, N., Nabekura, T., Inoue, H., Shimizu,
is involved in the control of physiological K+ lev- T., Okada, Y. (2004). Expression of novel iso-
els by astrocytes. Brain Res. 343:4451. forms of the CIC-1 chloride channel in astrocytic
Wang, M., Song, J., Xiao, W., Yang, L., Yuan, J., glial cells in vitro. Glia 47:4657.
Wang, W., Yu, Z., Xie, M. (2012). Changes in Zhang, Y., Zhang, H., Feustel, P.J., Kimelberg, H.K.
lipid-sensitive two-pore domain potassium (2008). DCPIB, a specific inhibitor of volume
channel TREK-1 expression and its involvement regulated anion channels (VRACs), reduces
in astrogliosis following cerebral ischemia in infarct size in MCAo and the release of gluta-
rats. J Mol Neurosci. 46:384392. mate in the ischemic cortical penumbra. Exp
Woo, S.K., Kwon, M.S., Ivanov, A., Gerzanich, Neurol. 210:514520.
V., Simard, J.M. (2013). The sulfonylurea Zhou, M., Xu, G., Xie, M., Zhang, X., Schools,
receptor 1 (Sur1)-transient receptor potential G.P., Ma, L., Kimelberg, H.K., Chen, H. (2009).
melastatin 4 (TRPM4) channel. J Biol Chem. TWIK-1 and TREK-1 are potassium channels
288:36553667. contributing significantly to astrocyte pas-
Wu, L.J., Sweet, T.B., Clapham, D.E. (2010). sive conductance in rat hippocampal slices.
International Union of Basic and Clinical J Neurosci. 29:85518564.
2
Homeostatic Control ofAdenosine Levels
and Functions intheBrain
XUESONG CHEN, M AHMOUD L .SOLIM A N, LIA NG HUI,
A N D J O N AT H A N D . G E I G E R

INTRODUCTION many different anabolic and catabolic enzymes


Applying the concept of homeostasis, as intro- and transporters that help regulate the levels;
duced and elaborated on by Claude Bernard and there are multiple receptor systems upon which
Walter Cannon, to the regulation of the levels the purines act to elicit their actions; multiple
and actions of the purines adenosine and ATP cell types even in the brain are involved; and all
in the brain is both complex and interesting. Of of these sites of actions are subject to transcrip-
course there are many homeostatically regulated tional, translational, and epigenetic modifica-
physiologically-important brain functions, and tions. Indeed, it seems reasonable to propose that
the levels and actions of a variety of important these regulations and modifications are, in fact,
physiologically active ions, gases, chemicals, homeostatic mechanisms. In this chapter we dis-
and compounds help control these functions, cuss issues important to understanding where,
including ATP and adenosinethe focus of this when, and how these homeostatically regulated
chapter. brain energy metabolites act in the brain with a
When thinking about the homeostatic regu- focus on adenosine.
lation of purines, one needs to pay close attention
to not only normal fluctuations in their levels BR AIN LEVELS
and actions, something akin to noise (Woods OFADENOSINE
and Wilson, 2013)but also to signals caused by As a neuromodulator and a homeostatic regula-
increased levels and actions of ATP and adenosine tor, adenosine is involved in a variety of impor-
as occurs during neuronal stimulation, depolari- tant central nervous system (CNS) functions,
zation, metabolic insults, and pathological states. including bioenergetics, sleep and arousal,
It is in this latter context of signals that Andrew learning and memory, and motor control. These
Newby coined the term retaliatory metabolite actions of adenosine depend not only on the
(Newby et al., 1990); although not specifically levels of extracellular adenosine but also on its
discussed at that time some 25years ago, inher- access to and the distribution of adenosine recep-
ent in that term are the concepts of homeostatic tor subtypes through which the actions of adeno-
regulation and negative feedback. sine are mediated at the cellularlevel.
Adenosine is produced largely from the Under physiological conditions, extracel-
metabolism of the excitatory purine ATP, and the lular adenosine concentrations in the brain are
largely inhibitory actions of adenosine (certainly in the 30 to 200 nanomolar range (Fredholm
those mediated through adenosine A1 receptors) et al., 2011). These levels reflect a balance and
help to provide negative feedback (retaliation) homeostatic regulation of the production,
against initial brain stimuli such as increases release, metabolism, and uptake of adenosine
in energy demand and decreases in energy sup- and its precursors, as shown in Figure 2.1. In the
plies. However, understanding the homeostatic brain, purine salvage is key to the maintenance
regulation of purine levels and actions is com- of purine levels because the de novo synthe-
plicated because adenosine can be formed both sis of adenine nucleotides is limited and the de
intracellularly and extracellularly; there are novo synthesis of adenosine is absent. Adenosine
32 Part I: Homeostatic Regulators

FIGURE 2.1: Adenosine formation, metabolism, and transport. Neurons and astrocytes express
enzymes that catalyze the synthesis and removal of adenosine. Adenosine can be formed intracellularly from
catabolism of ATP and S-adenosylhomocysteine (SAH) or extracellularly from breakdown of released ATP by
ecto 5-nucleotidase (5N). Equilibrative nucleoside transporters (ENTs) are bidirectional and facilitate the move-
ment of adenosine into or out of cells along adenosine concentration gradients.

levels are regulated intracellularly and extracel- intracellular and extracellular levels of adeno-
lularly by enzymes that catalyze its synthesis, sine (King et al., 2006; Latini and Pedata, 2001;
5-nucleotidases and S-adenosylhomocysteine Peng et al., 2005). Equilibrative and concentra-
hydrolase, and enzymes that catalyze its removal, tive (sodium-dependent) nucleoside transporters
S-adenosylhomocysteine hydrolase, adenosine are present in the brain (Geiger and Fyda, 1991;
deaminase (ADA), and adenosine kinase (ADK; Parkinson et al., 1994). Equilibrative nucleoside
King eta l., 2006; Latini and Pedata, 2001; Peng eta l., transporters (ENTs) are bidirectional and facili-
2005). Intracellularly, adenosine formation is cat- tate the movement of adenosine into and out of
alyzed from 5-AMP by cytosolic 5-nucleotidases cells along adenosine concentration gradients
and from S-adenosylhomocysteine by (Baldwin et al., 2005). In contrast, adenosine
S-adenosylhomocysteine hydrolase. Extra movement into and out of cells through con-
cellularly, adenosine formation from ATP centrative nucleoside transporters follows Na+
and cAMP is catalyzed by ATPase, apyrase, electrochemical gradients. Of these nucleoside
and ecto-5-nucleotidase. Adenosine catabo- transporters, ENTs play a major role in adeno-
lism appears to occur mainly intracellularly sine transport (Geiger and Fyda, 1991; Parkinson
where S-adenosylhomocysteine hydrolase etal., 1994). Not only do these transporters par-
catalyzes the conversion of adenosine to ticipate in the homeostatic control of adenosine
S-adenosylhomocysteine, ADA catalyzes the levels, but the transporters themselves are home-
conversion of adenosine to inosine, and ADK ostatically controlled by brain levels of adenosine
catalyzes the conversion of adenosine to 5-AMP. (Sebastio, 2011; Delicado etal., 1994; Hertz and
The activities and expression levels of these Matz,1989).
enzymes are themselves homeostatically regu- Adenosine levels can also be controlled
lated by various influences, including the levels through action potential dependent release
of adenine nucleotides and adenosine (Cui etal., mechanisms. Unlike more traditional neu-
2009; Kowaluk and Jarvis, 2000; Orford and rotransmitters, adenosine is not packaged in
Saggerson,1996). secretory vesicles but, as mentioned, can be
In addition to the aforementioned enzymes, released through ENTs. Under conditions that
nucleoside transporters help regulate the increase neuronal metabolic load by, for instance,
Homeostatic Control of Adenosine Levels and Functions in the Brain 33

neuronal firing, the intracellular catabolism of 2001; Fredholm et al., 2005), all of which are
ATP can increase thus causing a concentration seven-transmembrane G-protein coupled recep-
gradient-dependent rapid efflux of adenosine tors (Figure 2.2). Adenosine receptors exhibit dif-
through ENTs. Alternatively, ATP is packaged ferent binding affinities for adenosine (Dunwiddie
in secretory vesicles and can be released where- and Masino, 2001; Olah and Stiles, 1995); A1
upon it can be metabolized to adenosine albeit receptors have the highest affinity (70 nM),
somewhat more slowly (Wall and Dale, 2013). A 2A receptors have high affinity (150 nM); and
Activation of excitatory neurons can result in A 2B and A3 have lower affinities ( 5000 nM). In
glutamate release whereupon it can activate the CNS, expression levels of adenosine recep-
excitatory amino acid receptors and increase lev- tors vary between regions: A1 receptors are the
els of intracellular Ca2+ and increased exocytotic most abundant and widespread with enrich-
release of ATP that can then be metabolized to ment in hippocampus, cerebellum, and cerebral
adenosine (Pankratov etal., 2007). Extracellular cortex; A2A receptors are highly concentrated
adenosine once formed may be preferentially in basal ganglia and limbic areas; and A2B and
taken up into astrocytes via ENTs because astro- A3 receptors are expressed at much lower levels
cytes maintain very low levels of intracellular (Fredholm etal., 2005; Gomes etal., 2011). At the
adenosine due to high activity levels of ADK, an cellular level, expression levels of A1 receptors
enzyme that catalyzes the conversion of adeno- are higher in neurons than in glial cells (astro-
sine to 5-AMP (Boison etal.,2010). cytes, microglia, and oligodendrocytes), whereas
A 2A receptors are expressed on neurons, glial
ADENOSINE RECEPTORS cells, and endothelial cells. For neurons, A1 and
AND DISTRIBUTION A 2A receptors are densely enriched in presynap-
There are four subtypes of cell surface adeno- tic active zones as well as in postsynaptic den-
sine receptors; A1, A 2A, A 2B, and A3 (Benarroch, sities, but for striatal neurons A 2A receptors are
2008; Boison etal., 2010; Dunwiddie and Masino, predominantly located at postsynaptic densities

FIGURE 2.2 Adenosine as a neuromodulator and homeostatic regulator of brain bioenergetics.


All four subtypes of adenosine receptors are expressed on neurons, astrocytes, and endothelial cells that make up
tripartite synapses. With their direct contacts with neuronal synapses and cerebral vasculature, astrocytes are
uniquely situated to regulate the involvement of adenosine in the homeostatic regulation of brain bioenergetics
and functions. On one hand, adenosine derived from astrocytes regulates both neurotransmitter release and syn-
aptic integrity, thus playing a major role in establishing set points of inhibitory tone within neuronal networks. On
the other hand, astrocyte-derived adenosine can regulate blood flow and blood-brain barrier integrity and modu-
late the entry of energy-yielding nutrients into the brain, thus regulating brain bioenergetics homeostatically.
34 Part I: Homeostatic Regulators

(Cunha, 2005). The effects of adenosine on brain adenosines functions. Moreover, prolonged acti-
functions are regulated mainly through activa- vation of adenosine receptors can lead to aden-
tion of A1 and A 2A receptors because the normal osine receptor desensitization and changes in
levels of extracellular adenosine preferentially adenosine receptor trafficking. In addition, and
activate A1 and A 2A receptors, because A1 and A2A depending on the subtype of adenosine receptor,
receptor expression levels are relatively high, and trafficking kinetics can be differently regulated
because manipulation of A 2B and A3 receptors has (Klaasse et al., 2008; Mundell and Kelly, 2011).
been shown to have relatively modest impacts on Such activity-dependent desensitization and traf-
brain function (Cunha, 2005; Fredholm et al., ficking mechanisms add another layer of homeo-
2005; Gomes etal., 2011). This contrasts with the static control of adenosine signaling.
actions of adenosine under pathological condi-
tions when adenosine levels rise dramatically and ADENOSINE ASA
activate lower affinity A2B and A3 receptors. H O M E O S TAT I C
R E G U L AT O R O F B R A I N
ADENOSINE RECEPTOR BIOENERGETICS
SIGNALING The brain in general and neurons in particu-
For many years adenosine receptor signaling lar require a large and almost constant supply
was thought to be restricted to cAMP formation, of energy (Rolfe and Brown, 1997). Because the
with adenosine A1 and A3 receptors coupling to brain has a very limited capacity to store read-
Gi proteins that once activated inhibit adenylate ily useable bioenergetic molecules, it requires
cyclase and adenosine A2A and A2B receptors cou- a continuous supply of blood containing oxy-
pling to Gs proteins that once activated activate gen and energy-yielding nutrients. Thus even
adenylate cyclase. However, adenosine receptor very brief interruptions (seconds) in blood flow
signaling is far more complex than this; depend- and/or absence of oxygen results in dramatic
ing on sites and degree of activation, adenosine losses of cellular energy, loss of consciousness,
receptor subtype activation can lead to coupling and profound abnormalities in brain function
to more than one G protein and to different sig- up to and including increased susceptibility to
nal transduction systems. For example, A1 recep- insult-induced neuronal celldeath.
tors can couple to Gi1/2/3 and Go; A2A receptors ATP is not only a readily available source
can couple to Gs, Golf, G15, and G16; A2B recep- of adenosine; it is also essential for a variety of
tors can couple to Gs and Gq/11; A3 receptors can neuronal functions, notably the maintenance
couple to Gi/2,3 and Gq/11 (Fredholm etal., 2001; of transmembrane ion gradients, cAMP forma-
Fredholm et al., 2011); and adenosine receptors tion, kinase-mediated phosphorylation, vesicu-
can couple to G subunits (Schwindinger etal., lar storage of neurotransmitters, and glutamate
2010; Yao etal.,2002). metabolism. Brain levels of adenosine are nearly
Adenosine receptor signaling is further 10,000-fold lower than ATP (Delaney and Geiger,
complicated by the degree to which A1 and A2A 1996; Pazzagli et al., 1993; Van Wylen et al.,
receptors can oligomerize as homodimers and 1986), which means that huge increases in adeno-
their ability to form heterodimers with, for sine levels can originate from minor fluctuations
example, ATP, dopamine, and glutamate recep- in ATP levels. In general, adenosine inhibits neu-
tors (Ciruela etal., 2012; Fredholm etal., 2011). ronal activity and in so doing allows target cells
A1 receptors form heterodimers with P2Y1- and like neurons and astrocytes to homeostatically
P2Y2-purinergic receptors (Suzuki et al., 2006; regulate their energy demands to match energy
Yoshioka et al., 2001), dopamine D1 receptors supplies (Meghji and Newby, 1990; Newby etal.,
(Ismayilova et al., 2004), and metabotropic glu- 1990). However, it appears clear that the ability of
tamate type 1 receptors (mGluR1; Ciruela etal., adenosine levels to increase as brain energy lev-
2001). A2A receptors form heterodimers with els decrease is quite different depending on the
dopamine D2 receptors (Hakansson etal., 2006; severity of the stimulation (Shepel et al., 2005).
Higley and Sabatini, 2010), dopamine D3 recep- With no significant external excitatory stimula-
tors, metabotropic glutamate type 5 receptors tion, ATP levels remain within a physiological
(mGluR5; Nishi et al., 2003), and cannabinoid range and adenosine increases modestly to lev-
CB1 receptors (Carriba et al., 2007). Such oli- els where adenosine A1 receptors would be pref-
gomerization can enable the fine-tuning of erentially activated. With significant external
adenosine signaling, and such aspects of sign- excitatory stimulation as occurs during excito-
aling can add to the homeostatic regulation of toxicity, ATP levels decrease below physiological
Homeostatic Control of Adenosine Levels and Functions in the Brain 35

thresholds and adenosine increases to levels nutrients into the brain (ORegan, 2005;
capable of upregulating and activating adeno- Pelligrino etal.,2011).
sine A2A receptors. When the brain is exposed Clearly, functional coupling between neu-
to more drastic insults such as global ischemia, ronal activity and blood flow is central to the
ATP levels drop dramatically and adenosine lev- homeostatic regulation of brain bioenergetics.
els increase markedly as the brain attempts to For example, when neurons are activated, cer-
homeostatically prevent cell death (Atkinson and ebral blood flow increases in localized brain
Walton, 1967) by activating low affinity adeno- regions thereby increasing temporally and spa-
sine A3 receptors and mobilizing tissue repair tially the supply of energy-yielding nutrients.
mechanisms (Abbracchio etal., 1997; von Lubitz Mechanistically this occurs because neuronal
etal.,1999). stimulation leads to glutamate release, the release
Astrocytes can outnumber neurons 9:1 of ATP from astrocytes, the metabolism of ATP
(White and Venkatesh, 2011)and are one of the to adenosine, and the activation of adenosine A2A
major sites for adenosine formation (Pascual receptors that are highly expressed on cerebral
etal., 2005). Activation of excitatory amino acid blood vessels; the end result is vasodilation and
receptors on astrocytes can lead to increased increased cerebral blood flow (Mills etal., 2011;
synaptic levels of adenosine originating from ORegan, 2005; Pelligrino etal., 2010). In addition
ATP released through vesicles or hemichannels to neurovascular coupling, astrocyte-derived
(Iglesias etal., 2009; Kang etal., 2008). Astrocytes adenosine can affect the permeability of the
can also increase extracellular levels of adeno- blood brain barrier through coordinated activa-
sine originating from ATP released through tion of A1 and A2A receptors, both of which are
volume-regulated ion channels (Darby et al., expressed on brain endothelial cells (Carman
2003). In addition, adenosine can be released via etal.,2011).
nucleoside transporters (King etal., 2006; Latini As such, adenosine is able to homeostatically
and Pedata, 2001; Peng et al., 2005). Astrocytes regulate brain microenvironments for optimal
may also play an important role in enzyme-linked functioning of neuronal networks. Moreover,
regulation of adenosine levels because ADK is these same homeostatically-controlled mecha-
mainly expressed in astrocytes and is known nisms and set points are activated in a variety of
to catalyze the salvage of adenosine to 5-AMP acute and chronic neurological and neuropsy-
(Boison etal., 2010; Studer etal.,2006). chiatric disorders such as brain ischemia, sleep
With their direct contacts with neuronal syn- deprivation, epilepsy, Huntingtons disease,
apses and cerebral vasculature, astrocytes are Parkinsons disease, Alzheimers disease, depres-
uniquely situated to regulate the involvement of sion, bipolar disorder, and schizophrenia (Gomes
adenosine in the homeostatic regulation of brain etal., 2011), and altering adenosine signaling may
bioenergetics and functions. Astrocytes, neurons, therefore be exploited therapeutically against
and endothelial cells make up tripartite synapses, these acute and chronic neurological disorders
and together these cells function to coordinate (Cunha, 2005; Gomes etal.,2011).
synaptic activity and networks. Moreover, aden-
osine derived from astrocytes in the functional ADENOSINE ASA
vicinity of tripartite synapses appears to play a N E U R O M O D U L AT O R
major role in establishing set points of inhibitory As a neuromodulator, adenosine working
tone within neuronal networks (Figure 2.2). For through A1 and A2A receptors helps tune and fine-
example, in actively functioning cells, energy lev- tune synapses, in part, by homeostatically con-
els decline and adenosine levels increase to levels trolling inhibition and excitation (Ribeiro and
that preferentially activate inhibitory cell surface Sebastiao, 2010; Sebastiao and Ribeiro, 2009a,
adenosine A1 receptors. Thus at a very local level, 2009b). An important aspect of this synaptic
adenosine formation provides a rapidly acting tuning is the control of neurotransmitter release
negative feedback mechanism that prevents fur- and synaptic plasticity through the coordinated
ther brain energy demands and homeostatically actions of inhibitory adenosine A1 and excita-
regulates brain energy metabolism. Alternatively, tory adenosine A2A receptors (Dias et al., 2013).
endfeet of astrocytes are integral elements of the Indeed, adenosine has been shown to modulate
blood-brain barrier where astrocyte-derived excitatory neurotransmitters, including gluta-
adenosine can homeostatically regulate blood mate, acetylcholine, and serotonin, as well as
flow, maintain blood brain barrier integrity, the inhibitory neurotransmitter GABA. Such
and modulate the entry of energy-yielding actions underlie critical physiological functions
36 Part I: Homeostatic Regulators

of adenosine, including sleep/arousal (Brown expression on astrocytes, where activation of A1


etal., 2012), learning and memory (Prediger and receptors within A2A/A1 oligomers decreases the
Takahashi, 2005; Wei et al., 2011), and motor expression of GABA transporters thus inhibiting
control (Ciruela et al., 2012; Cunha, 2005; Fuxe GABA transport into astrocytes. On the other
et al., 2010). Here we focus on how adenosine hand, activation of A 2A receptors within A 2A/
adjusts dynamically the strength of excitatory A1 oligomers increases the expression of GABA
and inhibitory inputs reaching a given neuron transporters thus facilitating GABA transport
thus controlling neuronal firingrates. into astrocytes (Cristovao-Ferreira et al., 2013).
The tonic release of adenosine via neuronal In addition, adenosine could affect GABA recep-
ENTs can activate presynaptic A1 receptors and tor signaling at postsynaptic densities because
inhibit the presynaptic release of glutamate by A1 receptor-dependent activation of potassium
inhibiting calcium influx through voltage-gated channels increases cell membrane conductance
calcium channels, including N-type and and thus has a shunting effect on GABA AR cur-
P/Q-type calcium channels (Ambrosio etal., 1997; rents (Ilie etal.,2012).
Brambilla etal., 2005; Gundlfinger etal., 2007). Such actions of adenosine on the strength
Such tonic release can also activate postsynap- of excitatory and inhibitory input may play an
tic A1 receptors, inhibit N-methyl-D-aspartate important role in the regulation of sleep/arousal,
(NMDA) receptors (de Mendonca et al., 1995; quite possibly the least well-understood homeo-
Hartwick et al., 2004) and activate potas- static mechanism of any organism (Brown etal.,
sium channels (including G protein-coupled 2012). Prolonged wakefulness can increase
inwardly rectifying potassium channels [GIRKs], adenosine levels (Kalinchuk et al., 2011) and
ATP-sensitive potassium channels, and small might promote sleep through activation of A1
conductance calcium-activated potassium chan- and A2A receptors in specific brain regions. In
nels), thus leading to postsynaptic neuronal basal forebrain, activation of A1 receptors, which
hyperpolarization (Chung et al., 2009; Clark are upregulated during prolonged wakefulness
et al., 2009; Hosseinzadeh and Stone, 1998; (Basheer etal., 2007), inhibit glutamatergic neu-
Kawamura etal., 2010). On the other hand, aden- rons and wake-promoting cholinergic neurons
osine derived from the release of ATP can activate (Yang et al., 2013). In the tuberomammillary
A2A receptors abundantly located in presynaptic hypothalamic nucleus, activation of A1 recep-
active zones, where it can facilitate glutamate tors results in inhibition of wake-promoting
release (Ciruela etal., 2006)and inhibit glutamate histaminergic neurons (Hong et al., 2005). In
uptake into astrocytes (Matos etal., 2012). A1 and the ventrolateral preoptic area, activation of A2A
A2A receptors are expressed pre- and postsynapti- receptors activates sleep-promoting neurons and
cally, where these receptors can heterodimerize inhibits arousal-promoting regions (Urade and
and help functionally tune adenosine and gluta- Hayaishi, 2011). In addition, A 2A receptors in the
mate signaling. At low adenosine levels, adeno- nucleus accumbens may play a role in sleep-wake
sine preferentially activates A1 receptors, and at regulation, especially caffeines wake-promoting
higher levels it activates A 2A receptors within A 2A/ effects (Lazarus etal., 2011; Zhang etal.,2013).
A1 oligomers. Such a hierarchical activation of A1 Through coordinated activation of high-
receptors or A2A receptors allows functional tun- affinity inhibitory A1 and excitatory A 2A recep-
ing of adenosine signaling and the control of glu- tors, adenosine has been recognized as an impor-
tamate release (Dias etal.,2013). tant homeostatic regulator of synaptic plasticity
Compared to glutamatergic transmis- (Dias etal., 2013)with a unique ability to allow
sion, much less is known about how adenosine neurons to dynamically modify their synaptic
affects GABAergic transmission. It is generally structure and function as a result of activity.
accepted that adenosine does not affect directly Here we discuss how adenosine affects synaptic
the presynaptic release of GABA (Thompson plasticity of glutamatergic and dopaminergic
et al., 1992) and that the effects of adenosine systems.
on GABA synaptic transmission are mainly Adenosine has been shown to provide
achieved by inhibiting the excitatory synaptic homeostatic regulation of glutamatergic sys-
inputs to GABAergic neurons via A1 receptors tems, essential for two well-described forms
(Yang et al., 2013). Recent studies indicate that of synaptic plasticity: long-term potentia-
adenosine regulates GABA levels directly at the tion (LTP) and long-term depression (LTD;
synaptic cleft by altering GABA transporter Abraham, 2008; Queenan et al., 2012). LTP is
Homeostatic Control of Adenosine Levels and Functions in the Brain 37

a form of activity-dependent plasticity that through D2 receptors. In general, adenosine


results in a persistent enhancement of synap- exerts antagonist effects on dopamine recep-
tic efficacy, whereas LTD is a long-lasting and tors. In direct pathway neurons, A1 receptors
activity-dependent decrease in synaptic efficacy. expressed at presynaptic dopaminergic nerve
LTP and LTD are expressed by synapse-specific terminals exert an inhibitory modulation
regulation by AMPA and NMDA glutamate on dopamine release (Ferre et al., 1997). At a
receptors (Lee et al., 2010). Tonic release of postsynaptic site, A1 receptors form heterodi-
adenosine through ENTs can activate postsyn- mers with D1 receptors, thus negatively affect-
aptic A1 receptors, GIRK channels, and home- ing the binding characteristics of D1 receptors
ostatically control LTP (Chung et al., 2009). (Ismayilova et al., 2004). Behaviorally, adeno-
Activation of postsynaptic A1 receptors also sine A1 receptor antagonists potentiate the
affects LTD. Activation of A1 receptors blocks motor effects of D1R agonists through antago-
mGluR1-mediated LTD by decreasing the nistic A1D1 receptor interactions (Popoli etal.,
ligand sensitivity of mGluR1 (Kamikubo etal., 1996). In direct pathway neurons, adenosine
2013), possibly through direct interactions A 2A receptors are more densely expressed in
between A1 receptors and mGluR1 in the form the striatum than anywhere else in the brain
of A1/mGluR1 heterodimers (Ciruela et al., (Cunha, 2001; Cunha, 2005), and striatal post-
2001). High-frequency bursts of stimulation, as synaptic A 2A receptors are selectively localized
occur in the induction of LTP, can trigger the in the dendritic membranes of medium spiny
release of ATP, and adenosine levels formed neurons in the indirect-pathway, where they
from the released ATP are high enough to acti- can form heterodimers with D2 receptors and
vate postsynaptic A 2A receptors (Cunha, 2001). exert strong functional antagonistic interac-
Such activation of postsynaptic A 2A recep- tions with D2 receptors (Hakansson etal., 2006;
tors can induce different forms of short- and Higley and Sabatini, 2010). Stimulation or
long-term synaptic plasticity by enhancing the blockade of striatal postsynaptic A 2A receptors
surface expression of GluR1 (Dias etal., 2012). counteracts and potentiates, respectively, the
Activation of A 2A receptors can also potentiate motor-activating effects of D2 receptor stimula-
NMDA receptor-mediated LTP (Rebola et al., tion (Ferre etal., 1997; Schiffmann etal., 2007).
2008), possibly through co-activation of A 2A and In addition, A1 receptors expressed in neurons
mGluR5 in the form of A 2A/mGluR5 heterodi- of indirect pathway could affect the function
mers (Tebano et al., 2005). Thus, homeostatic of A 2A receptors though A 2A-A1 heterodimeri-
regulation of synaptic plasticity in the form of zation (Karcz-Kubicha etal., 2003). Such find-
LTP and LTD by adenosine suggests that aden- ings suggest that the coupling of adenosine and
osine plays an important role in learning and dopamine receptors is a key mechanism for
memory. Indeed, altering A1 and A 2A receptor the homeostatic regulation of the dopaminer-
signaling affects learning and memory in ani- gic system (Stromberg etal., 2000). Given that
mal models (Prediger and Takahashi, 2005; Wei dopamine plays an important role in control-
et al., 2011), and this opens the door to possi- ling locomotor activity, motivation, and addic-
ble exploitation of therapeutic interventions tion (Arias-Carrion et al., 2010; Beninger,
against such conditions as Alzheimers disease 1983; Salamone and Correa, 2002), the ability
where memory is often seriously impaired. of adenosine to homeostatically regulate dopa-
Adenosine also homeostatically regulates minergic systems reinforces the concept that
dopaminergic systems in basal ganglia where A1 adenosine levels and/or signaling plays a role in
and A 2A receptors are highly expressed (Ciruela the pathogenesis of Parkinsons disease, schizo-
et al., 2012; Cunha, 2005; Fuxe et al., 2010). phrenia, and drug addiction (Fuxe etal.,2010).
Dopamine D1 receptors are primarily expressed
on striato-nigral neurons of the direct pathway ADENOSINE-BASED
where they couple to stimulatory G s proteins, THER APEUTICS
and dopamine D2 receptors are highly expressed Because adenosine, as a neuromodulator and a
on striato-pallidal neurons of the indirect path- homeostatic regulator, is involved in a variety
way where they are coupled to inhibitory G i of important CNS functions, including bioener-
proteins. Dopamine induces motor activation getics, sleep and arousal, learning and memory,
by activating the direct pathway through D1 and motor control, and because altered adeno-
receptors and depressing the indirect pathway sine signaling is involved in a number of acute
38 Part I: Homeostatic Regulators

and chronic neurological disorders, includ- cell death (von Lubitz etal., 1994). Furthermore,
ing brain ischemia, sleep deprivation, epilepsy, homeostatic regulation by adenosine appears
Huntingtons disease, Parkinsons disease, to be different in newborns and adults (Bona
Alzheimers disease, drug addiction, depres- etal., 1995; Mody and MacDonald, 1995; Turner
sion, bipolar disorder, and schizophrenia (Gomes etal.,2004).
et al., 2011), adenosine-based therapies might
be developed. Such adenosine-based thera- SUMMARY
pies include strategies to alter adenosine levels Homeostatic regulation of the levels and
(Boison, 2013; Chu etal., 2013)and/or adenosine actions of ATP and adenosine in the brain
receptor signaling (Fishman etal., 2012; Gomes is complex. There are normal f luctuations
etal., 2011; Sachdeva and Gupta,2013). (noise), and there are increased levels and
With respect to the development of phar- actions of purines (signals) as occurs during
macological inhibitors aimed at augmenting neuronal stimulation, depolarization, meta-
the effect of endogenous adenosine, ADK has bolic insults, and pathological states. Some
received considerable attention (Boison, 2013; 25 years ago, adenosine was described as act-
Boison et al., 2002). However, ADK inhibitors ing like a retaliatory metabolite because
have been linked to increased risks of liver its largely inhibitory actions (certainly those
toxicity (Boison etal., 2002)and brain hemor- mediated through adenosine A1 receptors)
rhages (McGaraughty et al., 2005). Therefore, help to provide negative feedback (retali-
there has been a growing effort to manipulate ation) against initial brain stimuli such as
ADK and adenosine levels site and event specif- increases in energy demand and decreases in
ically by transplanting stem cells with deletions energy supplies. However, understanding the
of ADK and the use of gene therapy vectors homeostatic regulation of purine levels and
to downregulate ADK expression (Shen et al., actions is complicated because of the many
2011; Theofilas et al., 2011). Another approach factors that regulate the intracellular and
might be to alter extracellular adenosine levels extracellular levels of adenosine including
by manipulating ENTs. Indeed, blocking ENTs the transcriptional, translational and epige-
with, for example, dipyridamole decreased netic regulation of adenosines anabolic and
adenosine efflux in neurons but not astrocytes catabolic enzymes, transporters; and receptor
in a cell culture model of ischemia (Parkinson systems. Nevertheless, there remains value in
and Xiong, 2004), and ENT1 knockout mice understanding all of this further because of
have decreased motor incoordination and the need to understand better pathological
hypnosis after ethanol administration (Choi mechanisms as well as the need to create new
etal.,2004). and/or better therapeutics.
With regard to modulating adenosine
receptor signaling, subtype-specific adenosine ACK NOWLEDGMENTS
receptor agonists and antagonists have been Our research activities in this area have been
examined for their use for neurological diseases. supported consistently by grants awarded by the
Neuroprotective effects of adenosine are mostly Canadian Institutes of Health Research (Canada)
attributed to A1Rs that decrease excitotoxic- and the National Institutes of Health (United
ity and reduce cellular metabolic needs. On the States), including R01NS065957, P30GM103329,
other hand, activation of A 2ARs can exacerbate R01MH100972 and R01MH105329.
neuronal injury (Fredholm etal., 2005). Thus A1R References
agonists and A2AR antagonists are neuroprotec- Abbracchio, M.P., S. Ceruti, R. Brambilla, C.
tive (Blum etal., 2002; Delle Donne and Sonsalla, Franceschi, W. Malorni, K.A. Jacobson, D.K.von
1994; MacGregor etal.,1997). Lubitz, and F. Cattabeni. 1997. Modulation of
Finally, for homeostatic regulations by apoptosis by adenosine in the central nervous
adenosine it is important to consider duration- system: A possible role for the A3 receptor.
dependent effects of adenosine receptor manipu- Pathophysiological significance and therapeutic
lation as well as age-dependent effects (Fredholm implications for neurodegenerative disorders.
et al., 2005). Indeed, effect inversion has been Ann NY Acad Sci. 825:1122.
observed where, for example, acute activation of Abraham, W.C. 2008. Metaplasticity: Tuning syn-
A1Rs is neuroprotective while chronic treatment apses and networks for plasticity. Nat Rev
with A1R agonists is associated with neuronal Neurosci.9:387.
Homeostatic Control of Adenosine Levels and Functions in the Brain 39

Ambrosio, A.F., J.O. Malva, A.P. Carvalho, and C.M. Brown, R.E., R. Basheer, J.T. McKenna, R.E. Strecker,
Carvalho. 1997. Inhibition of N-P/Qand other and R.W. McCarley. 2012. Control of sleep and
types of Ca2+ channels in rat hippocampal nerve wakefulness. Physiol Rev. 92:10871187.
terminals by the adenosine A1 receptor. Eur J Carman, A.J., J.H. Mills, A. Krenz, D. G.Kim, and
Pharmacol. 340:301310. M.S. Bynoe. 2011. Adenosine receptor signaling
Arias-Carrion, O., M. Stamelou, E. Murillo-Rodriguez, modulates permeability of the blood-brain bar-
M. Menendez-Gonzalez, and E. Poppel. 2010. rier. J Neurosci. 31:1327213280.
Dopaminergic reward system:Ashort integrative Carriba, P., O. Ortiz, K. Patkar, Z. Justinova, J. Stroik,
review. Int Arch Med.3:24. A. Themann, C. Muller, A.S. Woods, B.T. Hope,
Atkinson, D.E., and G.M. Walton. 1967. Adenosine F. Ciruela, V. Casado, E.I. Canela, C. Lluis, S.R.
triphosphate conservation in metabolic regu- Goldberg, R. Moratalla, R. Franco, and S. Ferre.
lation: Rat liver citrate cleavage enzyme. J Biol 2007. Striatal adenosine A2A and cannabinoid
Chem. 242:32393241. CB1 receptors form functional heteromeric com-
Baldwin, S.A., S.Y. Yao, R.J. Hyde, A.M. Ng, S. plexes that mediate the motor effects of cannabi-
Foppolo, K. Barnes, M.W. Ritzel, C.E. Cass, and noids. Neuropsychopharmacology 32:22492259.
J.D. Young. 2005. Functional characterization of Choi, D.S., M.G. Cascini, W. Mailliard, H. Young,
novel human and mouse equilibrative nucleo- P. Paredes, T. McMahon, I. Diamond, A. Bonci,
side transporters (hENT3 and mENT3) located and R.O. Messing. 2004. The type 1 equilibrative
in intracellular membranes. J. Biol. Chem. nucleoside transporter regulates ethanol intoxi-
280:1588015887. cation and preference. Nat Neurosci. 7:855861.
Basheer, R., A. Bauer, D. Elmenhorst, V. Ramesh, and Chu, S., W. Xiong, D. Zhang, H. Soylu, C. Sun, B.C.
R.W. McCarley. 2007. Sleep deprivation upregu- Albensi, and F.E. Parkinson. 2013. Regulation of
lates A1 adenosine receptors in the rat basal fore- adenosine levels during cerebral ischemia. Acta
brain. Neuroreport 18:18951899. Pharmacol Sin. 34:6066.
Benarroch, E.E. 2008. Adenosine and its recep- Chung, H.J., W.P. Ge, X. Qian, O. Wiser, Y.N. Jan,
tors:Multiple modulatory functions and poten- and L.Y. Jan. 2009. G protein-activated inwardly
tial therapeutic targets for neurologic disease. rectifying potassium channels mediate depoten-
Neurology 70:231236. tiation of long-term potentiation. Proc Natl Acad
Beninger, R.J. 1983. The role of dopamine in Sci USA 106:635640.
locomotor activity and learning. Brain Res. Ciruela, F., V. Casado, R.J. Rodrigues, R. Lujan,
287:173196. J. Burgueno, M. Canals, J. Borycz, N. Rebola,
Blum, D., D. Gall, M.C. Galas, P. dAlcantara, K. S.R. Goldberg, J. Mallol, A. Cortes, E.I. Canela,
Bantubungi, and S.N. Schiffmann. 2002. The J.F. Lopez-Gimenez, G. Milligan, C. Lluis, R.A.
adenosine A1 receptor agonist adenosine amine Cunha, S. Ferre, and R. Franco. 2006. Presynaptic
congener exerts a neuroprotective effect against control of striatal glutamatergic neurotransmis-
the development of striatal lesions and motor sion by adenosine A1-A2A receptor heteromers.
impairments in the 3-nitropropionic acid model J Neurosci. 26:20802087.
of neurotoxicity. J Neurosci. 22:91229133. Ciruela, F., M. Escriche, J. Burgueno, E. Angulo, V.
Boison, D. 2013. Adenosine kinase:Exploitation for Casado, M.M. Soloviev, E.I. Canela, J. Mallol,
therapeutic gain. Pharmacol Rev. 65:906943. W.Y. Chan, C. Lluis, R.A. McIlhinney, and R.
Boison, D., J.F. Chen, and B.B. Fredholm. 2010. Franco. 2001. Metabotropic glutamate 1alpha
Adenosine signaling and function in glial cells. and adenosine A1 receptors assemble into func-
Cell Death Differ. 17:10711082. tionally interacting complexes. J Biol Chem.
Boison, D., L. Scheurer, V. Zumsteg, T. Rulicke, P. 276:1834518351.
Litynski, B. Fowler, S. Brandner, and H. Mohler. Ciruela, F., V. Fernandez-Duenas, J. Llorente, D.
2002. Neonatal hepatic steatosis by disruption Borroto-Escuela, M.L. Cuffi, L. Carbonell, S.
of the adenosine kinase gene. Proc Natl Acad Sci Sanchez, L.F. Agnati, K. Fuxe, and C.I. Tasca.
USA 99:69856990. 2012. G protein-coupled receptor oligomeriza-
Bona, E., U. Aden, B.B. Fredholm, and H. Hagberg. tion and brain integration:Focus on adenosiner-
1995. The effect of long term caffeine treatment gic transmission. Brain Res. 1476:8695.
on hypoxic-ischemic brain damage in the neo- Clark, B.D., Z.L. Kurth-Nelson, and E.A. Newman.
nate. Pediatr Res. 38:312318. 2009. Adenosine-evoked hyperpolariza-
Brambilla, D., D. Chapman, and R. Greene. 2005. tion of retinal ganglion cells is mediated by
Adenosine mediation of presynaptic feed- G-protein-coupled inwardly rectifying K+ and
back inhibition of glutamate release. Neuron small conductance Ca2+-activated K+ channel
46:275283. activation. J Neurosci. 29:1123711245.
40 Part I: Homeostatic Regulators

Cristovao-Ferreira, S., G. Navarro, M. Brugarolas, K. Ferre, S., B.B. Fredholm, M. Morelli, P. Popoli,
Perez-Capote, S.H. Vaz, G. Fattorini, F. Conti, C. and K. Fuxe. 1997. Adenosine-dopamine
Lluis, J.A. Ribeiro, P.J. McCormick, V. Casado, receptor-receptor interactions as an integrative
R. Franco, and A.M. Sebastiao. 2013. A1R-A2AR mechanism in the basal ganglia. Trends Neurosci.
heteromers coupled to Gs and G i/0 proteins 20:482487.
modulate GABA transport into astrocytes. Fishman, P., S. Bar-Yehuda, B.T. Liang, and K.A.
Purinergic Signal. 9:433449. Jacobson. 2012. Pharmacological and therapeu-
Cui, X.A., B. Singh, J. Park, and R.S. Gupta. 2009. tic effects of A3 adenosine receptor agonists.
Subcellular localization of adenosine kinase in Drug Discov Today 17:359366.
mammalian cells: The long isoform of AdK is Fredholm, B.B., A.P. IJzerman, K.A. Jacobson, K.N.
localized in the nucleus. Biochem Biophys Res Klotz, and J. Linden. 2001. International Union
Commun. 388:4650. of Pharmacology. XXV. Nomenclature and clas-
Cunha, R.A. 2001. Adenosine as a neuromodulator sification of adenosine receptors. Pharmacol Rev.
and as a homeostatic regulator in the nervous 53:527552.
system:Different roles, different sources and dif- Fredholm, B.B., A.P. IJzerman, K.A. Jacobson, J.
ferent receptors. Neurochem Int. 38:107125. Linden, and C.E. Muller. 2011. International
Cunha, R.A. 2005. Neuroprotection by adenosine Union of Basic and Clinical Pharmacology.
in the brain: From A(1) receptor activation to LXXXI. Nomenclature and classification of
A (2A) receptor blockade. Purinergic Signal. adenosine receptorsan update. Pharmacol Rev.
1:111134. 63:134.
Darby, M., J.B. Kuzmiski, W. Panenka, D. Feighan, Fredholm, B.B., J.F. Chen, R.A. Cunha, P.
and B.A. MacVicar. 2003. ATP released from Svenningsson, and J.M. Vaugeois. 2005.
astrocytes during swelling activates chloride Adenosine and brain function. Int Rev Neurobiol.
channels. J Neurophysiol. 89:18701877. 63:191270.
de Mendonca, A., A.M. Sebastiao, and J.A. Ribeiro. Fuxe, K., D. Marcellino, D.O. Borroto-Escuela, M.
1995. Inhibition of NMDA receptor-mediated Guescini, V. Fernandez-Duenas, S. Tanganelli,
currents in isolated rat hippocampal neurones by A. Rivera, F. Ciruela, and L.F. Agnati. 2010.
adenosine A1 receptor activation. Neuroreport Adenosine-dopamine interactions in the patho-
6:10971100. physiology and treatment of CNS disorders. CNS
Delaney, S.M., and J.D. Geiger. 1996. Brain regional Neurosci Ther. 16:e18e42.
levels of adenosine and adenosine nucleotides in Geiger, J.D., and D.M. Fyda. 1991. Adenosine trans-
rats killed by high-energy focused microwave port in nervous system tissues. In Adenosine in the
irradiation. J Neurosci Methods 64:151156. nervous system. T. Stone, ed. London:Academic
Delicado, E.G., T. Casillas, R.P. Sen, and M.T. Press,123.
Miras-Portugal. 1994. Evidence that adenine Gomes, C.V., M.P. Kaster, A.R. Tome, P.M.
nucleotides modulate nucleoside-transporter Agostinho, and R.A. Cunha. 2011. Adenosine
function: Characterization of uridine transport receptors and brain diseases: Neuroprotection
in chromaffin cells and plasma membrane vesi- and neurodegeneration. Biochim Biophys Acta.
cles. Eur J Biochem. 225:355362. 1808:13801399.
Delle Donne, K.T., and P.K. Sonsalla. 1994. Protection Gundlfinger, A., J. Bischofberger, F.W. Johenning,
against methamphetamine-induced neurotoxic- M. Torvinen, D. Schmitz, and J. Breustedt. 2007.
ity to neostriatal dopaminergic neurons by ade- Adenosine modulates transmission at the hip-
nosine receptor activation. J Pharmacol Exp Ther. pocampal mossy fibre synapse via direct inhibi-
271:13201326. tion of presynaptic calcium channels. J Physiol.
Dias, R.B., J.A. Ribeiro, and A.M. Sebastiao. 2012. 582:263277.
Enhancement of AMPA currents and GluR1 Hakansson, K., S. Galdi, J. Hendrick, G. Snyder, P.
membrane expression through PKA-coupled Greengard, and G. Fisone. 2006. Regulation of
adenosine A(2A) receptors. Hippocampus phosphorylation of the GluR1 AMPA recep-
22:276291. tor by dopamine D2 receptors. J Neurochem.
Dias, R.B., D.M. Rombo, J.A. Ribeiro, J.M. Henley, 96:482488.
and A.M. Sebastiao. 2013. Adenosine:Setting the Hartwick, A.T., M.R. Lalonde, S. Barnes, and W.H.
stage for plasticity. Trends Neurosci. 36:248257. Baldridge. 2004. Adenosine A1-receptor modu-
Dunwiddie, T.V., and S.A. Masino. 2001. The role lation of glutamate-induced calcium influx in rat
and regulation of adenosine in the central ner- retinal ganglion cells. Invest Ophthalmol Vis Sci.
vous system. Annu Rev Neurosci. 24:3155. 45:37403748.
Homeostatic Control of Adenosine Levels and Functions in the Brain 41

Hertz, L., and H. Matz. 1989. Inhibition of adenos- involves cooperation among pannexin hemi-
ine deaminase activity reveals an intense active channels, adenosine receptors, and KATP chan-
transport of adenosine into neurons in primary nels. J Neurosci. 30:38863895.
cultures. Neurochem Res. 14:755760. King, A.E., M.A. Ackley, C.E. Cass, J.D. Young,
Higley, M.J., and B.L. Sabatini. 2010. Competitive and S.A. Baldwin. 2006. Nucleoside transport-
regulation of synaptic Ca2+ influx by D2 ers:From scavengers to novel therapeutic targets.
dopamine and A2A adenosine receptors. Nat Trends Pharmacol Sci. 27:416425.
Neurosci. 13:958966. Klaasse, E.C., A.P. Ijzerman, W.J.de Grip, and M.W.
Hong, Z.Y., Z.L. Huang, W.M. Qu, N. Eguchi, Y. Beukers. 2008. Internalization and desensitiza-
Urade, and O. Hayaishi. 2005. An adenosine tion of adenosine receptors. Purinergic Signal.
A receptor agonist induces sleep by increas- 4:2137.
ing GABA release in the tuberomammillary Kowaluk, E.A., and M.F. Jarvis. 2000. Therapeutic
nucleus to inhibit histaminergic systems in rats. potential of adenosine kinase inhibitors. Expert
J Neurochem. 92:15421549. Opin Investig Drugs 9:551564.
Hosseinzadeh, H., and T.W. Stone. 1998. Tolbutamide Latini, S., and F. Pedata. 2001. Adenosine in the
blocks postsynaptic but not presynaptic effects central nervous system: Release mechanisms
of adenosine on hippocampal CA1 neurones. J and extracellular concentrations. J Neurochem.
Neural Transm. 105:161172. 79:463484.
Iglesias, R., G. Dahl, F. Qiu, D.C. Spray, and E. Lazarus, M., H.Y. Shen, Y. Cherasse, W.M. Qu, Z.L.
Scemes. 2009. Pannexin 1: The molecular sub- Huang, C.E. Bass, R. Winsky-Sommerer, K.
strate of astrocyte hemichannels. J Neurosci. Semba, B.B. Fredholm, D. Boison, O. Hayaishi,
29:70927097. Y. Urade, and J.F. Chen. 2011. Arousal effect of
Ilie, A., J.V. Raimondo, and C.J. Akerman. 2012. caffeine depends on adenosine A2A receptors in
Adenosine release during seizures attenuates the shell of the nucleus accumbens. J Neurosci.
GABAA receptor-mediated depolarization. J 31:1006710075.
Neurosci. 32:5321532. Lee, M.C., R. Yasuda, and M.D. Ehlers. 2010.
Ismayilova, N., A. Crossman, A. Verkhratsky, Metaplasticity at single glutamatergic synapses.
and J. Brotchie. 2004. Effects of adenosine A1, Neuron 66:859870.
dopamine D1 and metabotropic glutamate MacGregor, D.G., D.I. Graham, and T.W. Stone.
5 receptors-modulating agents on locomo- 1997. The attenuation of kainate-induced neuro-
tion of the reserpinised rats. Eur J Pharmacol. toxicity by chlormethiazole and its enhancement
497:187195. by dizocilpine, muscimol, and adenosine recep-
Kalinchuk, A.V., R.W. McCarley, T. Porkka- tor agonists. Exp Neurol. 148:110123.
Heiskanen, and R. Basheer. 2011. The time course Matos, M., E. Augusto, A.D. Santos-Rodrigues,
of adenosine, nitric oxide (NO) and inducible NO M.A. Schwarzschild, J.F. Chen, R.A. Cunha, and
synthase changes in the brain with sleep loss and P. Agostinho. 2012. Adenosine A2A receptors
their role in the non-rapid eye movement sleep modulate glutamate uptake in cultured astro-
homeostatic cascade. J Neurochem. 116:260272. cytes and gliosomes. Glia 60:702716.
Kamikubo, Y., T. Shimomura, Y. Fujita, T. Tabata, T. McGaraughty, S., M. Cowart, M.F. Jarvis, and R.F.
Kashiyama, T. Sakurai, K. Fukurotani, and M. Berman. 2005. Anticonvulsant and antinocicep-
Kano. 2013. Functional cooperation of metabo- tive actions of novel adenosine kinase inhibitors.
tropic adenosine and glutamate receptors regu- Curr Top Med Chem. 5:4358.
lates postsynaptic plasticity in the cerebellum. J Meghji, P., and A.C. Newby. 1990. Sites of adenosine
Neurosci. 33:1866118671. formation, action and inactivation in the brain.
Kang, J., N. Kang, D. Lovatt, A. Torres, Z. Zhao, J. Neurochem Int. 16:227232.
Lin, and M. Nedergaard. 2008. Connexin 43 Mills, J.H., L. Alabanza, B.B. Weksler, P.O. Couraud,
hemichannels are permeable to ATP. J Neurosci. I.A. Romero, and M.S. Bynoe. 2011. Human brain
28:47024711. endothelial cells are responsive to adenosine
Karcz-Kubicha, M., D. Quarta, B.T. Hope, K. receptor activation. Purinergic Signal. 7:265273.
Antoniou, C.E. Muller, M. Morales, C.W. Mody, I., and J.F. MacDonald. 1995. NMDA
Schindler, S.R. Goldberg, and S. Ferre. 2003. receptor-dependent excitotoxicity: The role of
Enabling role of adenosine A1 receptors in ade- intracellular Ca2+ release. Trends Pharmacol Sci.
nosine A2A receptor-mediated striatal expres- 16:356359.
sion of c-fos. Eur J Neurosci. 18:296302. Mundell, S., and E. Kelly. 2011. Adenosine receptor
Kawamura, M., Jr., D.N. Ruskin, and S.A. Masino. desensitization and trafficking. Biochim Biophys
2010. Metabolic autocrine regulation of neurons Acta 1808:13191328.
42 Part I: Homeostatic Regulators

Newby, A.C., Y. Worku, P. Meghji, M. Nakazawa, and Prediger, R.D., and R.N. Takahashi. 2005.
A.C. Skladanowski. 1990. Adenosine:Aretalia- Modulation of short-term social memory in rats
tory metabolite or not? Physiology 5:6770. by adenosine A1 and A(2A) receptors. Neurosci
Nishi, A., F. Liu, S. Matsuyama, M. Hamada, H. Lett. 376:160165.
Higashi, A.C. Nairn, and P. Greengard. 2003. Queenan, B.N., K.J. Lee, and D.T. Pak. 2012.
Metabotropic mGlu5 receptors regulate adenos- Wherefore art thou, homeo(stasis)? Functional
ine A2A receptor signaling. Proc Natl Acad Sci diversity in homeostatic synaptic plasticity.
USA 100:13221327. Neural Plast. 2012:718203.
ORegan, M. 2005. Adenosine and the regulation of Rebola, N., R. Lujan, R.A. Cunha, and C. Mulle.
cerebral blood flow. Neurol Res. 27:175181. 2008. Adenosine A2A receptors are essential
Olah, M.E., and G.L. Stiles. 1995. Adenosine recep- for long-term potentiation of NMDA-EPSCs
tor subtypes: Characterization and therapeu- at hippocampal mossy fiber synapses. Neuron
tic regulation. Annu Rev Pharmacol Toxicol. 57:121134.
35:581606. Ribeiro, J.A., and A.M. Sebastiao. 2010. Modulation
Orford, M.R., and E.D. Saggerson. 1996. A low-Km and metamodulation of synapses by adenosine.
5-nucleotidase from rat brain cytosolic frac- Acta Physiol (Oxf). 199:161169.
tion: Purification, kinetic properties, and Rolfe, D.F., and G.C. Brown. 1997. Cellular energy
description of regulation by a novel factor that utilization and molecular origin of standard met-
increases sensitivity to inhibition by ATP and abolic rate in mammals. Physiol Rev. 77:731758.
ADP. J Neurochem. 67:795804. Sachdeva, S., and M. Gupta. 2013. Adenosine and its
Pankratov, Y., U. Lalo, A. Verkhratsky, and R.A. receptors as therapeutic targets: An overview.
North. 2007. Quantal release of ATP in mouse Saudi Pharm J. 21:245253.
cortex. J Gen Physiol. 129:257265. Salamone, J.D., and M. Correa. 2002. Motivational
Parkinson, F.E., K.A. Rudolphi, and B.B. Fredholm. views of reinforcement:Implications for under-
1994. Propentofylline: A nucleoside transport standing the behavioral functions of nucleus
inhibitor with neuroprotective effects in cerebral accumbens dopamine. Behav Brain Res. 137:325.
ischemia. Gen Pharm. 25:10531058. Schiffmann, S.N., G. Fisone, R. Moresco, R.A.
Parkinson, F.E., and W. Xiong. 2004. Stimulus- Cunha, and S. Ferre. 2007. Adenosine A2A
and cell-type-specific release of purines in cul- receptors and basal ganglia physiology. Prog
tured rat forebrain astrocytes and neurons. J Neurobiol. 83:277292.
Neurochem. 88:13051312. Schwindinger, W.F., L.J. Mihalcik, K.E. Giger, K.S.
Pascual, O., K.B. Casper, C. Kubera, J. Zhang, R. Betz, A.M. Stauffer, J. Linden, D. Herve, and J.D.
Revilla-Sanchez, J.Y. Sul, H. Takano, S.J. Moss, Robishaw. 2010. Adenosine A2A receptor signal-
K. McCarthy, and P.G. Haydon. 2005. Astrocytic ing and golf assembly show a specific require-
purinergic signaling coordinates synaptic net- ment for the gamma7 subtype in the striatum. J
works. Science 310:113116. Biol Chem. 285:2978729796.
Pazzagli, M., F. Pedata, and G. Pepeu. 1993. Effect Sebastio, Ana M., S. Cristovao-Ferreira, and J.A.
of K+ depolarization, tetrodotoxin, and NMDA Ribeiro. 2011. Downstream pathways of adenos-
receptor inhibition on extracellular adenosine ine. In Adenosine: A key link between metabo-
levels in rat striatum. Eur J Pharmacol. 234:6165. lism and brain activity. S.M.D. Boison, ed.
Pelligrino, D.A., F. Vetri, and H.L. Xu. 2011. NewYork:Springer, 131156.
Purinergic mechanisms in gliovascular cou- Sebastiao, A.M., and J.A. Ribeiro. 2009a. Adenosine
pling. Semin Cell Dev Biol. 22:229236. receptors and the central nervous system. Handb
Pelligrino, D.A., H.L. Xu, and F. Vetri. 2010. Caffeine Exp Pharmacol:471534.
and the control of cerebral hemodynamics. J Sebastiao, A.M., and J.A. Ribeiro. 2009b. Tuning and
Alzheimers Dis. 20 (Suppl 1):S51S62. fine-tuning of synapses with adenosine. Curr
Peng, L., R. Huang, A.C. Yu, K.Y. Fung, M.P. Neuropharmacol. 7:180194.
Rathbone, and L. Hertz. 2005. Nucleoside trans- Shen, H.Y., T.A. Lusardi, R.L. Williams-Karnesky,
porter expression and function in cultured J.Q. Lan, D.J. Poulsen, and D. Boison. 2011.
mouse astrocytes. Glia 52:2535. Adenosine kinase determines the degree of brain
Popoli, P., L. Gimenez-Llort, A. Pezzola, R. Reggio, E. injury after ischemic stroke in mice. J Cereb
Martinez, K. Fuxe, and S. Ferre. 1996. Adenosine Blood Flow Metab. 31:16481659.
A1 receptor blockade selectively potentiates Shepel, P.N., D. Ramonet, P. Stevens, and J.D.
the motor effects induced by dopamine D1 Geiger. 2005. Purine level regulation dur-
receptor stimulation in rodents. Neurosci Lett. ing energy depletion associated with graded
218:209213.
Homeostatic Control of Adenosine Levels and Functions in the Brain 43

excitatory stimulation in brain. Neurol Res. von Lubitz, D.K., R.C. Lin, N. Melman, X.D. Ji,
27:139148. M.F. Carter, and K.A. Jacobson. 1994. Chronic
Stromberg, I., P. Popoli, C.E. Muller, S. Ferre, and K. administration of selective adenosine A1 recep-
Fuxe. 2000. Electrophysiological and behavioural tor agonist or antagonist in cerebral ischemia.
evidence for an antagonistic modulatory role of Eur J Pharmacol. 256:161167.
adenosine A2A receptors in dopamine D2 recep- von Lubitz, D.K., W. Ye, J. McClellan, and R.C. Lin.
tor regulation in the rat dopamine-denervated 1999. Stimulation of adenosine A3 receptors in
striatum. Eur J Neurosci. 12:40334037. cerebral ischemia: Neuronal death, recovery, or
Studer, F.E., D.E. Fedele, A. Marowsky, C. Schwerdel, both? Ann NY Acad Sci. 890:93106.
K. Wernli, K. Vogt, J.M. Fritschy, and D. Boison. Wall, M.J., and N. Dale. 2013. Neuronal trans-
2006. Shift of adenosine kinase expression from porter and astrocytic ATP exocytosis underlie
neurons to astrocytes during postnatal develop- activity-dependent adenosine release in the hip-
ment suggests dual functionality of the enzyme. pocampus. J Physiol. 591:38533871.
Neuroscience 142:125137. Wei, C.J., P. Singer, J. Coelho, D. Boison, J. Feldon,
Suzuki, T., K. Namba, H. Tsuga, and H. Nakata. B.K. Yee, and J.F. Chen. 2011. Selective inacti-
2006. Regulation of pharmacology by vation of adenosine A(2A) receptors in striatal
hetero-oligomerization between A1 adenosine neurons enhances working memory and reversal
receptor and P2Y2 receptor. Biochem Biophys Res learning. Learn Mem. 18:459474.
Commun. 351:559565. White, H., and B. Venkatesh. 2011. Clinical
Tebano, M.T., A. Martire, N. Rebola, R. Pepponi, M.R. review: Ketones and brain injury. Crit Care
Domenici, M.C. Gro, M.A. Schwarzschild, J.F. 15:219.
Chen, R.A. Cunha, and P. Popoli. 2005. Adenosine Woods, H.A., and J.K. Wilson. 2013. An informa-
A2A receptors and metabotropic glutamate 5 tion hypothesis for the evolution of homeostasis.
receptors are co-localized and functionally inter- Trends Ecol Evol. 28:283289.
act in the hippocampus: A possible key mecha- Yang, C., S. Franciosi, and R.E. Brown. 2013.
nism in the modulation of N-methyl-D-aspartate Adenosine inhibits the excitatory synap-
effects. J Neurochem. 95:1188200. tic inputs to basal forebrain cholinergic,
Theofilas, P., S. Brar, K.A. Stewart, H.Y. Shen, GABAergic, and parvalbumin neurons in mice.
U.S. Sandau, D. Poulsen, and D. Boison. 2011. Front Neurol.4:77.
Adenosine kinase as a target for therapeutic anti- Yao, L., M.P. Arolfo, D.P. Dohrman, Z. Jiang, P.
sense strategies in epilepsy. Epilepsia 52:589601. Fan, S. Fuchs, P.H. Janak, A.S. Gordon, and
Thompson, S.M., H.L. Haas, and B.H. Gahwiler. I. Diamond. 2002. Betagamma dimers medi-
1992. Comparison of the actions of adenosine at ate synergy of dopamine D2 and adenosine A2
pre- and postsynaptic receptors in the rat hippo- receptor-stimulated PKA signaling and regulate
campus in vitro. J Physiol. 451:347363. ethanol consumption. Cell 109:733743.
Turner, C.P., M.R. Blackburn, and S.A. Rivkees. Yoshioka, K., O. Saitoh, and H. Nakata. 2001.
2004. A1 adenosine receptors mediate Heteromeric association creates a P2Y-like
hypoglycemia-induced neuronal injury. J Mol adenosine receptor. Proc Natl Acad Sci USA
Endocrinol. 32:129144. 98:76177622.
Urade, Y., and O. Hayaishi. 2011. Prostaglandin D2 and Zhang, J.P., Q. Xu, X.S. Yuan, Y. Cherasse, S.N.
sleep/wake regulation. Sleep Med Rev. 15:411418. Schiffmann, A. de Kerchove dExaerde, W.M.
Van Wylen, D.G., T.S. Park, R. Rubio, and R.M. Qu, Y. Urade, M. Lazarus, Z.L. Huang, and R.X.
Berne. 1986. Increases in cerebral interstitial Li. 2013. Projections of nucleus accumbens ade-
fluid adenosine concentration during hypoxia, nosine A2A receptor neurons in the mouse brain
local potassium infusion, and ischemia. J Cereb and their implications in mediating sleep-wake
Blood Flow Metab. 6:522528. regulation. Front Neuroanat.7:43.
3
Glutamate Homeostasis asa Regulator
ofNeurotransmitter Recycling
and Synaptic Function
M A R Y E L L E N K E L LY A N D D O U G L A S A . C O U LT E R

INTRODUCTION monoamines, and acetylcholine, among others


To ensure appropriate temporal fidelity and (for review, see Eiden, 2000). The GABA vesicu-
maintenance of maintain proper synaptic trans- lar transporter (VIAAT), like other vesicular
mission, neurotransmitters must be removed transporters, relies on the proton gradient within
rapidly from the leave the synaptic cleft follow- synaptic vesicles to transport GABA against its
ing release, and metabolic intermediates must be concentration gradient. Evidence has emerged
returned to presynaptic terminals for subsequent demonstrating complexity in this process of
regeneration of neurotransmitters to sustain sequestering GABA. VIAAT activity may be
subsequent synaptic function. For the central tightly linked to local GABA synthesis, due to the
nervous system (CNS)neurotransmitters gluta- fact that it may participate in a supramolecular
mate and GABA, these requirements are fulfilled complex including glutamic acid decarboxylase
through partitioned and segregated transporter (GAD), and Ca2+/calmodulin-dependent pro-
and enzyme expression and function in neurons tein kinase II (Jin etal. 2003). This suggests that
and astrocytes. Unlike monamine and acetyl- glutamate, in addition to serving as an excita-
choline uptake, the majority of neurotransmitter tory neurotransmitter in principal neurons, may
transporter expression (and associated trans- be the critical precursor in inhibitory synaptic
mitter uptake) at glutamatergic and GABAergic terminals, regulating intravesicular GABA lev-
synapses is evident on perisynaptic astrocytic els. Two patch clamp recording studies have lent
processes (Danbolt, 2001; Schousboe, 2000). support to this hypothesis (Sepkuty et al. 2002;
Once taken up into astrocytes, GABA and gluta- Mathews and Diamond,2003).
mate are metabolized, and metabolic precursors
for subsequent glutamate and GABA re-synthesis T H E G L U TA M AT E - G L U TA M I N E
are shuttled to neuronal terminals to allow main- CYCLE AS A LOCAL , R APID
tenance of synaptic transmission. ECONOM Y IN SY NAPSES
Specialized machinery exists to synthesize Much of what we know about the various stages
and package neurotransmitter in membrane- of neurotransmitter recycling comes from bio-
delimited packets (synaptic vesicles) for use in chemical studies in tissue homogenates, or stud-
intercellular communication. Since the early cal- ies utilizing sampling of extracellular amino
culations of molar content of cholinergic vesicles acid concentrations in labelled tissue sam-
over 30years ago (Kuffler and Yoshikami, 1975), ples. Although this confers information about
it has been known that vesicular content of neu- global transfer of amino acids through the
rotransmitter is extremely high, approaching the glutamate-glutamine cycle in brain regions in
physical limits for packaging sufficient numbers general, it misses significant information con-
of molecules within the limited lumenal space cerning the local recycling of neurotransmitter in
of a vesicle. Several structurally related proteins synaptic compartments, which are isolated and
have been identified for mediating small mole- remote from cellular metabolism occurring in
cule neurotransmitter vesicle packaging, includ- neuronal somata. Synapses can be quite distant
ing vesicular transporters for glutamate, GABA, in both space and time relative to overall cellular
Glutamate Homeostasis as a Regulator of Neurotransmitter 45
metabolism of amino acids. There are signifi- inhibitory neurons may act as a significant pre-
cant temporal delays imposed by the necessity of cursor for GABA synthesis. Several systems are
transferring metabolic substrates from the cell present within inhibitory synaptic terminals
somata to the synaptic compartment. Synapses that provide GABA used for vesicular packag-
overcome this constraint by conducting the ing by VIAAT. These include proteins that facili-
majority of their neurotransmitter metabolism tate reuptake of GABA and glutamate into the
locally, within the tripartite compartment cre- inhibitory terminal following release (GABA
ated by the perisynaptic, sastrocyte process and transporters such as GAT-1 and the neuronal glu-
the pre- and postsynaptic cellular components. tamate transporter EAAC-1), other transporters
Within this tripartite necessary there is a that take up glutamine (system A[slc38-family]
tightly coupled linkage between glutamate uptake transporters; Chaudry et al. 2002), alanine (asc
in astrocytes and subsequent release and transfer family transporters; Palacin et al. 1998; Helboe
of glutamine to neurons (Uwechue etal., 2012). As et al. 2003; Kanai and Hediger, 2004), and leu-
a primary contributory mechanism to this cou- cine (system L transporters, Segawa etal. 1999),
pled function, both the glutamate transporter-1 all amino acids that are released from astrocytes.
(GLT-1) and the System N glutamine trans- Once in the inhibitory presynaptic terminal,
porter (SN1) are colocalized on the fine astro- these amino acids can all serve as precursors for
cytic processes ensheathing synapses (Boulland local glutamate synthesis by the enzymatic activ-
etal., 2002, Chaudry etal., 1995; Danbolt, 2001). ity of glutaminase, alanine aminotransferase,
Possibly interacting with the precise colocaliza- and branched-chain aminotransferase, respec-
tion of these astrocytic transporters, the milli- tively (Yudkoff etal. 1996; Yudkoff, 1997; Sweatt
second precision in coupling between glutamate etal. 2004). In addition, other substrates may be
uptake into astrocytes and glutamine resupply provided by the tricarboxylic acid (TCA) cycle,
to neurons (Uwechue etal., 2012)may be gener- localized in mitochondria within the synaptic
ated through the interacting stoichiometry of the terminal. Since neurons lack pyruvate carboxy-
various transporters involved. Glutamate uptake lase, an enzyme necessary to synthesize gluta-
via GLT-1 is linked to the cotransport of three mate directly from CO2 (Shank et al. 1985; Yu
sodium ions and a proton and reverse transport et al. 1983), and these cells constantly release
of a potassium ion and so is accompanied by a glutamate and/or GABA as neurotransmitters,
rapid rise in intracellular sodium within astro- they rely exclusively on astrocytes to provide
cytes (Langer and Rose, 2009; Langer etal., 2011; substrates for continued glutamate synthesis.
Uwechue et al., 2012). Glutamine transport out Otherwise, provision of TCA intermediates to
of astrocytes is mediated by System N transport- subserve this function will result in their deple-
ers, which cotransport a sodium ion with glu- tion and the rapid failure of intraterminal mito-
tamie and countertransport a proton, resulting in chondrial metabolism and energy production.
electroneutral release of glutamine (Broer et al., Evidence shows that in a normal brain the major-
2002). Because System N transporters operate in ity of neurotransmitter recycling is accomplished
a near equilibrium, the sodium influx occurring through the astrocytic glutamate-glutamine
during glutamate uptake in astrocytes (with a 3:1 cycle (Laake et al. 1995; Battiglioli and Martin,
stoichiometry) may be sufficient to couple GLT-1 1996; Rae etal.2003).
function to the rapid initiation and amplification
of glutamine efflux via System N transport (with THE ASTROCYTIC
their 1:1 stoichiometry for Na+ and glutamine; G L U TA M AT E - G L U TA M I N E
Broer et al., 2002). This rapid coupling may be CYCLE ASA GABA
enhanced by its occurrence within the fine astro- R ECYCLING MECHANISM
cytic processes investing synapses, which will While interneurons do recycle GABA from
generate a much larger increase in net sodium the synaptic cleft through GABA transporters
concentration during glutamate uptake due to (including GAT-1), the principal source of syn-
the small cytoplasmic compartment in which it thesized GABA packed into synaptic vesicles is
occurs. derived from decarboxylation of glutamate by
glutamic acid decarboxylase (Martin etal. 2000).
SOURCES There are two primary sources for this gluta-
OFPRESY NAPTICGABA mate in interneurons. One is glutamate uptake
In addition to serving as a neurotransmitter via excitatory amino acid carrier-1 transporters
in excitatory neurons, glutamate supplied to (EAAC-1) located in the presynaptic terminals of
46 Part I: Homeostatic Regulators

GABAergic neurons (Arriza et al., 1994; Bjoras In contrast, in similar experimental stud-
etal., 1996; Nakayama etal., 1996; Eskandari etal., ies, excitatory synaptic transmission was found
2000). A second source of glutamate is derived to be much less sensitive to disruption of the
from the astrocytic glutamate-glutaminecycle. glutamate-glutamine cycle. Although several
The glutamate-glutamine cycle begins with studies have found that epileptiform activity
the uptake of synaptically released glutamate by in vitro depends on glutamine supply (Bacci
glutamate transporters in ensheathing astrocytic et al., 2002; Tani et al., 2007, 2010), excitatory
processes. In the forebrain, glutamate is taken synaptic transmission exhibited less evidence
up by astrocytes through the astrocytic specific for a similar reliance. Under conditions where
glutamate transporter-1 (GLT-1) and converted the glutamate-glutamine cycle was blocked by
into glutamine by astrocyte-specific enzyme, application of a glutamine synthetase antago-
glutamine synthetase (Pines et al., 1992; Arriza nist (methionine sulfoximine), no compromise
etal., 1994). Glutamine is then shuttled back to in excitatory synaptic function was evident
presynaptic terminals of neurons through spe- until prolonged periods of synaptic release were
cific glutamine transporters localized in astro- induced, encompassing hours or thousands of
cytes and neurons. Glutamine is released from synaptic stimuli (Kam and Nicoll, 2007; Tani
astrocytes by system N transporters and trans- et al., 2014). So, although inhibitory synaptic
ferred into neurons by system A transporters function appears directly linked to astrocytic
(both members of the slc38 family; Chaudhry derived glutamine, continued excitatory synap-
et al., 2002). Once in the presynaptic terminal, tic transmission appears less dependent on this
glutamine is then reconverted to glutamate by astrocytic-derived glutamate precursor. This
the mitochondrial enzyme, phosphate-activated may be due to higher reserve pools of cytoplas-
glutaminase. Following its resynthesis from glu- mic glutamate being accessible in excitatory
tamine, glutamate is available directly as a neu- relative to inhibitory neurons, buffering the
rotransmitter for packaging in synaptic vesicles immediate effects of loss of glutamate-glutamine
in excitatory terminals or is converted to GABA cycle function for significant periods of activ-
by decarboxylation in inhibitory terminals (the ity. Comparing the distinct effects of disrupting
glutamate-glutamine-GABA cycle). Release of the glutamate-glutamine cycle in inhibitory and
synaptic glutamate or GABA then completes excitatory synapses, it is likely that the predomi-
the cycle (reviewed in Bak etal., 2006; Chaudry nant proximate effect would be to reduce GABA
etal.,2002). release from inhibitory synapses, potentially
Given the primary role of the glutamate- enhancing circuit excitability.
glutamine cycle in taking up and inactivating One notable consequence of the depend-
glutamate, and the linkage between this cycle ence of GABA replenishment on the glutamate-
and the local neurotransmitter economy in syn- glutamine-GABA cycle is that this may serve
apses (discussed earlier), it would be expected as an intimate link, scaling inhibitory synaptic
that pharmacological approaches interfering efficacy to ongoing levels of glutamate release.
with the cycle at any stage would rapidly impact Since appropriate inhibition is necessary to con-
neurotransmitter supply and synaptic function. strain the potential for excessive glutamater-
Several studies have examined the role of the gic excitation, this linkage to the astrocytic
glutamate-glutamine cycle in maintaining inhib- glutamate-glutamine cycle may provide GABA
itory synaptic transmission. In hippocampal and on demand when excitation levels are high and
thalamic inhibitory synapses, there was a marked reduce GABA levels under low activity condi-
dependence of transmitter release on continued tions. As already mentioned, this glutamate
function of the glutamate-glutamine cycle. In release to glutamine resupply linkage occurs
functional studies, it was found that application within milliseconds, enhancing the potential for
of a glutamine synthetase antagonist, glutamate an intimate interrelationship between excitatory
uptake blockers, or glutamine transporter block- activity and supply of metabolites to sustain inhi-
ers all rapidly depleted synaptic release of GABA, bition (Uwechue etal., 2012). Interestingly, many
which could be reversed by a supply of exogenous of the key players in the glutamate-glutamine
glutamine (Fricke etal., 2007; Liang etal., 2006; cycle exhibit functional regulation at both the
Yang and Cox, 2011). This is entirely consistent transcriptional and the posttranslational level
with the astrocytic glutamine-glutamate cycle (e.g., Broer etal., 2004; Nissen-Meyer etal., 2011;
plays a primary role in providing inhibitory syn- Yang etal., 2009), suggesting that differing activ-
apses with substrates for GABA synthesis. ity levels in brain circuits may alter the efficacy of
Glutamate Homeostasis as a Regulator of Neurotransmitter 47

this cycle by influencing expression and function sclerosis in TLE. This supports a causal/con-
of these key players. tributory linkage between glutamine synthetase
downregulation and the circuit hyperexcitabil-
T H E G L U TA M AT E - G L U TA M I N E ity generating seizures, the hallmark symptom
CYCLE IN EPILEPSY ofTLE.
Many central nervous system disease states,
including epilepsy, Parkinsons disease, ASTROGLIOSIS EFFECTS ON
Alzheimers disease, stroke, and traumatic brain SY NAPTIC TR ANSMISSION
injury, are associated with the development of A N D C I R C U I T E X C I TA B I L I T Y
a series of astrocytic alterations, collectively Astrogliosis is a pathological hallmark of the
termed reactive astrocytosis. The disease in hippocampus in TLE patients. As discussed
which reactive astrocytosis has arguably received previously, the development of astrogliosis is
the most experimental attention is temporal lobe accompanied by a number of important changes
epilepsy (TLE). The pathological hallmark of in astrocyte structure and function, including
TLE is mesial temporal sclerosis, defined as seg- a marked downregulation in expression of glu-
mental neuronal loss and reactive astrocytosis in tamine synthetase. Recent experiments have
the hippocampus and associated temporal lobe explored the potential role of astrogliosis in excit-
structures. ability defects that underly seizure predisposi-
In addition to proliferation of astrocytes tion in TLE. The role of astroglial reactivityin
and anatomic changes in cellular shape and the etiology of epilepsy is poorly understood.
size, there are numerous functional and bio- This is in large part because of the many poten-
chemical changes in astrocytes accompanying tially contributory changes in diverse processes
the development of reactive astrogliosis. These evident following an epileptogenic injury, which
include increased expression of glial fibrillary complicate the interpretation of results. To iso-
acidic protein (GFAP) and vimentin, downreg- late the role that reactive astrogliosis may play
ulation in both Kv4.1 and aquaporin-4 expres- in generating hippocampal excitability defects,
sion, reduced gap junction coupling between Ortinski etal. (2010) induced reactive astrocyto-
astrocytes, and decreased expression of GLT-1 sis in isolation using a cell-specific viral strategy
and glutamine synthetase (Eid etal., 2004; van and then examined the effects of gliosis on circuit
der Hel et al., 2005; reviewed in Wetherington excitability.
et al., 2008). Associated with the presence of Experimental astrogliosis induced by the
astrogliosis and loss of glutamine synthetase in viral strategy employed by Ortinski and col-
the hippocampus of patients with TLE, there leagues (2010) had several similarities to reac-
are abnormally high concentrations of extracel- tivity seen in vivo. Astrocytes overexpressing
lular glutamate (Cavus etal., 2005; During and Green Fluorescent Protein following a high titer
Spencer,1993). viral infection exhibited hypertrophy, increased
In patients with TLE, there is a juxtaposition vimentin, and GFAP expression, as well as a
between pathological hyperexcitability (seizure downregulation in glutamine synthetase expres-
predisposition), astrogliosis, and accompany- sion, all of which were viral-titer dependent.
ing loss of glutamine synthetase expression. This Importantly, neighboring pyramidal neurons
raises the related questions:Is loss of glutamine and interneurons showed no alterations in their
synthetase (and the concomitant damaged func- anatomy and intrinsic properties. In hippocam-
tion of the glutamate-glutamine cycle) a cause pal regions with reactive astrocytes, GABAergic
or a consequence of epilepsy? Do these changes inhibition, but not glutamatergic excitation, was
contribute to seizure susceptibility, or are they significantly compromised. To assess effects
merely a reflection of the severe damage seen of gliosis on circuit excitability in area CA1 of
in the hippocampi of patients with TLE? To the hippocampus, the response to activation of
address these questions, Eid and colleagues direct cortical inputs to the distal dendrites of
(2008) continuously infused the glutamine syn- CA1 neurons was compared between controls
thetase antagonist methionine sulfoximine into and astrogliotic hippocampal slices. This path-
the hippocampus of rats for 28 days and moni- way has been previously shown to be extensively
tored animals for epilepsy development (see also hyperactive in epileptic animals (Ang etal., 2006;
Wang etal., 2009). They found that methionine Denslow etal., 2001; Wozny etal., 2005)and to be
sulfoximinetreated animals developed epilepsy powerfully regulated by local circuit inhibition
and neuropathology similar to mesial temporal (Ang etal., 2005; Empson and Heinemann, 1995;
48 Part I: Homeostatic Regulators

Soltesz, 1995). In regions of extensive gliosis, there Arriza, J.L., W.A. Fairman, J.L. Wadiche, G.H.
was significant hyperactivation of CA1 by direct Murdoch, M.P. Kavanaugh, and S.G. Amara.
cortical inputs, and this was reversed by supply 1994. Functional comparisons of three glutamate
of exogenous glutamine (Ortinski et al., 2010). transporter subtypes cloned from human motor
Finally, all of these astrogliosis-mediated effects cortex. J Neurosci. 14:55595569.
on inhibitory synaptic function could be mim- Bacci, A., G. Sancini, C. Verderio, S. Armano, E.
icked by blockers of the glutamate-glutamine Pravettoni, R. Feace, S. Franceschetti, and M.
cycle, and these alterations in gliotic hippocampi Matteoli. 2002. Block of glutamate-glutamine
were occluded by these same blockers and cycle between astrocytes and neurons inhib-
reversed by supply of exogenous glutamine, all its epileptiform activity in hippocampus.
supporting a role for gliosis-induced glutamine J Neurophysiol. 88:23022310.
synthetase downregulation in the circuit excit- Bak, L.K., A. Schousboe, and H.S. Waagepetersen.
ability defects underlying epilepsy. 2006. The glutamate/GABA-glutamine cycle:
Aspects of transport, neurotransmitter homeo-
stasis and ammonia transfer. J Neurochem.
CONCLUSIONS
98:641653.
One primary mechanism contributing to regu-
Battiglioli, G., and D.L. Martin. 1996. Glutamine
lation of glutamate homeostasis in the brain
stimulates gamma-aminobutyric acid synthesis
is the glutamate-glutamine cycle. This cycle
in synaptosomes but other putative astrocyte-to-
involves partitioned expression of various trans-
neuron shuttle substances do not. Neurosci Lett.
porters and enzymes in perisynaptic astro-
209:129133.
cytic processes and presynaptic terminals of Bjoras, M., O. Gjesdal, J.D. Erickson, R. Torp,
excitatory and inhibitory neurons. It is critical L.M. Levy, O.P. Ottersen, M. Degree, J.
in regulating pathological extracellular accu- Storm-Mathisen, E. Seeberg, and N.C. Danbolt.
mulation of glutamate, termination of synaptic 1996. Cloning and expression of a neuronal rat
responses, and in resupply of neurotransmitter brain glutamate transporter. Brain Res Mol Brain
in both excitatory and inhibitory neurons. This Res. 36(1):163168.
glutamate-glutamine cycle is not only critical Boulland, J.L., K.K. Osen, L.M. Levy, N.C. Danbolt,
in normal functions of synapses. Dysfunction R.H. Edwards, J. Storm-Mathiesen, and F.A.
in this cycle may contribute to the etiology of Chaudry. 2002. Cell-specific expression of the
many neurological disease states, including epi- glutamine transporter SN1 suggests differences
lepsy. Epilepsy is associated with the develop- in dependence on the glutamine cycle. Eur J
ment of significant gliosis, which, among other Neurosci. 15:16151631.
changes, results in a significant downregula- Broer, A., A. Albers, I. Setiawan, R.H. Edwards,
tion in expression of the keystone astrocytic F.A. Chaudry, F. Lang, C.A. Wagner, and S.
enzyme in the glutamate-glutamine cycle, glu- Broer. 2002. Regulation of the glutamine trans-
tamine synthetase. Experimental induction of porter SN1 by extracellular pH and intracellular
reduced glutamine synthetase function results sodium ions. J Physiol. 539:314.
in epilepsy and triggers hippocampal circuit Broer, A., J.W. Deitmer, and S. Broer. 2004. Astroglial
hyperexcitability and disinhibition. These data glutamine transport by system N is upregulated
not only provide mechanistic insight into cir- by glutamate. Glia 48:298310.
cuit dysfunction underlying epilepsy and other Cavus, I., W.S. Kasoff, M.P. Cassaday, R. Jacob,
disorders associated with gliosis but also sug- R. Gueorguieva, R.S. Sherwin, J.H. Krystal,
gest novel, metabolic strategies for the develop- D.D. Spencer, and W.M. Abu-Saab. 2005.
ment of therapeutic agents to better treat such Extracellular metabolites in the cortex and
diseases. hippocampus of epileptic patients. Ann Neurol.
57:226235.
References Chaudry, F.A., K.P. Lehre, M. van Lookeren
Ang, C.W., G.C. Carlson, and D.A. Coulter. 2005. Campagne, O.P. Ottersen, N.P. Danbolt, and J.
Hippocampal CA1 circuitry dynamically gates Storm-Mathisen. 1995. Glutamate transporters
direct cortical inputs preferentially at theta fre- in glial plasma membranes:Highly differentiated
quencies. J Neurosci. 25:95679580. localizations revealed by quantitative ultrastruc-
Ang, C.W., G.C. Carlson, and D.A. Coulter. 2006. tural immunohistochemistry. Neuron 15:711720.
Massive and specific dysregulation of direct cor- Chaudry, F.A., R.J. Reimer, and R.H. Edwards. 2002.
tical inputs to the hippocampus in temporal lobe The glutamine commute: Take the N line and
epilepsy. J Neurosci. 26:1185011856. transfer to the A. J Cell Biol. 157:349355.
Glutamate Homeostasis as a Regulator of Neurotransmitter 49

Danbolt, N.C. 2001. Glutamate uptake. Prog SLC1: Molecular, physiological and pharmaco-
Neurobiol. 65:1105. logical aspects. Pflugers Arch. 447(5):469479.
Denslow, M.J., T. Eid, F. Du, R. Schwarcz, E.W. Kuffler, S.W., and D. Yoshikami. 1975. The dis-
Lothman, and O. Steward. 2001. Disruption tribution of acetylcholine sensitivity at the
of inhibition in area CA1 of the hippocam- post-synaptic membrane of vertebrate skeletal
pus in a rat model of temporal lobe epilepsy. J twitch muscles: Iontophoretic mapping in the
Neurophysiol. 86:22312245. micron range. J Physiol. 244(3):703730.
During, M.J., and D.D. Spencer. 1993. Extracellular Laake, J.H., T.A. Slyngstad, F.M. Haug, and O.P.
hippocampal glutamate and spontaneous sei- Ottersen. 1995. Glutamine from glial cells is
zure in the conscious human brain. Lancet essential for the maintenance of the nerve ter-
341:16071610. minal pool of glutamate:Immunogold evidence
Eid, T., A. Ghosh, Y. Wang, H. Beckstrom, H.P. Zaveri, from hippocampal slice cultures. J Neurochem.
T.S. Lee, J.C. Lai, G.H. Malthankkar-Phatak, and 65:871881.
N.C.de Lanerolle. 2008. Recurrent seizures and Langer, J., and C.R. Rose. 2009. Synaptically induced
brain pathology after inhibition of glutamine sodium signals in hippocampal astrocytes in
synthetase in the hippocampus in rats. Brain situ. J Physiol. 587:58595877.
131:20612070. Langer, J., J. Stephan, M. Theis, and C.R. Rose. 2011.
Eid, T., M.J. Thomas, D.D. Spencer, E. Runden-Pran, Gap junctions mediate intercellular spread of
J.C. Lai, G.V. Malthankar, J.H. Kim, N.C. sodium between hippocampal astrocytes in situ.
Danbolt, O.P. Ottersen, and N.C. De Lanerolle. Glia 60:239252.
2004. Loss of glutamine synthetase in the human Liang, S.-L., G.C. Carlson, and D.A. Coulter. 2006.
epileptogenic hippocampus: Possible mecha- Dynamic regulation of synaptic GABA release by
nism for raised extracellular glutamate in mesial the glutamate-glutamine cycle in hippocampal
temporal lobe epilepsy. Lancet 363:2837. area CA1. J Neurosci. 26(33):85378548.
Eiden, L.E. 2000. The vesicular neurotransmitter Martin, D.L., H. Liu, S.B. Martin, and S.J. Wu. 2000.
transporters: Current perspectives and future Structural features and regulatory properties of
prospects. FASEB J. 14(15):23962400. the brain glutamate decarboxylases. Neurochem
Empson, R.M., and U. Heinemann. 1995. The per- Int. 37(23):111119.
forant path projection to the hippocampal area Mathews, G., and J. Diamond. 2003. Neuronal glu-
CA1 in the rat hippocampal-entorhinal cortex tamate uptake contributes to GABA synthesis
combined slice. J Physiol. 484:707720. and inhibitory synaptic strength. J Neurosci.
Eskandari, S., M. Kreman, M.P. Kavanaugh, E.M. 23:20402048.
Wright, and G.A. Zampighi. 2000 Pentameric Nakayama T., H. Kawakami, K. Tanaka, and S.
assembly of a neuronal glutamate transporter. Nakamura. 1996. Expression of three glutamate
Proc Natl Acad Sci USA 97(15):86418646. transporter subtype mRNAs in human brain
Fricke, M.N., D.M. Jones-Davis, and G.C. Mathews. regions and peripheral tissues. Brain Res Mol
2007. Glutamine uptake by system Atransport- Brain Res. 36:189192.
ers maintains neurotransmitter GABA syn- Nissen-Meyer, L.S., M.C. Popescu, E.H. Hamdani, and
thesis and inhibitory synaptic transmission. J F.A. Chaudry. 2011. Protein kinase C-mediated
Neurochem. 102:18951904. phosphorylation of a single serine residue on the
Helboe, L., J. Egebjerg, M. Moller, and C. Thomsen. rat glial glutamine transporter SN1 governs its
2003. Distribution and pharmacology of membrane trafficking. J Neurosci. 31:65656575.
alanine-serine-cysteine transporter 1 (asc-1) in Ortinski, P.O., J. Dong, A. Mungenast, C. Yue, H.
rodent brain. Eur J Neurosci. 18(8):22272238. Takano, D.J. Watson, P.G. Haydon, and D.A.
Jin H., H. Wu, G. Osterhaus, J. Wei, K. Davis, D. Sha, Coulter. 2010. Selective induction of astrocytic
E. Floor, C.C. Hsu, R.D. Kopke, and J.Y. Wu. 2003. gliosis generates deficits in neuronal inhibition.
Demonstration of functional coupling between Nat Neurosci. 13:584591.
gamma-aminobutyric acid (GABA) synthesis Palacin, M., R. Estevez, J. Bertran, and A. Zorzano.
and vesicular GABA transport into synaptic 1998. Molecular biology of mammalian plasma
vesicles. Proc Natl Acad Sci USA 100:42934298. membrane amino acid transporters. Physiol Rev.
Kam, K., and R.A. Nicoll. 2007. Excitatory syn- 78(4):9691054.
aptic transmission persists independently of Pines, G., N.C. Danbolt, M. Bjoras, Y. Zhang, A.
the glutamate-glutamine cycle. J Neurosci. Bendahan, L. Eide, H. Koepsell, J. Storm-Mathisen,
27:91929200. E. Seeberg, and B.I. Kanner. 1992. Cloning and
Kanai, Y., and M.A. Hediger. 2004. The gluta- expression of a rat brain L-glutamate transporter.
mate/neutral amino acid transporter family Nature 360:464467.
50 Part I: Homeostatic Regulators

Rae, C., N. Hare, W.A. Bubb, S. R. McEwan, Tani, H., C.G. Dulla, J.R. Huguenard, and
A. Broer, J.A. McQuillan, V.J. Balcar, A.D. R.J. Reimer. 2010. Glutamine is required for per-
Conigrave, and S. Broer. 2003. Inhibition of sistent epileptiform activity in the disinhibited
glutamine transport depletes glutamate and neocortical brain slice. J Neurosci. 30:12881300.
GABA neurotransmitter pools: Further evi- Uwechue, N.M., M.-C. Marx, Q. Chevy, and B.
dence for metabolic compartmentation. J Billups. 2012. Activation of glutamate transport
Neurochem. 85:503514. evokes rapid glutamine release from perisynaptic
Schoesboe, A. 2000. Pharmacological and func- astrocytes. J Physiol. 590:23172331.
tional characterization of astrocytic GABA van den Hel, W.S., R.G. Notenboom, I.W. Bos,
transport: A short review. Neurochem Res. P.C. van Rijen, C.W. van Veelen, and P.N. de
25:12411244. Graan. 2005. Reduced glutamine synthetase in
Segawa, H., Y. Fukasawa, K. Miyamoto, E. hippocampal areas with neuron loss in temporal
Takeda, H. Endou, and Y. Kanai. 1999. lobe epilepsy. Neurology 64:326333.
Identification and functional characterization Wang, Y., H.P. Zaveri, T.S. Lee, and T. Eid. 2009.
of a Na+-independent neutral amino acid trans- The development of seizures after continu-
porter with broad substrate selectivity. J Biol ous intrahippocampal infusion of methio-
Chem. 274(28):1974519751. nine sulfoximine in rats: A video-intracranial
Sepkuty, J., A. Cohen, C. Eccles, A. Rafiq, K. Behar, electroencephalographic study. Exp Neurol.
R. Ganel, D. A.Coulter, and J. Rothstein. 2002. 220:293302.
A neuronal glutamate transporter contributes to Wetherington J., G. Serrano, and R. Dingledine
neurotransmitter GABA synthesis and epilepsy. 2008. Astrocytes in the epileptic brain. Neuron
J Neurosci. 22:63726379. 58:168178.
Shank, R.P., G.S. Bennett, S.O. Freytag, and G.L. Wozny, C., S. Gabriel, K. Jandova, K. Schulze, U.
Campbell. 1985. Pyruvate carboxylase: An Heinemann, and J. Behr. 2005. Entorhinal
astrocyte-specific enzyme implicated in the cortex entrains epileptiform activity in CA1
replenishment of amino acid neurotransmitter in pilocarpine-treated rats. Neurobiol Dis.
pools. Brain Res. 329(12):364367. 19:451460.
Soltesz, I. 1995. Brief history of cortico-hippocampal Yang, S., and C.L. Cox. 2011. Attenuation of inhibi-
time with a special reference to the direct ento- tory synaptic transmission by glial dysfunction
rhinal input to CA1. Hippocampus 5:120124. in the rat thalamus. Synapse 65:12981308.
Sweatt, A.J., M.A. Garcia-Espinosa, R. Wallin, and Yang, Y., O. Gozen, A. Watkins, I. Lorenzini, A.
S.M. Hutson. 2004. Branched-chain amino acids Lepore, Y. Gao, S. Vidensky, J. Brennan, D.
and neurotransmitter metabolism: Expression Poulsen, J.W. Park, N.L. Jeon, M.B. Robinson,
of cytosolic branched-chain aminotransferase and J.D. Rothstein. 2009. Pre-synaptic regula-
(BCATc) in the cerebellum and hippocampus. J tion of astroglial excitatory neurotransmitter
Comp Neurol. 477(4):360370. transporter GLT-1. Neuron 61:880894.
Tani, H., A.E. Bandrowski, I. Parada, M. Wynn, Yu, A.C., J. Drejer, L. Hertz, and A. Schousboe. 1983.
J.R. Huguenard, D.A. Prince, and R.J. Reimer. Pyruvate carboxylase activity in primary cul-
2007. Modulation of epileptiform activity by tures of astrocytes and neurons. J Neurochem.
glutamine and system A transport in a model 41(5):14841487.
of post-traumatic epilepsy. Neurobiol Dis. Yudkoff, M. 1997. Brain metabolism of
25:230238. branched-chain amino acids. Glia 21:9298.
Tani, H., C.G. Dulla, Z. Farzampour, A. Yudkoff, M., Y. Daikhin, D. Nelson, I. Nissim, and
Taylor-Weiner, J.R. Huguenard, and R.J. Reimer. M. Erecinska. 1996. Neuronal metabolism of
2014. A local glutamate-glutamine cycle sustains branched-chain amino acids: Flux through the
synaptic excitatory transmitter release. Neuron aminotransferase pathway in synaptosomes. J
81:888900. Neurochem. 66:21362145.
4
Homeostasis ofNeuronal Excitability Via
Synaptic and Intrinsic Inhibitory Mechanisms
J O C H E N M E I E R , M A R C U S S E M T N E R , A N D J A K O B W O L FA R T

T H E M U LT I P L E FA C E T S chloride and bicarbonate, the concentrations of


OFINHIBITION VIA which are dynamically regulated by ionic trans-
G A B A A N D G LY C I N E port and enzyme activity, respectively (see later
RECEPTORS discussion for details). Generally, the opening
In the mid-20th century, the mysterious fac- of ion channels has two simultaneous effects: a
tor I included in brain extracts that inhibited decrease in membrane resistance (R m), and a
impulse generation in neurons was discovered shift of membrane potential (Vm) toward E.The
to be -aminobutyric acid (GABA), that is, the R m decrease always leads to shunting (inhibition)
main fast inhibitory transmitter of the brain due to Ohms law (U=R*I). The Vm effect depends
(Bazemore et al., 1956). The other fast central on the difference in E versus membrane poten-
inhibitory transmitter is glycine (Curtis et al., tial (Vrest, if the cell is at rest). Since E of chlo-
1968). The respective ionotropic receptors (i.e., ride (ECl) is close to the Vrest of many cell types,
ligand-gated ion channels) are GABA type the net effect of GABA A R/GlyR channel open-
A receptors (GABAA Rs) and glycine receptors ing is difficult to predict. The easiest case is an
(GlyRs), which are co-expressed in central neu- ECl negative of Vrest (Figure4.1a); here GABA AR/
rons (Jonas etal., 1998; Le-Corronc etal., 2011). GlyR activation is clearly inhibitory in the sense
They belong to the superfamily of acetylcholine that it prevents action potential (AP) genera-
receptor type ligand-gated ion channels (also tion. If ECl is positive of Vrest and positive of the
known as Cys-loop receptors), share common threshold for AP generation (e.g.45 mV), then
protein architecture and are permeable for both GABA A R/GlyR activation is initially excitatory.
chloride and bicarbonate (Bormann etal., 1987; However, this effect is reversed when GABA AR/
Kaila and Voipio, 1987). GABAA R and GlyR GlyR activation is slow and long-lasting, because
anion channels consist of five subunits, and each voltage-gated sodium (Nav) channels progres-
subunit consists of an extracellular N-terminal sively inactivate, which prevents AP generation
neurotransmitter binding domain stabilized (Figure4.1b, depolarization block). If ECl is posi-
by disulfide bridges, four transmembrane seg- tive or equal to Vrest but below the AP threshold,
ments, and a large cytosolic protein loop between GABA A R/GlyR activation will depolarize but
the third and fourth transmembrane domain itself not evoke APs; instead it will shunt fur-
at the C-terminus (Thompson et al., 2010). The ther incoming excitatory postsynaptic potentials
large cytoplasmic loop provides the structural (EPSPs), thus reducing the probability of AP
framework for functional diversification through generation (Figure4.1c, shunting inhibition). In
posttranscriptional and posttranslational addition to direct effects, GABA A R/GlyR acti-
mechanisms. vation may have secondary consequences; for
As a basic principle for all ion channels, the example hyperpolarization can trigger rebound
direction of ion currents (I) depends on the excitation via deinactivation of Nav channels.
equilibrium potential (E) of the respective ion Thus the transmembrane chloride gradient
across the plasma membrane, which in turn is needs a thorough homeostatic regulation, which
dependent on the ion concentration gradient. is achieved, among other mechanisms, via chlo-
The GABA A Rs and GlyRs are permeable for ride transporters.
52 Part I: Homeostatic Regulators

(a) (b) (c) activation, likely reflecting the KCC2-dependent


reduction of the intracellular chloride concentra-
Vm Vm Vm
+20 +20 +20 tion to about 4 to 7 mM (Rivera etal., 1999; Tyzio
etal.,2008).
Besides the developmental regulation of intra-
cellular chloride concentration through NKCC1
and KCC2 (Ben-Ari, 2002), spatial aspects of
chloride transporter function play a crucial role
for the nature of GABA action. The intracellular
45 45 45
ECl chloride concentration was found to be highly
variable between axonal, somatic, and den-
dritic compartments or even from one dendritic
Vrest Vrest Vrest ECl
branch to its neighbors (Figure4.2a, 4.2b; Duebel
ECl
et al., 2006; Waseem et al., 2010; Friedel et al.,
2013). Thus, depending on the cellular compart-
Hyper- Depol Shunt
pol block ment, activation of GABA ARs or GlyRs can have
different effects on the membrane potential of
FIGURE 4.1 Scheme depicting three forms of the same neuron and be inhibitory due to hyper-
inhibition via chloride channel opening (i.e., polarization or shunt inhibition (Banke and
GABAAR/GlyR activation). (a) When the chloride McBain, 2006; Tyzio etal., 2008; Glickfeld etal.,
equilibrium potential (ECl) is negative of the rest- 2009) or depolarizing and excitatory, the latter
ing membrane potential (Vrest), hyperpolarization eventually facilitating plasticity of glutamater-
predominates. (b) If ECl is around the threshold for gic synaptic transmission (Staley et al., 1995;
voltage-gated sodium (Nav) channel activation (around Turecek and Trussell, 2001; Ruiz et al., 2010).
-45mV) and depolarization relatively slow, Nav chan- Indeed, the intracellular chloride concentration
nels inactivate, decreasing the likelihood for AP gen- in the axonal compartment of glutamatergic
eration (depolarization block). (c) Shunting inhibition neurons is rather high (about 52 mM; Waseem
occurs due to the resistance reduction at any potential, and Fedorovich, 2010), and, consequently, activa-
but its effect becomes predominant if the chloride con- tion of chloride channels at the axon initial seg-
ductance is large and ECl is close to Vrest because then the ment or in presynaptic axon terminals is usually
cell is clamped to ECl and all inputs are shuntedaway. depolarizing (Khirug etal., 2008)and excitatory
as it facilitates glutamate release (Turecek and
Basically, two families of cation-chloride Trussell, 2001; Ruiz etal., 2010; Figure4.2a, 4.2b).
cotransporters exist and work in oppo- However, it should be noted that excitation via
site directions. Transporters of the sodium- GABA is less common than inhibition and the
potassium-chloride cotransporter (NKCC) type effect of GABAergic excitation is also less pro-
use the energy of the electrochemical sodium nounced than that of glutamatergic transmis-
gradient to load cells with chloride. On the other sion, due to the different equilibrium potential of
hand, potassium-chloride co-transporter (KCC)- chloride (~ 1020 mV) and sodium (~ 120 mV)
type transporter-mediated chloride extrusion is atVrest.
driven by the electrochemical potassium gradi- As GABA A Rs and GlyRs are also perme-
ent. Hence, both transporter types are fueled by able to bicarbonate (Bormann etal., 1987), ionic
the sodium-potassium ATPase, which establishes crosstalk between chloride, bicarbonate, and
the respective ionic gradients. For the purpose of potassium influences the functional versatil-
simplicity, we focus on NKCC1 andKCC2. ity of these receptors. Carbonic anhydrase is an
During early postnatal development, the important enzyme in this context as it catalyzes
intracellular (somatic) chloride concentration the rapid interconversion of carbon dioxide and
is estimated to range from 25 mM to 40 mM water to bicarbonate and protons (or vice versa).
(Blaesse etal., 2009), which likely reflects prepon- In pyramidal neurons, expression of carbonic
derant expression of NKCC1 at this developmen- anhydrase VII is developmentally upregulated
tal stage (Plotkin etal., 1997; Hbner etal., 2001). just like KCC2 but with a delayed time course
It has also been established that developmental (Rivera etal., 2005). The equilibrium potential of
upregulation of KCC2 expression and func- bicarbonate (EHCO3) is about -10mV (Figure4.2c);
tion correlates with the progressively decreas- that is, bicarbonate efflux during GABA AR or
ing depolarizing effect of GABAA R or GlyR GlyR activation is rather depolarizing (Kaila
Homeostasis of Neuronal Excitability 53

(a) (b) (c)

Soma Dendrites Axon Vm


Axon +20 +20 +20 +20
Soma AIS

10 EHCO3
Dendrites
45 45 45 ECl 45
ECl ECI
Vrest ECl Vrest Vrest Vrest

(d)

GABA/glycine

(Ecl) (Synchronization)
HCO3 Cl K+
(Depolarization)
GlyR
KCC2

GABAAR Gephyrin

H2O HCO3
CA7
CO2 H+ Cl
(Hyperpolarization, Ecl)

FIGURE 4.2 Subcellular heterogeneity of chloride and bicarbonate equilibrium potentials (ECl
and EHCO3, respectively). (a) Immunochemical staining of a hippocampal neuron in cell culture showing the
somato-dendritic compartment (Winkelmann and Meier, unpublished results). The axon with axon initial seg-
ment (AIS) is drawn schematically. (b) Scheme of ECl range between different neuronal compartments. In particu-
lar, the axonal ECl can be depolarized compared to the soma. See Figure 4.1 for functional consequences. (c) The
EHCO3 is more positive than ECl, irrespectively of the neuronal compartment. (d) Upon pronounced GABAergic
transmission (e.g., during seizure-like activity) in the presence of carbonic anhydrase (CA7) and the chloride
transporter KCC2, the ECl may increase due to loading of neurons with chloride (during hyperpolarization) if local
chloride extrusion capacity is saturated (KCC2), which facilitates HCO3-dependent depolarizing GABAergic
transmission in the adult nervous system. Furthermore, KCC2 can contribute to synchronization of neural net-
work activity by increasing the interstitial potassium concentration. For citation and details see maintext.

and Voipio, 1987). The net effect of GABA A R or facilitated by bicarbonate-dependent depo-
GlyR activation in the presence of carbon diox- larization (efflux; Kaila and Voipio, 1987; Kaila
ide and carbonic anhydrase depends on the rela- et al., 1989; Staley and Proctor, 1999). If the
tive permeability of bicarbonate versus chloride GABA AR-mediated current exceeds local chlo-
(PHCO3/chloride), which can be calculated using the ride transport capacity, a significant positive
Goldman-Hodgkin-Katz equation and ranges shift of ECl will occur (Staley etal., 1995), while
between 0.2 and 0.6 (Kaila, 1994). This ratio EHCO3 is kept fairly constant due to rapid buffering
suggests that GABA AR or GlyR activation can through carbonic anhydrase activity (Pasternack
trigger a biphasic change of Vm (Figure 4.2d). et al., 1993; Figure 4.2d). This mechanism of
Several studies have shown that tonic GABA A R neuronal aniondependent plasticity may pref-
activation loads neurons with chloride due erentially apply to dendrites as they show a high
to GABA AR-mediated chloride influx that is receptor-to-volume ratio (Qian and Sejnowski,
54 Part I: Homeostatic Regulators

1990). Thus, in response to GABA application or most frequently discussed (Abbott and Nelson,
high-frequency GABAergic synaptic transmis- 2000; Turrigiano and Nelson, 2000; Davis and
sion, dendritic GABA AR activation in mature Bezprozvanny, 2001; Daoudal and Debanne,
neurons gives rise to a biphasic response con- 2003; Zhang and Linden, 2003; Debanne and
sisting of initial chloride-dependent hyper- Poo, 2010). In particular, the possibility of scaling
polarization followed by bicarbonate-driven excitability without changing the relative differ-
depolarization; in acute slice preparations, hip- ences produced by synaptic plasticity in form of a
pocampal pyramidal neurons are actually excited metaplasticity is an attractive feature of intrin-
by this depolarization, which accounts for fre- sic plasticity (Abraham and Bear, 1996; Abbott
quency modulation of synaptic NMDA receptor and Nelson, 2000; Turrigiano and Nelson, 2000;
activation and synchronous gamma-frequency Davis and Bezprozvanny, 2001; Zhang and
(2080 Hz) firing in response to high-frequency Linden, 2003; Narayanan and Johnston, 2010).
stimulation (Staley etal., 1995; Ruusuvuori etal., Somewhat interfering, but also popular, is the
2004). In addition, (pathological) neuronal net- concept that intrinsic plasticity could itself be
work synchronization can occur under these a mechanism for learning and memory (Alkon,
conditions (Figure4.2d) and involve an increase 1984; Marder etal., 1996; Daoudal and Debanne,
in interstitial potassium concentration through 2003; Disterhoft and Oh, 2006; Mozzachiodi and
the KCC2-dependent net efflux of potassium and Byrne, 2010; Turrigiano, 2011). Additionally,
chloride (Viitanen etal., 2010), which depolarizes intrinsic plasticity has been discussed in the
the neuronal membrane potential but may also context of cell type identity and variability
reverse the neuronal chloride load in a homeo- (Golowasch et al., 1999; Padmanabhan and
static way, provided that local chloride transport Urban, 2010; Marder and Taylor, 2011), develop-
capacity is not saturated (Voipio and Kaila, 2000; ment (Turrigiano and Nelson, 2004; Marder and
Rivera etal., 2005). This ionic crosstalk between Goaillard, 2006), and brain diseases, in particu-
chloride, bicarbonate, and potassium can locally lar epilepsy (Beck and Yaari, 2008; Wolfart and
and transiently change the efficacy ratio between Laker,2015).
glutamatergic and GABAergic (and/or glyciner- Much of neuronal development depends on
gic) neurotransmission and thereby contribute to intrinsic plasticity. The properties of immature
synaptic plasticity and homeostatic regulation of neurons are different from those of adult neurons
high-frequency network activity. However, such (Spitzer, 1999; Moody and Bosma, 2005; Seki
a complex interplay of ionic mechanisms must et al., 2012), and neurons need to develop their
be tightly controlled because runaway excitation functions while their morphology and synaptic
may result. inputs are being built (Llinas, 1988; Spitzer, 1999;
Collectively, these considerations suggest Moody and Bosma, 2005; Marder and Goaillard,
that homeostasis of neuronal activity involves an 2006; Overstreet-Wadiche, and Westbrook, 2006;
array of spatiotemporally regulated inhibitory Pratt and Aizenman, 2007). But also, follow-
mechanisms of ionic crosstalk. ing maturation, neurons need to maintain their
functional phenotype despite constant recycling
INTRINSIC PLASTICITY (turnover) of short-lived ion channel proteins
(Marder and Goaillard, 2006). Thus neurons
What Is Intrinsic Plasticity? cannot rely solely on a genetically encoded pro-
The plasticity of nonsynaptic ion channels is sum- tein expression in a feed-forward open loop man-
marized by the term intrinsic plasticity (Siegel ner; they constantly need homeostatic feedback
etal., 1994; Zhang and Linden, 2003; Marder and mechanisms to adjust their excitability (Davis,
Goaillard, 2006). Although in principle post- 2006; Marder and Goaillard,2006).
synaptic ion channels such as AMPA receptors
could count as intrinsic, the synapse is a special- Short-Term Intrinsic Plasticity
ized structure; therefore homeostatic plasticity Neuronal excitability is due to positive feedback
affecting postsynaptic receptors is differentiated effects of voltage-gated calcium (Cav) and Nav
as synaptic scaling (Davis and Goodman, 1998; channels on membrane voltage (with excitation
Turrigiano et al., 1998; Desai, 2003), and this producing excitation). Clearly, such feedback
subject is separately treated in chapters7 and 10. cannot exist without inhibitory control. Starting
Synaptic plasticity is a prominent cellular mecha- from the millisecond-fast APs up to day-to-day
nism involved in learning and memory, and this cellular milieu changes, evolution has connected
is also the context in which intrinsic plasticity is all excitatory signals (e.g. depolarization, calcium
Homeostasis of Neuronal Excitability 55

influx, and energy consumption) directly to a coupling reinforces synaptic plasticity in both
battery of appeasing potassium channels with directions: E-S potentiation supports LTP and
many subtypes outnumbering those of excita- E-S depression accompanies long-term synap-
tory channels (Coetzee et al., 1999; Hille, 2001; tic depression (LTD; Daoudal etal., 2002; Wang
Catterall etal., 2005; Gutman etal., 2005). Thus et al., 2003). However, since E-S regulation is
control of excitation is an important function of synapse-specific (Daoudal and Debanne, 2003),
potassium channels; however, their even more it lacks the advantage of cell-wide homeostatic
fundamental purpose is maintenance of Vrest for scaling. Also, behavioral experiments found that
membrane transport of metabolites, and this associative learning paradigms permanently
function is evolutionarily much older than the reduced potassium conductances mediated by
appearance of excitable cells with Nav and Cav rapidly inactivating A-type (IA) and KCa types
channels (Anderson and Greenberg, 2001). In of potassium channels in certain types of neu-
other words, compared to potassium channel rons (Alkon, 1984; Scholz and Byrne, 1987; de
homeostasis, synaptic plasticity is evolutionarily Jonge et al., 1990). Later, LTP-inducing proto-
a recent luxury (Jan and Jan, 1994; Anderson and cols were shown to increase the intrinsic excit-
Greenberg, 2001; Hedrich,2012). ability of cerebellar and hippocampal neurons
An important factor in intrinsic plas- in slice preparations (Aizenman and Linden,
ticity is the timing of the inducing stimuli. 2000; Armano etal., 2000; Xu etal., 2005). Not
Depolarization and calcium influx are thought to surprisingly, considering the inducing protocols,
be the key signals involved in both synaptic and the cellular mechanisms underlying this intrin-
intrinsic plasticity (Thoby-Brisson and Simmers, sic plasticity appear similar to those involved
1998; Narayanan and Johnston, 2010; Wiegert in LTP; that is, they depend on calcium influx,
and Bading, 2011). Atrain of APs directly elicits NMDA receptor activation, and activation of
calcium- and sodium-dependent potassium con- Ca 2+/calmodulin-dependent protein kinase II
ductances (KCa) mediating after hyperpolariza- (Aizenman and Linden, 2000; Armano et al.,
tions (AHPs) of different lengths (up to minutes) 2000; Xu etal., 2005; Groth etal., 2011). However,
depending on the duration of the depolarization such intrinsic plasticity accompanying synaptic
(Gustafsson and Wigstrom, 1983; Schwindt etal., plasticity can also be evoked via strong post-
1989; Kim and McCormick, 1998; Sanchez-Vives synaptic depolarization alone (Aizenman and
etal., 2000; Tanner etal., 2011). Although it may Linden,2000).
be confusing to count the AHP itself as intrin- In pyramidal neurons of hippocampus and
sic plasticity, many AHP-evoking stimuli are cortex, two currents prominently involved in
certainly overlapping with those evoking some intrinsic plasticity are the hyperpolarization-
forms of intrinsic memory (Marder etal., 1996; activated cation current (IH mediated via HCN
Golowasch et al., 1999; Aizenman and Linden, channels), which mediates mostly shunting inhi-
2000). Perhaps one may speak of short-term bition, and the aforementioned IA (members of
intrinsic plasticity in analogy to short-term syn- Kv1, Kv3, and Kv4 subfamilies). Both IA and IH are
aptic plasticity (changes in synaptic strength that downregulated during LTP induction (Wang
last up to few minutes; Zucker and Regehr,2002). etal., 2003; Frick etal., 2004), possibly by NMDA/
calcium-triggered internalization of membrane
Synaptic PlasticityRelated proteins (Kim etal., 2007; Hyun etal., 2013). In
Intrinsic Plasticity fact, the decrease of IA and IH during the plasticity
It is well established that experimental stimu- induction protocol can determine whether or not
lation protocols that elicit long-term synaptic the coincidence of synaptic excitation eventu-
plasticity simultaneously trigger long-lasting ally induces LTP (Hoffman etal., 1997; Markram
intrinsic plasticity. Already in the earliest in vivo et al., 1997; Magee, 1998; Spruston, 2008).
long-term potentiation (LTP) experiments, the Interestingly, the very same LTP protocols can
potentiation of extracellularly recorded popu- lead to a reduction of excitability via IH increase
lation spikes could not be fully explained by in the cornu ammonis region 1 (CA1) pyrami-
potentiation of the EPSP alone (Bliss and Lomo, dal dendrites (Fan etal., 2005). This discrepancy
1973). This effect involved alterations of syn- could arise from experimental differences, for
aptic inhibition and/or intrinsic plasticity and example, in the age of animals (Fan etal., 2005),
was called excitation-spike (E-S) coupling (Bliss but there may also be a window for spike timing
and Lomo, 1973; Andersen etal., 1980; Daoudal dependent neuronal dynamics of intracellular
and Debanne, 2003). In the hippocampus, E-S signaling cascades involving NMDA receptors or
56 Part I: Homeostatic Regulators

Cav channels, where the individual status/history realized via a regulation on the transcriptional
of the discerned neuron decides on the direction level (Kirchheim etal., 2013). In addition, com-
of plasticity (Roth-Alpermann etal., 2006). These plex activity patterns can be homeostatically
examples identify ion channel changes as a pow- controlled. For example, the spontaneously
erful metaplasticity mechanism for the bidirec- bursting phenotype of lobster stomatogastric
tional control of the neuronal network (Abraham ganglion (STG) neurons, which is lost when
and Bear, 1996; Narayanan and Johnston,2010). cut off from the network, is autonomously rees-
tablished in cell culture via intrinsic plasticity
Chronic Intrinsic Plasticity (Turrigiano etal., 1994; Turrigiano etal., 1995;
Homeostatic intrinsic plasticity and synaptic Thoby-Brisson and Simmers,1998).
scaling related to changes of spontaneous activ- Experimental gene or RNA modulation has
ity on the time scale of days have been best stud- revealed the enormous flexibility of intrinsic
ied in cell culture models (Marder et al., 1996; plasticity. For example, the deletion of GABA A R
Turrigiano, 1999). For example, bathing the cell channels responsible for a tonic leak current
culture in blockers of synaptic inhibition not (6 and subunits) resulted in upregulation of
only leads to synaptic downscaling of AMPA and the two-pore potassium (K2P) channels, which
NMDA receptors but also to a long-term reduc- exactly compensated for the lost conductance
tion of intrinsic excitability (Lissin et al., 1998; leak (Brickley etal., 2001). Furthermore, overex-
Karmarkar and Buonomano, 2006). Similarly, pression of Kv4 A-type channels via RNA injec-
chronic depolarization via elevated extracel- tion in cultured STG neurons led to a functional
lular potassium, a major excitotoxic burden for compensation through IH upregulation, which
the cells, leads to permanent downregulation of restored the STG pacemaker pattern (MacLean
excitatory currents and upregulation of inhibi- etal., 2003). Astonishingly, this IH plasticity was
tory currents, even when AP activity is blocked observed even when nonconducting Kv4 chan-
(Franklin et al., 1992; Golowasch et al., 1999; nels were employed, that is, in the absence of
Leslie etal., 2001; van Welie etal.,2004). any functional feedback (MacLean et al., 2003;
Although it is doubtful that there are physi- MacLean et al., 2005). The latter indicates that
ological conductances that can kill a neuron some forms of intrinsic plasticity may actually
rapidly by hyperpolarization, chronic silenc- risk feed-forward open loop regulation. Most of
ing can eventually kill neurons (Eichler et al., the discussed forms of intrinsic plasticity, how-
2008). Therefore, and to achieve functional ever, are realized most likely via proper home-
homeostasis, it makes sense that chronic reduc- ostasis, that is, via intracellular set point and
tion of spontaneous AP activity (e.g., by bath- feedback mechanisms that keep the neuronal
ing cell cultures in Nav channel blockers or activity constant even in a changing environ-
AMPA and NMDA receptor inhibitors) results ment (Davis, 2006). The targeted set point of
in a compensatory long-term downregulation AP firing appears in most cases to be imple-
of dendrotoxin-sensitive delayed rectifier (Kv1) mented via the intracellular calcium level, which
channels in cornu ammonis region 3 (CA3) can be detected by calcium sensors such as the
pyramidal cells (OBrien etal., 1998; Turrigiano calcium-calmodulin-dependent protein kinase
etal., 1998; Desai etal., 1999; Turrigiano, 1999; II; this instrument faithfully translates AP fre-
Cudmore and Turrigiano, 2004; Karmarkar quency into kinase activity and downstream
and Buonomano, 2006). Similarly, chronic activation of transcription factors (LeMasson
overexpression of inward rectifier potassium et al., 1993; De Koninck and Schulman, 1998;
(K ir) channels involved in the resting potential Davis and Bezprozvanny, 2001; Weston and
(K ir2.1), leads to a compensatory downregu- Baines,2007).
lation of Kv currents in CA1 pyramidal cells In summary, intrinsic homeostatic plas-
(Okada and Matsuda, 2008). In addition to the ticity is the capacity of cells to adapt ion chan-
quantitative ion channel expression, the sub- nel expression, distribution, and function. We
cellular location of ion channels is also under discussed two partially overlapping forms of
activity-dependent homeostatic control (Grubb intrinsic plasticity: (a) a form that accompa-
and Burrone, 2010; Kuba et al., 2010). All of nies synaptic LTP in a permissive manner (i.e.,
these mechanisms of neuronal intrinsic plastic- anti-homeostatic) and (b) an evolutionarily
ity contribute to a constancy of neuronal firing older form that counteracts deviations from Vrest,
rates (Marder et al., 1996) and most likely are in particular via potassium channels.
Homeostasis of Neuronal Excitability 57

POSTTR ANSCRIPTIONAL ATP-dependent regulation of run-down of


A N D P O S T T R A N S L AT I O N A L GABA-induced currents upon successive GABA
MECHANISMS FOR applications (Zhao etal., 2006; Zhao etal., 2009),
SY NAPTIC AND INTRINSIC indicating that this site takes part in the regula-
H O M E O S TA S I S O F tion of the excitation/inhibition ratio. Moreover,
INHIBITION the genomic sequence environment of the exon
The aforementioned homeostatic inhibitory 10 is a hotspot for single nucleotide polymor-
mechanisms often involve intracellular signal- phisms that give rise to expression of differently
ing on RNA and protein levels, which includes spliced GABA AR 2 variants in patients with
RNA mechanisms (splicing and editing) and schizophrenia (Zhao et al., 2006; Zhao et al.,
posttranslational modification of proteins (phos- 2009), a disease associated with deregulation of
phorylation). Here we discuss examples of such the ratio between glutamatergic and GABAergic
molecular plasticity. synaptic transmission (Eichler and Meier, 2008).
RNA splicing of the GABA AR 2 subunit also
R NA SPLICING AND changes the protein sequence in the large cyto-
EDITING INNEURONAL plasmic loop domain; the long 2L GABA A R
INHIBITORY PLASTICITY subunit contains eight additional amino acids
As mentioned previously, chloride homeostasis that harbor a protein kinase C phosphorylation
via chloride transporters such as NKCC1 is an site (Whiting etal., 1990; Moss etal., 1992). RNA
important determinant of GABAergic/glyciner- splicing of GABA AR 2 is developmentally regu-
gic inhibition. More than 10years ago, two RNA lated (Henneberger etal., 2005), varies in different
splice variants of NKCC1 (NKCC1a/NKCC1b) brain regions (Gutierrez etal., 1994), and is altered
were isolated from human tissue (Vibat et al., in patients with schizophrenia (Huntsman etal.,
2001), but information regarding their functional 1998). Phosphorylation of the protein kinase C
role is still sparse. It is known that NKCC1b is site in the GABAA R 2L splice variant was impli-
the preponderant RNA variant in the brain; in cated in interaction with gephyrin (Meier and
contrast to NKCC1a, it lacks a protein kinase Grantyn, 2004b), a postsynaptic scaffold protein
Aconsensus site but contains a casein kinase II that facilitates neurotransmitter receptor stabi-
site (Vibat et al., 2001). Regarding KCC2, two lization at GABAergic and glycinergic synapses
splice variants (KCC2a/KCC2b) with different (Kirsch et al., 1993; Essrich et al., 1998; Meier
N-terminal parts were identified (Uvarov et al., etal., 2001; Lardi-Studler etal., 2007). RNA splic-
2007), and KCC2b seems to be upregulated ing of GlyR-coding gene transcripts also leads to
during development, whereas KCC2a expres- sequence variation in the large cytoplasmic loop
sion apparently prevails in the neonatal brain domain of 1 and 3 subunits (Malosio et al.,
(Uvarov et al., 2009). Here again, RNA splicing 1991; Nikolic etal., 1998)or changes the protein
creates additional sites for protein modifica- sequence of the ligand-binding domain of GlyR
tion through phosphorylation, and, according 2 (Kuhse etal., 1991). While the functional rel-
to their subcellular distribution, the different evance of GlyR 1 RNA splicing remains largely
KCC2a and KCC2b RNA splice variants seem unclear, it is well established that GlyR 2 splic-
to function preferentially at distal and proximal ing changes apparent neurotransmitter affinity
somatodendritic compartments, respectively (Miller et al., 2004). In contrast, GlyR 3 splic-
(Markkanen et al., 2014). However, RNA splice ing impacts on subcellular receptor distribution,
variantspecific functional roles of KCC2 remain receptor clustering, and desensitization kinetics
to be determined. (Nikolic etal., 1998; Eichler etal., 2009; Notelaers
Several RNA splice variants of GlyR-coding et al., 2012; Winkelmann et al., 2014). In par-
gene transcripts exist, whereas only GABA A R 2 ticular, expression of 2B- and 3K-GlyR RNA
and 2 subunits seem to undergo functional diver- splice variants were shown to be upregulated in
sification through RNA splicing. GABA AR 2L the sclerotic hippocampus of patients with idi-
contains a 38 amino acidlong splice insert that opathic temporal lobe epilepsy (TLE), suggesting
adds a potential calcium-calmodulin-dependent that they may contribute to neurodegeneration
kinase II phosphorylation site to the large cyto- (Eichler et al., 2008). In fact, nonsynaptic tonic
plasmic loop domain (McKinley et al., 1995). GlyR activation and the resulting decrease of AP
Use of phosphomimetic molecular constructs firing due to shunting inhibition were shown to
revealed that this site may be important for be harmful to neurons with low KCC2 protein
58 Part I: Homeostatic Regulators

expression (Eichler et al., 2008). On the other of neuron where this form of molecular plasticity
hand, upregulation of tonic GABA A R currents occurs (Gonzalez,2013).
in the dentate gyrus of the epileptic tissue may RNA splicing can furthermore contribute to
have protective effects for some cell types (Young intrinsic potassium channel plasticity mentioned
etal., 2009), emphasizing the implications of cell before. For example, alternative splicing in the C
type- and brain region-specific mechanisms for terminal domaincoding region changes subcel-
homeostatic inhibitory mechanisms. lular channel trafficking of Kv3.1 channels while
Gephyrin is a central organizer of postsynap- it does not affect biophysical channel proper-
tic GABAergic and glycinergic domains (Tretter ties. The differential propensities of Kv3.1a and
etal., 2012). It consists of three domains (G, C, and Kv3.1b splice variants for supporting fast spik-
E), which evolved from exon shuffling of homolo- ing could rely on axonal trafficking of Kv3.1b
gous bacterial proteins involved in molybdenum (Gu etal., 2012). Indeed, splicing of Kv3.1-coding
cofactor synthesis (Schwarz etal., 2009). Due to RNA can act as a regulatory switch between neu-
its central role in postsynaptic receptor stabiliza- ronal compartmentspecific forms of neuronal
tion, RNA splicing of gephyrin-coding mRNA plasticity because only the axonal Kv3.1b effec-
could play a significant role in regulation of the tively converted slow-spiking young neurons to
strength of GABAergic and glycinergic synapses. fast-spiking ones, while somatodendritic expres-
Indeed, a considerable number of RNA splice sion of Kv3.1a was far less effective at increasing
variants were isolated (Fritschy etal., 2008)and the maximal firing frequency (Gu et al., 2012).
basically fall into two groups. The glia cell-specific This finding suggests different properties of som-
RNA splice variant (containing the C3 peptide atodendritic and axonal compartments in the
insert in the C domain), for example, was shown expression of synaptic and/or intrinsic homeo-
to be involved in molybdenum cofactor synthesis static neuronal plasticity.
in the brain (Smolinsky etal., 2008), while RNA In addition to the functional diversification
splicing in the G domaincoding part of gephy- through RNA splicing, neurotransmitter recep-
rin was reported to regulate synaptic receptor tors and potassium channels can undergo RNA
clustering in neurons (Meier and Grantyn, 2004a; editing. Adenosine-to-inosine RNA editing of
Bedet etal., 2006). Sequence variations of the G GABA AR 3 is developmentally upregulated,
domain could be disease relevant as different influences receptor surface expression (Ohlson
gephyrin splice variants lacking several exons in et al., 2007; Daniel et al., 2011), and eventually
the G domain are expressed in the hippocampus contributes to the glutamate-dependent develop-
of TLE patients and downregulate the strength mental and cognitively relevant 3-to-1 subu-
of GABAergic synaptic transmission (Frstera nit switch of GABA AR expression (Henneberger
etal., 2010). In fact, hippocampal regionspecific et al., 2005). Cytidine-to-uridine RNA edit-
dysfunction of the RNA splice machinery and ing of GlyR 2 and 3 subunits changes the
the resulting impairment of GABAergic synaptic apparent neurotransmitter affinity by about
inhibition in the CA3 region may contribute to 10-fold (glycine) or even 50-fold (taurine), ren-
increased excitability of CA3 pyramidal neurons ders GlyRs responsive to GABA (Meier et al.,
(Frstera et al., 2010). In addition to cell stress 2005; Legendre et al., 2009), and even leads to
as a reason for exon skipping during gephyrin spontaneous receptor activity in the nominal
RNA splicing (Frstera et al., 2010), deletions absence of a neurotransmitter (Kletke et al.,
in the G domaincoding part of gephyrin can 2013; Winkelmann et al., 2014). The RNA edit-
also have a genetic origin, as was evidenced in a ing of GlyR 2- and 3-coding gene transcripts
wide range of patients with different neuropsy- is increased in TLE patients (Eichler etal., 2008).
chiatric symptoms (Lionel et al., 2013). These Depending on spatiotemporal coregulation of
findings may define a new genetic and RNA GlyR RNA splicing and editing and subcellular
processingdependent hotspot for pathological differences in receptor location, the resulting
gephyrin protein sequence variation. However, GlyR protein variants can elicit neurodegenera-
whether or not downregulation of postsynaptic tion via increased tonic inhibition of neuronal
gephyrin is a pathological or beneficial mecha- AP firing (Eichler et al., 2008; Legendre et al.,
nism of neuronal plasticity depends on many 2009) and/or trigger neuropsychiatric symp-
factors, including spatiotemporal regulation of toms reminiscent of the psychopathology of
ionic equilibrium potentials and crosstalk with epilepsy due to enhancement of presynaptic neu-
intrinsic neuronal plasticity, as well as the type ronal function (Winkelmann et al., 2014). This
Homeostasis of Neuronal Excitability 59

suggests that RNA processing of GlyR-coding or maladaptive disease-promoting mechanisms


gene transcripts is a disease-promoting mecha- depends on developmental characteristics of
nism of neuronal molecular plasticity. On the glycinergic and GABAergic neurotransmission.
other hand, adenosine-to-inosine RNA editing Undoubtedly, the ratio between NKCC1 and
of Kv1.1 channels and the resulting change in the KCC2 activity is relevant for regulation of the
current-voltage relationship may serve as a com- nature (excitatory or inhibitory) of glycinergic
pensatory mechanism against epileptic seizures and GABAergic signaling in the central nervous
(Streit etal., 2011; Kirchheim etal.,2013). system, and altered chloride homeostasis due to
Collectively, RNA processing of gene tran- the dysfunction of NKCC1 and/or KCC2 impacts
scripts coding for anion permeable neuro- neuronal excitability and cell volume regulation
transmitter receptors, the postsynaptic scaffold in disease (Kahle etal.,2008).
protein gephyrin, and the plasticity of potassium There is compelling evidence for
channels create considerable functional diversity phosphory lation-dependent modification of
and an important level for the regulation of inter- GABA AR function. Sixteen GABA A R subunits
action of homeostatic synaptic and intrinsic plas- are known to date, and most of them can be phos-
ticity in the healthy and diseasedbrain. phorylated involving serine/threonine-directed
protein kinases or kinases with substrate speci-
Phosphorylation ficity for tyrosine residues (Luscher etal., 2011).
One important switch for the regulation of Basically, most of the phosphorylation sites are
protein function that also plays a major role in located in the large cytoplasmic loop domain,
functional homeostasis is protein phospho- which is a central determinant of subcellular
rylation and dephosphorylation via kinases receptor distribution and biophysical receptor
and phosphatases, respectively. For exam- properties.
ple, the functional properties of NKCC1 and The pioneering studies by Stephen Moss dis-
KCC2 cotransporters are reciprocally regulated closed many different phosphorylation sites and
by serine/threonine phosphorylation. With their impact on regulation of GABA AR surface
no lysine = K kinase-dependent activation expression (Kittler and Moss, 2003; Vithlani
of NKCC1 and inhibition of KCC2 through and Moss, 2009). Phosphorylation of GABAA R
Ste20-related proline alanine-rich/oxidative 2S (Krishek etal., 1994)may be relevant in the
stress response-1 signaling cascades (Uvarov context of homeostatic regulation of brain func-
et al., 2007; Kahle et al., 2010) may be consid- tion as it modulates GABA AR stabilization in an
ered homeostatic mechanisms because these activity-dependent way, involving activation of
pathways are activated in conditions of hyper- the NMDA receptor and the protein phosphatase
tonic cell stress (Xu etal., 2000)and depletion of calcineurin in rapid dispersal of postsynaptic
intracellular chloride (Ponce-Coria etal., 2008). 2-containing GABA ARs (Muir etal., 2010). This
Activity-dependent bidirectional regulation of may present an antihomeostatic mechanism as it
KCC2 function through phosphorylation is well supports glutamate-biased weighting of excita-
established (Chamma et al., 2012; Kahle et al., tory and inhibitory synapses in the context
2013), and several different serine/threonine and of learning-associated synaptic plasticity. Serine
tyrosine phosphorylation sites in the C-terminal phosphorylation of the GlyR subunit through
domain can contribute to up- or downregula- protein kinase C may also contribute to maladap-
tion of KCC2-mediated chloride extrusion (Lee tive forms of synaptic plasticity as it downregu-
et al., 2007; Rinehart et al., 2009; Watanabe lates postsynaptic gephyrin clustering (Specht
et al., 2009). Oxidative stress and induction of etal.,2011).
seizure activity change the KCC2 phosphoryla- Gephyrin phosphorylation plays a major role
tion status through signaling events that involve in GABAergic synaptic plasticity. In particular,
brain-derived neurotrophic factor, calcium, pro- glycogen synthase kinase GSK3-dependent and
tein kinase C, TrkB tyrosine kinase, and protein extracellular signal-related kinasemediated
phosphatases, which triggers downregulation of phosphorylation of two adjacent serine residues
KCC2-mediated chloride extrusion in the adult in the C domain of gephyrin reduce the num-
brain (Rivera et al., 2002; Rivera et al., 2004; ber of postsynaptic gephyrin clusters and func-
Wake etal., 2007; Lee etal., 2010; Lee etal., 2011). tional GABAergic synapses (Tyagarajan et al.,
Whether or not downregulation of KCC2 chlo- 2011; Tyagarajan etal., 2013), which may present
ride transport activity contributes to homeostatic another antihomeostatic mechanism. Lithium is
60 Part I: Homeostatic Regulators

a GSK3 inhibitor that is used as mood-stabilizing we assume that the hypothesis of seizure-evoking
drug, which again suggests that gephyrin mal- mechanisms via hyperexcitable CA1 pyramidal
function contributes to neuropsychiatric disease cells applies for the TLE patient population with-
(Frstera etal., 2010; Lionel etal., 2013). However, out hippocampal sclerosis (which also exists) or
we also know that GSK3-signaling impacts on a represents a presclerosis stage of human TLE.
wide range of cellular processes in the healthy Many other dysfunctional potassium chan-
and diseased brain, including Alzheimers dis- nels have been implicated in epileptic disor-
ease, bipolar disorder, Fragile X syndrome, schiz- ders (Wolfart and Laker, 2015). Examples are
ophrenia, and stroke (Kaidanovich-Beilin etal., G proteincoupled inward rectifier (Girk, K ir3)
2012). Therefore, whether or not GSK3-dependent channels (Signorini et al., 1997), KCa channels
regulation of postsynaptic gephyrin contributes of the big conductance (Brenner etal., 2005; Du
to disease or represents a beneficial homeostatic etal., 2005)and small conductance type (Schulz
mechanism of GABAergic synapse plasticity et al., 2012), as well as Kv channels (e.g., Kv7.2/3
remains to be determined. [Biervert etal., 1998; Main etal.,2000] and Kv1.1
Also, potassium channels are phosphoryl- channels [Smart et al., 1998]). A new channel-
ated, and phosphorylation influences surface epsy nomenclature was even proposed for potas-
expression and function of potassium channels. sium channelrelated epilepsies (DAdamo etal.,
There is a large number of involved potassium 2013). Less frequently reported but also impor-
channels and an even larger number of phos- tant are gain-of-function mutations in potas-
phorylation sites (Vacher and Trimmer, 2011). sium channels leading to enhanced excitability,
Here we limit ourselves to emphasizing that as in the case of big conductancetype potassium
phosphorylation-dependent internalization has channels, which allow higher firing frequencies
been implicated as an intracellular mechanism when upregulated (Brenner etal.,2005).
in the functional downregulation of potassium In contrast to the aforementioned procon-
channels as it occurs during LTP protocols and vulsive channelepsies, anticonvulsive ion chan-
epilepsy (Bernard etal., 2004; Lugo etal., 2008; nel changes have received much less attention.
Hyun etal.,2013). More recently though, several forms of such
homeostatic ion channel regulations have been
E P I L E P S Y- R E L AT E D highlighted (Figure 4.3a). Dentate gyrus gran-
INTRINSIC AND ule cells downregulate their excitability not only
SY NAPTIC PLASTICIT Y in an animal model of TLE but also in sclerotic
hippocampi of TLE patients (Stegen etal., 2009;
Epilepsy-Related Intrinsic Young etal., 2009; Stegen etal., 2012; Kirchheim
Plasticity etal., 2013). It is perhaps related to the gate-keeper
It is not coincidental that neuronal plasticity function of the dentate (Hsu, 2007)that granule
and epilepsy share mechanisms: LTP-evoking cells appear as veritable experts in the downscal-
protocols resemble epileptic seizures very much ing of intrinsic excitability.
(Leite et al., 2005). Those most frequently dis- A whole range of inhibitory ion channels was
cussed are cases of pathological ion channel found permanently upregulated in granule cells
changes (channelopathies), which lend them- of epileptic tissue (e.g., leak channels control-
selves to be potentially seizure-causing ones ling Vrest and input resistance, such as Kir2.1-4
(Chang and Lowenstein, 2003; George, 2004; channels and K2P weak inward rectifier chan-
Rogawski and Loscher, 2004; Beck and Yaari, nels; Stegen etal., 2009; Young etal., 2009)and
2008). Again, the usual suspects IA (e.g., Kv4.2) axonal Kv channels (e.g., Kv1.1), which control AP
and IH (e.g., HCN1) but also T-type Cav channels delay (Kirchheim et al., 2013). There are inter-
were reported as misregulated; particularly in esting molecular differences between animal
TLE models without hippocampal sclerosis such models of TLE and the human TLE; while in the
as systemic pilocarpine-induced status epilepti- severely sclerotic tissue of a mouse TLE model,
cus, CA1 pyramidal cells were found hyperex- a tonic GABA A leak conductance is increased,
citable due to such ion channel changes (Tsaur respective human granule cells of TLE patients
et al., 1992; Castro et al., 2001; Su et al., 2002; showed enhanced IH currents (Young etal., 2009;
Bernard et al., 2004; Bender and Baram, 2007; Stegen etal., 2012). Both IH and tonic GABA A are
Dyhrfjeld-Johnsen etal., 2009; Noam etal., 2011). slightly depolarizing but shunting inhibitory for
Because in TLE patients with hippocampal scle- dentate granule cells and thereby counterbalance
rosis this CA1 cell population is severely depleted, the effect of increased potassium conductances
Homeostasis of Neuronal Excitability 61

on Vrest without reversing their decrease of excit- here on a few selected target proteins involved
ability (Young et al., 2009; Stegen et al., 2012). in inhibitory synaptic transmission. Calpain
The differences between human and mouse gran- belongs to the family of calcium-dependent,
ule cells could be due to the more severe epileptic nonlysosomal cysteine proteases. It cleaves a
phenotype of the animal model (Isokawa, 1996; wide range of proteins most likely because ter-
Haussler etal., 2012)or species differences. The tiary protein structural determinants rather than
similar functional outcome of these changes is primary amino acid sequences define substrate
consistent with the hypothesis that neurons have specificity.
perhaps many alternative (ion channel) routes to Gephyrin is a calpain substrate (Kawasaki
achieve output homeostasis via intrinsic excit- et al., 1997). In fact, ERK and GSK3 cooper-
ability (Brickley et al., 2001; Prinz et al., 2004; ate and phosphorylate serine residues 268 and
Young etal., 2009; Stegen etal., 2012; Figure4.3a). 270, respectively, in the central C domain of
Furthermore, it appears that the intrinsic plastic- gephyrin, which may render gephyrin more
ity is not installed to simply switch off these gran- susceptible to calpain-mediated proteolysis due
ule cells in overcompensation but instead to scale to phosphorylation-dependent conformational
down their excitability in a function-maintaining changes (Tyagarajan etal., 2011; Tyagarajan etal.,
manner during upstream hyperexcitation. For 2013). Furthermore, we know that gephyrin is
the dentate gyrus, the upstream driving source extensively spliced in this protein region, which
likely is the entorhinal cortex, which during TLE may involve RNA splicing in conformational
shows enhanced excitatory output (Kobayashi changes and increased susceptibility of gephyrin
et al., 2003). Consistent with the hypothesis of to proteolysis (Herweg and Schwarz,2012).
granule cell output homeostasis, a recent mod- Evidence from in vivo studies may support
eling study showed that the observed intrinsic a pathophysiological role for proteolytic gephy-
plasticity is in a position to restore the network rin processing in TLE patients (Frstera et al.,
performance in a task of pattern separation 2010; Fang et al., 2011), and recent studies have
despite epileptic wiring (Yim etal.,2015). also demonstrated downregulation of gephyrin
In addition to antiexcitability strategies, and GABA ARs in an animal model of epilepsy
pharmacological support of neuronal intrinsic where the muscarinic acetylcholine receptor
anticonvulsive mechanisms of neurons could agonist pilocarpine is used for induction of sta-
be a powerful concept for antiepileptic drugs. tus epilepticus (Fang etal., 2011; Gonzalez etal.,
However, one has to carefully examine the tar- 2013). However, KCC2 is also a calpain target
geted cell type because, depending on the cel- (Puskarjov et al., 2012), and NMDA receptor
lular conductance environment, increasing a activation enhances calpain-mediated proteoly-
hyperpolarizing current can eventually enhance sis of KCC2. Here again, phosphorylation seems
the network excitability, for example by deinac- to play a key role in the regulation of calpain
tivation of excitatory channels or by supporting susceptibility because pilocarpine increased
inhibitory synchronization that underlies certain tyrosine phosphorylation and lysosomal deg-
epilepsy forms (McCormick and Contreras, 2001; radation of KCC2 (Lee et al., 2010). Hence,
Wickenden, 2002; Brenner etal., 2005). Another activity-dependent calpain action on gephy-
important aspect is changed network connectiv- rin and KCC2 should restrain both GABAergic
ity during epilepsy. In particular, the imbalance of neurotransmission-dependent chloride load and
different forms of inhibition in the CA regions and potassium-dependent (KCC2-mediated) rebound
the subiculum has to be studied carefully when burst activity of neurons with expression of car-
considering the potential impact of intrinsic plas- bonic anhydrase (see Figure4.2d), suggesting that
ticity during TLE (Cohen etal., 2002; Magloczky this seizure-induced molecular plasticity may
and Freund, 2005; Cohen et al., 2006; Wittner represent a homeostatic form of neuronal synap-
et al., 2009; Frstera et al., 2010; see also previ- tic and intrinsic plasticity. Indeed, that neonatal
ous discussion for RNA processingdependent seizure activity increases KCC2 function (Khirug
mechanisms of neuronal plasticity). etal., 2010)in the absence of carbonic anhydrase
(Ruusuvuori etal., 2004)supports the view that
Epilepsy-Related Synaptic seizure-dependent bidirectional regulation of
Plasticity KCC2 function is part of a neuronal homeostatic
Activity (calcium)-dependent proteolysis impacts adaptive mechanism that aims at protecting the
on the entire cellular proteome, but we focus system against pathological hypersynchronous
62 Part I: Homeostatic Regulators

(a)

(b)

FIGURE 4.3 Scheme summarizing a selection of intrinsic and synaptic plasticity mechanisms
relevant to hippocampal and other epilepsies. (a) In reaction to seizures invading the hippocampus
via the perforant path (red triangle) to the dentate gyrus (DG), granule cells homeostatically downscale their
intrinsic excitability via transcriptional upregulation of leak channels such as classic inward rectifier K+ (K ir2.1-4),
two-pore K+ (K 2P) channels, voltage-gated K+ (Kv1.1) channels, as well as shunting conductances such as tonic
chloride currents via GABA type Areceptors (GABA AR, * in a mouse TLE model) or hyperpolarization-activated
cation (HCN1) channels (** in sclerotic tissue of TLE patients) (Stegen et al., 2009; Young et al., 2009; Stegen
et al., 2012; Kirchheim et al., 2013). In TLE models without hippocampal sclerosis, inhibitory synaptic influ-
ences are decreased in the cornu ammonis (CA) region of the hippocampus (right panel, green triangle), and the
intrinsic excitability of particularly CA1 pyramidal neurons is further increased due to reduced expression of
dendritic A-type (Kv4) and HCN channels, as well as increased influence of T-type voltage-gated calcium chan-
nels (Cav3) (Tsaur etal., 1992; Castro etal., 2001; Su etal., 2002; Bernard etal., 2004; Bender and Baram, 2007;
Dyhrfjeld-Johnsen et al., 2009; Noam et al., 2011). (b) Epilepsy-related changes of presynaptic molecules can
increase (red, orange) or decrease (green) network excitability when expressed in glutamatergic neurons while
the opposite holds true if the changes occur in GABAergic neurons. For a review see Meier etal. (2014). These
examples emphasize the importance of specific neuron types in the regulation of neural network homeostasis.
Further abbreviations:Nav,=voltage-gated sodium channels; GlyR=glycine receptors; GABABR=GABA type B
receptors. Panel b:modified with permission from Meier etal. (2014).

neural network activity. The fact that respiratory Knockout of the presynaptic protein syndapin
CO2-regulatory regimes are able to interrupt sei- I also results in an epileptic phenotype (Koch
zure activity (Schuchmann etal., 2006)further- et al., 2011). Furthermore, presynaptic GABA B
more accentuates the relevance of bidirectional autoreceptor-mediated interneuron type-specific
homeostatic adaptive regulation of KCC2 expres- reduction of inhibition is associated with net-
sion as a function of neuronal carbonic anhy- work hyperexcitability in an animal model of
drase expression. epilepsy (Dugladze et al., 2013; Figure 4.3b).
However, the homeostatic machinery may Also, ectopic AP generation was shown to be a
also fail in disease conditions. Actually, there critical mechanism of network hyperexcitability,
is increasing evidence that supports a critical identifying axo-axonic connecting neurons as
role for the axonal and/or presynaptic compart- possible targets for new therapeutic strategies to
ment in epilepsy as it may escape homeostatic treat epilepsy (Dugladze etal.,2012).
regulation (Figure4.3b; Eichler and Meier, 2008; The multifaceted clinical picture of patients
Meier et al., 2014). For example, changes in the with epilepsy involves sudden episodes of cogni-
expression of presynaptic vesicleassociated tive dysfunction and psychiatric comorbidities,
proteins such as synapsins can elicit neural including depression and anxiety (Currie et al.,
network hyperexcitability (Fassio et al., 2011). 1971; Beyenburg etal., 2005; Garcia-Morales etal.,
Homeostasis of Neuronal Excitability 63

2008), which suggests common neuronal and Banke, T.G., and McBain, C.J. (2006). GABAergic
molecular mechanisms. Arecent study identified input onto CA3 hippocampal interneurons
a presynaptic molecule that can trigger cognitive remains shunting throughout development. J
dysfunction and anxiety depending on whether Neurosci. 26, 1172011725.
it is expressed in principal glutamatergic neurons Bazemore, A., Elliott, K.A., and Florey, E. (1956).
or a specific type of GABAergic interneuron (i.e., Factor Iand gamma-aminobutyric acid. Nature
parvalbumin-positive interneurons), respectively 178, 10521053.
(Winkelmann et al., 2014). In agreement with Beck, H., and Yaari, Y. (2008). Plasticity of intrinsic
possible common molecular mechanisms of epi- neuronal properties in CNS disorders. Nat. Rev.
lepsy, increased presynaptic functionality may Neurosci. 9, 357369.
represent a critical factor in the dysregulation of Bedet, C., Bruusgaard, J.C., Vergo, S., Groth-Pedersen,
neural network homeostasis (Figure 4.3b) and L., Eimer, S., Triller, A., and Vannier, C. (2006).
identify specific neuron types that can trigger the Regulation of gephyrin assembly and glycine
diverse neuropsychiatric symptoms of the dis- receptor synaptic stability. J. Biol. Chem. 281,
3004630056.
ease (Winkelmann etal., 2014; Meier etal.,2014).
Ben-Ari, Y. (2002). Excitatory actions of gaba dur-
In conclusion, the homeostasis of neuronal
ing development:The nature of the nurture. Nat.
excitability via intrinsic and synaptic inhibitory
Rev. Neurosci. 3, 728739.
mechanisms accompanies neurons throughout
Bender, R.A., and Baram, T.Z. (2007). Epileptogenesis
their lifes and becomes crucial during chronic
in the developing brain:What can we learn from
hyperexcitability as in epilepsy. The changes are
animal models? Epilepsia. 48 Suppl 5,26.
often very specific for cell types and even for Bernard, C., Anderson, A., Becker, A., Poolos, N.P.,
subcellular compartments. Therefore, further Beck, H., and Johnston, D. (2004). Acquired den-
cell typespecific investigations are needed to dritic channelopathy in temporal lobe epilepsy.
better understand the impact of molecules, neu- Science 305, 532535.
rons, and local networks in neuropsychiatric Beyenburg, S., Mitchell, A.J., Schmidt, D., Elger, C.E.,
diseases. and Reuber, M. (2005). Anxiety in patients with
epilepsy: Systematic review and suggestions for
References clinical management. Epilepsy Behav. 7, 161171.
Abbott, L.F., and Nelson, S.B. (2000). Synaptic plas- Biervert, C., Schroeder, B.C., Kubisch, C., Berkovic,
ticity:Taming the beast. Nat Neurosci. 3(Suppl.), S.F., Propping, P., Jentsch, T.J., and Steinlein,
11781183. O.K. (1998). A potassium channel mutation in
Abraham, W.C., and Bear, M.F. (1996). neonatal human epilepsy. Science 279, 403406.
Metaplasticity: The plasticity of synaptic plastic- Blaesse, P., Airaksinen, M.S., Rivera, C., and Kaila,
ity. Trends Neurosci. 19, 126130. K. (2009). Cation-chloride cotransporters and
Aizenman, C.D., and Linden, D.J. (2000). Rapid, neuronal function. Neuron 61, 820838.
synaptically driven increases in the intrinsic Bliss, T.V., and Lomo, T. (1973). Long-lasting potenti-
excitability of cerebellar deep nuclear neurons. ation of synaptic transmission in the dentate area
Nat. Neurosci. 3, 109111. of the anaesthetized rabbit following stimulation
Alkon, D.L. (1984). Calcium-mediated reduction of the perforant path. J Physiol 232, 331356.
of ionic currents: A biophysical memory trace. Bormann, J., Hamill, O.P., and Sakmann, B. (1987).
Science. 226, 10371045. Mechanism of anion permeation through chan-
Andersen, P., Sundberg, S.H., Sveen, O., Swann, J.W., nels gated by glycine and gamma-aminobutyric
and Wigstrom, H. (1980). Possible mechanisms acid in mouse cultured spinal neurones. J Physiol
for long-lasting potentiation of synaptic trans- 385, 243286.
mission in hippocampal slices from guinea-pigs. Brenner, R., Chen, Q.H., Vilaythong, A., Toney, G.M.,
J. Physiol. 302, 463482. Noebels, J.L., and Aldrich, R.W. (2005). BK
Anderson, P.A., and Greenberg, R.M. (2001). channel beta4 subunit reduces dentate gyrus
Phylogeny of ion channels:Clues to structure and excitability and protects against temporal lobe
function. Comp Biochem. Physiol B Biochem. seizures. Nat. Neurosci. 8, 17521759.
Mol. Biol. 129,1728. Brickley, S.G., Revilla, V., Cull-Candy, S.G., Wisden, W.,
Armano, S., Rossi, P., Taglietti, V., and DAngelo, E. and Farrant, M. (2001). Adaptive regulation of
(2000). Long-term potentiation of intrinsic neuronal excitability by a voltage-independent
excitability at the mossy fiber-granule cell potassium conductance. Nature 409,8892.
synapse of rat cerebellum. J. Neurosci. 20, Castro, P.A., Cooper, E.C., Lowenstein, D.H., and
52085216. Baraban, S.C. (2001). Hippocampal heterotopia
64 Part I: Homeostatic Regulators

lack functional Kv4.2 potassium channels in the hippocampal pyramidal neurons. Proc. Natl.
methylazoxymethanol model of cortical malfor- Acad. Sci. U.S. A. 99, 1451214517.
mations and epilepsy. J. Neurosci. 21, 66266634. Davis, G.W. (2006). Homeostatic control of neu-
Catterall, W.A., Goldin, A.L., and Waxman, S.G. ral activity: From phenomenology to molecu-
(2005). International Union of Pharmacology. lar design. Annu. Rev. Neurosci. 29:30723.,
XLVII. Nomenclature and structure-function 307323.
relationships of voltage-gated sodium channels. Davis, G.W., and Bezprozvanny, I. (2001).
Pharmacol. Rev. 57, 397409. Maintaining the stability of neural func-
Chamma, I., Chevy, Q., Poncer, J.C., and Levi, S. tion: A homeostatic hypothesis. Annu. Rev.
(2012). Role of the neuronal K-Cl co-transporter Physiol. 63, 847869.
KCC2 in inhibitory and excitatory neurotrans- Davis, G.W., and Goodman, C.S. (1998). Genetic
mission. Front Cell Neurosci.6,5. analysis of synaptic development and plastic-
Chang, B.S., and Lowenstein, D.H. (2003). Epilepsy. ity:Homeostatic regulation of synaptic efficacy.
N. Engl. J.Med. 349, 12571266. Curr. Opin. Neurobiol. 8, 149156.
Coetzee, W.A., Amarillo, Y., Chiu, J., Chow, A., de Jonge, M.C., Black, J., Deyo, R.A., and Disterhoft,
Lau, D., McCormack, T., Moreno, H., Nadal, J.F. (1990). Learning-induced afterhyperpolar-
M.S., Ozaita, A., Pountney, D., Saganich, M., ization reductions in hippocampus are specific
Vega-Saenz de, M.E., and Rudy, B. (1999). for cell type and potassium conductance. Exp.
Molecular diversity of K+ channels. Ann. N. Y. Brain Res. 80, 456462.
Acad. Sci. 868, 233285. De Koninck, P., and Schulman, H. (1998). Sensitivity
Cohen, I., Huberfeld, G., and Miles, R. (2006). of CaM kinase II to the frequency of Ca2+ oscil-
Emergence of disinhibition-induced synchrony lations. Science 279, 227230.
in the CA3 region of the guinea pig hippocam- Debanne, D., and Poo, M.M. (2010). Spike-timing
pus in vitro. J Physiol. 570, 583594. dependent plasticity beyond synapsepre- and
Cohen, I., Navarro, V., Clemenceau, S., Baulac, M., post-synaptic plasticity of intrinsic neuronal
and Miles, R. (2002). On the origin of interic- excitability. Front Synaptic. Neurosci.2,21.
tal activity in human temporal lobe epilepsy in Desai, N.S. (2003). Homeostatic plasticity in the
vitro. Science 298, 14181421. CNS: Synaptic and intrinsic forms. J. Physiol
Cudmore, R.H., and Turrigiano, G.G. (2004). Paris. 97, 391402.
Long-term potentiation of intrinsic excitability Desai, N.S., Rutherford, L.C., and Turrigiano, G.G.
in LV visual cortical neurons. J. Neurophysiol. (1999). Plasticity in the intrinsic excitability of cor-
92, 341348. tical pyramidal neurons. Nat. Neurosci. 2, 515520.
Currie, S., Heathfield, K.W., Henson, R.A., and Scott, Disterhoft, J.F., and Oh, M.M. (2006). Learning,
D.F. (1971). Clinical course and prognosis of aging and intrinsic neuronal plasticity. Trends
temporal lobe epilepsy:Asurvey of 666 patients. Neurosci. 29, 587599.
Brain 94, 173190. Du, W., Bautista, J.F., Yang, H., Diez-Sampedro, A.,
Curtis, D.R., Hosli, L., and Johnston, G.A. (1968). A You, S.A., Wang, L., Kotagal, P., Luders, H.O.,
pharmacological study of the depression of spi- Shi, J., Cui, J., Richerson, G.B., and Wang, Q.K.
nal neurones by glycine and related amino acids. (2005). Calcium-sensitive potassium channelo-
Exp. Brain Res. 6,118. pathy in human epilepsy and paroxysmal move-
DAdamo, M.C., Catacuzzeno, L., Di, G.G., ment disorder. Nat. Genet. 37, 733738.
Franciolini, F., and Pessia, M. (2013). K(+) Duebel, J., Haverkamp, S., Schleich, W., Feng, G.,
channelepsy: Progress in the neurobiology of Augustine, G.J., Kuner, T., and Euler, T. (2006).
potassium channels and epilepsy. Front. Cell. Two-photon imaging reveals somatodendritic
Neurosci. 7,134. chloride gradient in retinal ON-type bipolar cells
Daniel, C., Wahlstedt, H., Ohlson, J., Bjork, P., expressing the biosensor Clomeleon. Neuron
and Ohman, M. (2011). Adenosine-to-inosine 49,8194.
RNA editing affects trafficking of the gamma- Dugladze, T., Maziashvili, N., Borgers, C.,
aminobutyric acid type A(GABA(A)) receptor. J Gurgenidze, S., Haussler, U., Winkelmann, A.,
Biol. Chem. 286, 20312040. Haas, C.A., Meier, J.C., Vida, I., Kopell, N.J., and
Daoudal, G., and Debanne, D. (2003). Long-term Gloveli, T. (2013). GABAB autoreceptor-mediated
plasticity of intrinsic excitability:Learning rules cell type-specific reduction of inhibition in epi-
and mechanisms. Learn. Mem. 10, 456465. leptic mice. Proc. Natl. Acad. Sci. U. S. A 110,
Daoudal, G., Hanada, Y., and Debanne, D. (2002). 1507315078.
Bidirectional plasticity of excitatory postsyn- Dugladze, T., Schmitz, D., Whittington, M.A., Vida, I.,
aptic potential (EPSP)-spike coupling in CA1 and Gloveli, T. (2012). Segregation of axonal and
Homeostasis of Neuronal Excitability 65

somatic activity during fast network oscillations. imaging of intracellular Cl() with Cl-Sensor
Science. 336, 14581461. using conventional fluorescence setup. Front
Dyhrfjeld-Johnsen, J., Morgan, R.J., and Soltesz, I. Mol. Neurosci.6,7.
(2009). Double trouble? Potential for hyperex- Fritschy, J.M., Harvey, R.J., and Schwarz, G. (2008).
citability following both channelopathic up- Gephyrin:Where do we stand, where do we go?
and downregulation of I(h) in epilepsy. Front Trends Neurosci. 31, 257264.
Neurosci. 3,2533. Garcia-Morales, I., de la Pena, M.P., and Kanner,
Eichler, S.A., Frstera, B., Smolinsky, B., Juttner, R., A.M. (2008). Psychiatric comorbidities in epi-
Lehmann, T.N., Fahling, M., Schwarz, G., lepsy: Identification and treatment. Neurologist
Legendre, P., and Meier, J.C. (2009). Splice- 14, S15S25.
specific roles of glycine receptor 3 in the hippo- George, A.L., Jr. (2004). Inherited channelopathies
campus. Eur. J. Neurosci. 30, 10771091. associated with epilepsy. Epilepsy Curr. 4,6570.
Eichler, S.A., Kirischuk, S., Jttner, R., Schfermeier, Glickfeld, L.L., Roberts, J.D., Somogyi, P., and
P.K., Legendre, P., Lehmann, T.N., Gloveli, T., Scanziani, M. (2009). Interneurons hyperpolar-
Grantyn, R., and Meier, J.C. (2008). Glycinergic ize pyramidal cells along their entire somatoden-
tonic inhibition of hippocampal neurons with dritic axis. Nat. Neurosci. 12,2123.
depolarising GABAergic transmission elicits his- Golowasch, J., Abbott, L.F., and Marder, E. (1999).
topathological signs of temporal lobe epilepsy. J. Activity-dependent regulation of potassium
Cell. Mol. Med. 12, 28482866. currents in an identified neuron of the stoma-
Eichler, S.A., and Meier, J.C. (2008). E-I balance and togastric ganglion of the crab Cancer borealis. J.
human diseasesfrom molecules to networking. Neurosci. 19,RC33.
Front. Mol. Neurosci.1,2. Gonzalez, M.I. (2013). The possible role of GABAA
Essrich, C., Lorez, M., Benson, J.A., Fritschy, J.M., and receptors and gephyrin in epileptogenesis. Front
Luscher, B. (1998). Postsynaptic clustering of major Cell Neurosci. 7,113.
GABAA receptor subtypes requires the gamma 2 Gonzalez, M.I., Cruz Del, A.Y., and Brooks-Kayal,
subunit and gephyrin. Nat. Neurosci. 1, 563571. A. (2013). Down-regulation of gephyrin and
Fan, Y., Fricker, D., Brager, D.H., Chen, X., Lu, GABAA receptor subunits during epileptogen-
H.C., Chitwood, R.A., and Johnston, D. (2005). esis in the CA1 region of hippocampus. Epilepsia
Activity-dependent decrease of excitability in rat 54, 616624.
hippocampal neurons through increases in I(h). Groth, R.D., Lindskog, M., Thiagarajan, T.C.,
Nat. Neurosci. 8, 15421551. Li, L., and Tsien, R.W. (2011). Beta Ca2+/
Fang, M., Shen, L., Yin, H., Pan, Y.M., Wang, L., CaM-dependent kinase type II triggers upregu-
Chen, D., Xi, Z.Q., Xiao, Z., and Wang, X.F. lation of GluA1 to coordinate adaptation to syn-
(2011). Down-regulation of gephyrin in tempo- aptic inactivity in hippocampal neurons. Proc.
ral lobe epilepsy neurons in humans and a rat Natl. Acad. Sci. U.S. A. 108, 828833.
model. Synapse 65, 10061014. Grubb, M.S., and Burrone, J. (2010).
Fassio, A., Raimondi, A., Lignani, G., Benfenati, F., Activity-dependent relocation of the axon initial
and Baldelli, P. (2011). Synapsins: From syn- segment fine-tunes neuronal excitability. Nature
apse to network hyperexcitability and epilepsy. 465, 10701074.
Semin. Cell Dev. Biol. 22, 408415. Gu, Y., Barry, J., McDougel, R., Terman, D., and Gu,
Frstera, B., Belaidi, A.A., Jttner, R., Bernert, C., C. (2012). Alternative splicing regulates kv3.1
Tsokos, M., Lehmann, T.N., Horn, P., Dehnicke, C., polarized targeting to adjust maximal spiking
Schwarz, G., and Meier, J.C. (2010). Irregular frequency. J. Biol. Chem. 287, 17551769.
RNA splicing curtails postsynaptic gephyrin in Gustafsson, B., and Wigstrom, H. (1983).
the cornu ammonis of patients with epilepsy. Hyperpolarization following long-lasting tetanic
Brain 133, 37783794. activation of hippocampal pyramidal cells. Brain
Franklin, J.L., Fickbohm, D.J., and Willard, A.L. Res. 275, 159163.
(1992). Long-term regulation of neuronal cal- Gutierrez, A., Khan, Z.U., and De Blas, A.L. (1994).
cium currents by prolonged changes of mem- Immunocytochemical localization of gamma
brane potential. J. Neurosci. 12, 17261735. 2 short and gamma 2 long subunits of the
Frick, A., Magee, J., and Johnston, D. (2004). LTP is GABAA receptor in the rat brain. J. Neurosci. 14,
accompanied by an enhanced local excitability 71687179.
of pyramidal neuron dendrites. Nat. Neurosci. 7, Gutman, G.A., Chandy, K.G., Grissmer, S.,
126135. Lazdunski, M., McKinnon, D., Pardo, L.A.,
Friedel, P., Bregestovski, P., and Medina, I. Robertson, G.A., Rudy, B., Sanguinetti, M.C.,
(2013). Improved method for efficient Stuhmer, W., and Wang, X. (2005). International
66 Part I: Homeostatic Regulators

Union of Pharmacology. LIII. Nomenclature and Modulation of neuronal activity by phosphory-


molecular relationships of voltage-gated potas- lation of the K-Cl cotransporter KCC2. Trends
sium channels. Pharmacol. Rev. 57, 473508. Neurosci. 36(12), 726737.
Haussler, U., Bielefeld, L., Froriep, U.P., Wolfart, J., Kahle, K.T., Rinehart, J., and Lifton, R.P. (2010).
and Haas, C.A. (2012). Septotemporal position Phosphoregulation of the Na-K-2Cl and K-Cl
in the hippocampal formation determines epi- cotransporters by the WNK kinases. Biochim.
leptic and neurogenic activity in temporal lobe Biophys. Acta 1802, 11501158.
epilepsy. Cereb. Cortex. 22,2636. Kahle, K.T., Staley, K.J., Nahed, B.V., Gamba, G.,
Hedrich, R. (2012). Ion channels in plants. Physiol Hebert, S.C., Lifton, R.P., and Mount, D.B.
Rev. 92, 17771811. (2008). Roles of the cation-chloride cotrans-
Henneberger, C., Jttner, R., Schmidt, S.A., Walter, J., porters in neurological disease. Nat. Clin. Pract.
Meier, J.C., Rothe, T., and Grantyn, R. (2005). Neurol. 4, 490503.
GluR- and TrkB-mediated maturation of GABA Kaidanovich-Beilin, O., Beaulieu, J.M., Jope, R.S.,
receptor function during the period of eye open- and Woodgett, J.R. (2012). Neurological func-
ing. Eur. J.Neurosci. 21, 431440. tions of the masterswitch protein kinaseGsk-3.
Herweg, J., and Schwarz, G. (2012). Splice-specific Front Mol. Neurosci.5,48.
glycine receptor binding, folding, and phosphor- Kaila, K. (1994). Ionic basis of GABAA receptor
ylation of the scaffolding protein gephyrin. J Biol. channel function in the nervous system. Prog.
Chem. 287, 1264512656. Neurobiol. 42, 489537.
Hille, B. (2001). Ion channels of excitable membranes. Kaila, K., Pasternack, M., Saarikoski, J., and Voipio, J.
(Sunderland, MA:Sinauer Associates,Inc.). (1989). Influence of GABA-gated bicarbonate
Hoffman, D.A., Magee, J.C., Colbert, C.M., and conductance on potential, current and intracel-
Johnston, D. (1997). K+ channel regulation of lular chloride in crayfish muscle fibres. J Physiol
signal propagation in dendrites of hippocampal 416, 161181.
pyramidal neurons. Nature 387, 869875. Kaila, K., and Voipio, J. (1987). Postsynaptic fall in
Hsu, D. (2007). The dentate gyrus as a filter or intracellular pH induced by GABA-activated
gate:Alook back and a look ahead. Prog. Brain bicarbonate conductance. Nature 330,
Res. 163, 601613. 163165.
Hbner, C.A., Stein, V., Hermans-Borgmeyer, I., Karmarkar, U.R., and Buonomano, D.V. (2006).
Meyer, T., Ballanyi, K., and Jentsch, T.J. (2001). Different forms of homeostatic plasticity are
Disruption of KCC2 reveals an essential role of engaged with distinct temporal profiles. Eur.
K-Cl cotransport already in early synaptic inhi- J.Neurosci. 23, 15751584.
bition. Neuron 30, 515524. Kawasaki, B.T., Hoffman, K.B., Yamamoto, R.S.,
Huntsman, M.M., Tran, B.V., Potkin, S.G., Bunney, and Bahr, B.A. (1997). Variants of the recep-
W.E.J., and Jones, E.G. (1998). Altered ratios of tor/channel clustering molecule gephyrin in
alternatively spliced long and short gamma2 brain: Distinct distribution patterns, develop-
subunit mRNAs of the gamma-amino butyrate mental profiles, and proteolytic cleavage by cal-
type A receptor in prefrontal cortex of schizo- pain. J Neurosci. Res. 49, 381388.
phrenics. Proc. Natl. Acad. Sci. U. S. A. 95, Khirug, S., Ahmad, F., Puskarjov, M., Afzalov, R.,
1506615071. Kaila, K., and Blaesse, P. (2010). A single sei-
Hyun, J.H., Eom, K., Lee, K.H., Ho, W.K., and Lee, zure episode leads to rapid functional activa-
S.H. (2013). Activity-dependent downregulation tion of KCC2 in the neonatal rat hippocampus.
of D-type K+ channel subunit Kv1.2 in rat hippo- J Neurosci. 30, 1202812035.
campal CA3 pyramidal neurons. J. Physiol. 591, Khirug, S., Yamada, J., Afzalov, R., Voipio, J.,
55255540. Khiroug, L., and Kaila, K. (2008). GABAergic
Isokawa, M. (1996). Decreased time constant depolarization of the axon initial segment in
in hippocampal dentate granule cells in cortical principal neurons is caused by the
pilocarpine-treated rats with progressive seizure Na-K-2Cl cotransporter NKCC1. J Neurosci. 28,
frequencies. Brain Res. 718, 169175. 46354639.
Jan, L.Y., and Jan, Y.N. (1994). Potassium channels Kim, J., Jung, S.C., Clemens, A.M., Petralia, R.S., and
and their evolving gates. Nature 371, 119122. Hoffman, D.A. (2007). Regulation of dendritic
Jonas, P., Bischofberger, J., and Sandkuhler, J. (1998). excitability by activity-dependent trafficking of
Corelease of two fast neurotransmitters at a cen- the A-type K+ channel subunit Kv4.2 in hippo-
tral synapse. Science 281, 419424. campal neurons. Neuron. 54, 933947.
Kahle, K.T., Deeb, T.Z., Puskarjov, M., Silayeva, L., Kim, U., and McCormick, D.A. (1998). Functional and
Liang, B., Kaila, K., and Moss, S.J. (2013). ionic properties of a slow afterhyperpolarization
Homeostasis of Neuronal Excitability 67

in ferret perigeniculate neurons in vitro. J. Le-Corronc, H., Rigo, J.M., Branchereau, P., and
Neurophysiol. 80, 12221235. Legendre, P. (2011). GABA(A) receptor and gly-
Kirchheim, F., Tinnes, S., Haas, C.A., Stegen, M., and cine receptor activation by paracrine/autocrine
Wolfart, J. (2013). Regulation of action potential release of endogenous agonists:More than a sim-
delays via voltage-gated potassium Kv1.1 chan- ple communication pathway. Mol. Neurobiol.
nels in dentate granule cells during hippocampal 44,2852.
epilepsy. Front. Cell. Neurosci. 7,248. Lee, H.H., Deeb, T.Z., Walker, J.A., Davies, P.A.,
Kirsch, J., Wolters, I., Triller, A., and Betz, H. (1993). and Moss, S.J. (2011). NMDA receptor activ-
Gephyrin antisense oligonucleotides prevent ity downregulates KCC2 resulting in depolar-
glycine receptor clustering in spinal neurons. izing GABAA receptor-mediated currents. Nat.
Nature 366, 745748. Neurosci. 14, 736743.
Kittler, J.T., and Moss, S.J. (2003). Modulation of Lee, H.H., Jurd, R., and Moss, S.J. (2010). Tyrosine
GABA(A) receptor activity by phosphoryla- phosphorylation regulates the membrane traf-
tion and receptor trafficking: Implications for ficking of the potassium chloride co-transporter
the efficacy of synaptic inhibition. Curr. Opin. KCC2. Mol. Cell Neurosci. 45, 173179.
Neurobiol. 13, 341347. Lee, H.H., Walker, J.A., Williams, J.R., Goodier, R.J.,
Kletke, O., Sergeeva, O.A., Lorenz, P., Oberland, Payne, J.A., and Moss, S.J. (2007). Direct protein
S., Meier, J.C., Hatt, H., and Gisselmann, G. kinase C-dependent phosphorylation regulates
(2013). New insights in endogenous modula- the cell surface stability and activity of the potas-
tion of ligand-gated ion channels: Histamine is sium chloride cotransporter KCC2. J Biol. Chem.
an inverse agonist at strychnine sensitive glycine 282, 2977729784.
receptors. Eur. J.Pharmacol. 710,5966. Legendre, P., Frstera, B., Jttner, R., and Meier,
Kobayashi, M., Wen, X., and Buckmaster, P.S. (2003). J.C. (2009). Glycine receptors caught between
Reduced inhibition and increased output of layer genome and proteomeFunctional implica-
II neurons in the medial entorhinal cortex in a tions of RNA editing and splicing. Front. Mol.
model of temporal lobe epilepsy. J. Neurosci. 23, Neurosci.2,23.
84718479. Leite, J.P., Neder, L., Arisi, G.M., Carlotti, C.G., Jr.,
Koch, D., Spiwoks-Becker, I., Sabanov, V., Sinning, A., Assirati, J.A., and Moreira, J.E. (2005). Plasticity,
Dugladze, T., Stellmacher, A., Ahuja, R., synaptic strength, and epilepsy: What can
Grimm, J., Schuler, S., Muller, A., Angenstein, we learn from ultrastructural data? Epilepsia
F., Ahmed, T., Diesler, A., Moser, M., Tom, 46(Suppl 5), 134141.
D.S., Spessert, R., Boeckers, T.M., Fassler, R., LeMasson, G., Marder, E., and Abbott, L.F. (1993).
Hubner, C.A., Balschun, D., Gloveli, T., Kessels, Activity-dependent regulation of conductances
M.M., and Qualmann, B. (2011). Proper syn- in model neurons. Science 259, 19151917.
aptic vesicle formation and neuronal network Leslie, K.R., Nelson, S.B., and Turrigiano, G.G.
activity critically rely on syndapin I. EMBO J. (2001). Postsynaptic depolarization scales quan-
30, 49554969. tal amplitude in cortical pyramidal neurons. J.
Krishek, B.J., Xie, X., Blackstone, C., Huganir, R.L., Neurosci. 21,RC170.
Moss, S.J., and Smart, T.G. (1994). Regulation of Lionel, A.C., Vaags, A.K., Sato, D., Gazzellone, M.J.,
GABAA receptor function by protein kinase C Mitchell, E.B., Chen, H.Y., Costain, G., Walker,
phosphorylation. Neuron 12, 10811095. S., Egger, G., Thiruvahindrapuram, B., Merico,
Kuba, H., Oichi, Y., and Ohmori, H. (2010). D., Prasad, A., Anagnostou, E., Fombonne, E.,
Presynaptic activity regulates Na(+) channel dis- Zwaigenbaum, L., Roberts, W., Szatmari, P.,
tribution at the axon initial segment. Nature 465, Fernandez, B.A., Georgieva, L., Brzustowicz,
10751078. L.M., Roetzer, K., Kaschnitz, W., Vincent, J.B.,
Kuhse, J., Kuryatov, A., Maulet, Y., Malosio, M.L., Windpassinger, C., Marshall, C.R., Trifiletti,
Schmieden, V., and Betz, H. (1991). Alternative R.R., Kirmani, S., Kirov, G., Petek, E., Hodge,
splicing generates two isoforms of the alpha 2 J.C., Bassett, A.S., and Scherer, S.W. (2013). Rare
subunit of the inhibitory glycine receptor. FEBS exonic deletions implicate the synaptic organizer
Lett. 283,7377. Gephyrin (GPHN) in risk for autism, schizo-
Lardi-Studler, B., Smolinsky, B., Petitjean, C.M., phrenia and seizures. Hum. Mol. Genet. 22,
Koenig, F., Sidler, C., Meier, J.C., Fritschy, J.M., 20552066.
and Schwarz, G. (2007). Vertebrate-specific Lissin, D.V., Gomperts, S.N., Carroll, R.C., Christine,
sequences in the gephyrin E-domain regulate C.W., Kalman, D., Kitamura, M., Hardy, S.,
cytosolic aggregation and postsynaptic cluster- Nicoll, R.A., Malenka, R.C., and von Zastrow,
ing. J. Cell Sci. 120, 13711382. M. (1998). Activity differentially regulates
68 Part I: Homeostatic Regulators

the surface expression of synaptic AMPA and Markram, H., Lubke, J., Frotscher, M., and Sakmann,
NMDA glutamate receptors. Proc. Natl. Acad. B. (1997). Regulation of synaptic efficacy by coin-
Sci. U.S. A. 95, 70977102. cidence of postsynaptic APs and EPSPs. Science.
Llinas, R.R. (1988). The intrinsic electrophysiological 275, 213215.
properties of mammalian neurons:Insights into McCormick, D.A., and Contreras, D. (2001). On the
central nervous system function. Science 242, cellular and network bases of epileptic seizures.
16541664. Annu. Rev. Physiol. 63, 815846.
Lugo, J.N., Barnwell, L.F., Ren, Y., Lee, W.L., McKinley, D.D., Lennon, D.J., and Carter, D.B.
Johnston, L.D., Kim, R., Hrachovy, R.A., Sweatt, (1995). Cloning, sequence analysis and expres-
J.D., and Anderson, A.E. (2008). Altered phos- sion of two forms of mRNA coding for the
phorylation and localization of the A-type chan- human beta 2 subunit of the GABAA receptor.
nel, Kv4.2 in status epilepticus. J. Neurochem. Brain Res. Mol. Brain Res. 28, 175179.
106, 19291940. Meier, J., and Grantyn, R. (2004a). A gephyrin-related
Luscher, B., Fuchs, T., and Kilpatrick, C.L. (2011). mechanism restraining glycine receptor anchor-
GABA(A) receptor trafficking-mediated plastic- ing at GABAergic synapses. J. Neurosci. 24,
ity of inhibitory synapses. Neuron 70, 385409. 13981405.
MacLean, J.N., Zhang, Y., Goeritz, M.L., Casey, R., Meier, J., and Grantyn, R. (2004b). Preferential accu-
Oliva, R., Guckenheimer, J., and Harris-Warrick, mulation of GABA(A) receptor 2L, not 2S,
R.M. (2005). Activity-independent coregulation cytoplasmic loops at rat spinal cord inhibitory
of IA and Ih in rhythmically active neurons. J. synapses. J. Physiol. 559, 355365.
Neurophysiol. 94, 36013617. Meier, J., Vannier, C., Serge, A., Triller, A., and
MacLean, J.N., Zhang, Y., Johnson, B.R., Choquet, D. (2001). Fast and reversible trapping
and Harris-Warrick, R.M. (2003). of surface glycine receptors by gephyrin. Nat.
Activity-independent homeostasis in rhythmi- Neurosci. 4, 253260.
cally active neurons. Neuron 37, 109120. Meier, J.C., Henneberger, C., Melnick, I., Racca, C.,
Magee, J.C. (1998). Dendritic hyperpolarization- Harvey, R.J., Heinemann, U., Schmieden, V., and
activated currents modify the integrative prop- Grantyn, R. (2005). RNA editing produces gly-
erties of hippocampal CA1 pyramidal neurons. cine receptor 3P185L resulting in high agonist
J. Neurosci. 18, 76137624. potency. Nat. Neurosci. 8, 736744.
Magloczky, Z., and Freund, T.F. (2005). Impaired and Meier, J.C., Semtner, M., Winkelmann, A., and
repaired inhibitory circuits in the epileptic human Wolfart, J. (2014). Presynaptic mechanisms of
hippocampus. Trends Neurosci. 28, 334340. neuronal plasticity and their role in epilepsy.
Main, M.J., Cryan, J.E., Dupere, J.R., Cox, B., Clare, J.J., Front. Cell. Neurosci. 8,164.
and Burbidge, S.A. (2000). Modulation of KCNQ2/3 Miller, P.S., Harvey, R.J., and Smart, T.G. (2004).
potassium channels by the novel anticonvulsant Differential agonist sensitivity of glycine receptor
retigabine. Mol. Pharmacol. 58, 253262. alpha2 subunit splice variants. Br. J.Pharmacol.
Malosio, M.L., Grenningloh, G., Kuhse, J., 143,1926.
Schmieden, V., Schmitt, B., Prior, P., and Betz, H. Moody, W.J., and Bosma, M.M. (2005). Ion chan-
(1991). Alternative splicing generates two vari- nel development, spontaneous activity, and
ants of the alpha 1 subunit of the inhibitory gly- activity-dependent development in nerve and
cine receptor. J. Biol. Chem. 266, 20482053. muscle cells. Physiol Rev. 85, 883941.
Marder, E., Abbott, L.F., Turrigiano, G.G., Liu, Z., Moss, S.J., Doherty, C.A., and Huganir, R.L. (1992).
and Golowasch, J. (1996). Memory from the Identification of the cAMP-dependent protein
dynamics of intrinsic membrane currents. Proc. kinase and protein kinase C phosphorylation
Natl. Acad. Sci. U.S. A. 93, 1348113486. sites within the major intracellular domains of
Marder, E., and Goaillard, J.M. (2006). Variability, the beta 1, gamma 2S, and gamma 2L subunits of
compensation and homeostasis in neuron and the gamma-aminobutyric acid type Areceptor. J.
network function. Nat. Rev. Neurosci. 7, 563574. Biol. Chem. 267, 1447014476.
Marder, E., and Taylor, A.L. (2011). Multiple models Mozzachiodi, R., and Byrne, J.H. (2010). More than
to capture the variability in biological neurons synaptic plasticity: Role of nonsynaptic plastic-
and networks. Nat. Neurosci. 14, 133138. ity in learning and memory. Trends Neurosci.
Markkanen, M., Karhunen, T., Llano, O., Ludwig, 33,1726.
A., Rivera, C., Uvarov, P., and Airaksinen, M.S. Muir, J., Arancibia-Carcamo, I.L., Macaskill, A.F.,
(2014). Distribution of neuronal KCC2a and Smith, K.R., Griffin, L.D., and Kittler, J.T. (2010).
KCC2b isoforms in mouse CNS. J. Comp Neurol. NMDA receptors regulate GABAA receptor
522, 18971914. lateral mobility and clustering at inhibitory
Homeostasis of Neuronal Excitability 69

synapses through serine 327 on the gamma2 Ponce-Coria, J., San-Cristobal, P., Kahle, K.T.,
subunit. Proc. Natl. Acad. Sci. U. S. A 107, Vazquez, N., Pacheco-Alvarez, D., de Los, H.P.,
1667916684. Juarez, P., Munoz, E., Michel, G., Bobadilla,
Narayanan, R., and Johnston, D. (2010). The h current N.A., Gimenez, I., Lifton, R.P., Hebert, S.C.,
is a candidate mechanism for regulating the slid- and Gamba, G. (2008). Regulation of NKCC2
ing modification threshold in a BCM-like synap- by a chloride-sensing mechanism involving the
tic learning rule. J. Neurophysiol. 104, 10201033. WNK3 and SPAK kinases. Proc. Natl. Acad. Sci.
Nikolic, Z., Laube, B., Weber, R.G., Lichter, P., U.S. A 105, 84588463.
Kioschis, P., Poustka, A., Mulhardt, C., and Pratt, K.G., and Aizenman, C.D. (2007). Homeostatic
Becker, C.M. (1998). The human glycine receptor regulation of intrinsic excitability and synaptic
subunit alpha3. Glra3 gene structure, chromo- transmission in a developing visual circuit. J.
somal localization, and functional characteriza- Neurosci. 27, 82688277.
tion of alternative transcripts. J. Biol. Chem. 273, Prinz, A.A., Bucher, D., and Marder, E. (2004).
1970819714. Similar network activity from disparate circuit
Noam, Y., Bernard, C., and Baram, T.Z. (2011). parameters. Nat. Neurosci. 7, 13451352.
Towards an integrated view of HCN channel role Puskarjov, M., Ahmad, F., Kaila, K., and Blaesse, A.
in epilepsy. Curr. Opin. Neurobiol. 21, 873879. (2012). Activity-dependent degradation of
Notelaers, K., Smisdom, N., Rocha, S., Janssen, D., the K-Cl-cotransporter KCC2 is mediated by
Meier, J.C., Rigo, J.M., Hofkens, J., and Ameloot, M. calcium-activated protease calpain. J. Neurosci.
(2012). Ensemble and single particle fluorimet- 32, 1135611364.
ric techniques in concerted action to study the Qian, N., and Sejnowski, T.J. (1990). When is an
diffusion and aggregation of the glycine receptor inhibitory synapse effective? Proc. Natl. Acad.
alpha3 isoforms in the cell plasma membrane. Sci. U.S. A 87, 81458149.
Biochim. Biophys. Acta 1818, 31313140. Rinehart, J., Maksimova, Y.D., Tanis, J.E., Stone, K.L.,
OBrien, R.J., Kamboj, S., Ehlers, M.D., Rosen, Hodson, C.A., Zhang, J., Risinger, M., Pan, W.,
K.R., Fischbach, G.D., and Huganir, R.L. (1998). Wu, D., Colangelo, C.M., Forbush, B., Joiner,
Activity-dependent modulation of synap- C.H., Gulcicek, E.E., Gallagher, P.G., and Lifton,
tic AMPA receptor accumulation. Neuron 21, R.P. (2009). Sites of regulated phosphorylation
10671078. that control K-Cl cotransporter activity. Cell
Ohlson, J., Pedersen, J.S., Haussler, D., and Ohman, M. 138, 525536.
(2007). Editing modifies the GABA(A) receptor Rivera, C., Li, H., Thomas-Crusells, J., Lahtinen, H.,
subunit alpha3. RNA. 13, 698703. Viitanen, T., Nanobashvili, A., Kokaia, Z.,
Okada, M. and Matsuda, H. (2008). Chronic len- Airaksinen, M.S., Voipio, J., Kaila, K., and
tiviral expression of inwardly rectifying K+ Saarma, M. (2002). BDNF-induced TrkB activa-
channels (Kir2.1) reduces neuronal activity and tion down-regulates the K+-Cl- cotransporter
downregulates voltage-gated potassium currents KCC2 and impairs neuronal Cl- extrusion. J. Cell
in hippocampus. Neuroscience 156, 289297. Biol. 159, 747752.
Overstreet-Wadiche, L.S., and Westbrook, G.L. Rivera, C., Voipio, J., and Kaila, K. (2005). Two devel-
(2006). Functional maturation of adult-generated opmental switches in GABAergic signalling:The
granule cells. Hippocampus 16, 208215. K+-Cl- cotransporter KCC2 and carbonic anhy-
Padmanabhan, K., and Urban, N.N. (2010). Intrinsic drase CAVII. J. Physiol 562,2736.
biophysical diversity decorrelates neuronal fir- Rivera, C., Voipio, J., Payne, J.A., Ruusuvuori, E.,
ing while increasing information content. Nat. Lahtinen, H., Lamsa, K., Pirvola, U., Saarma, M.,
Neurosci. 13, 12761282. and Kaila, K. (1999). The K+/Cl- co-transporter
Pasternack, M., Voipio, J., and Kaila, K. (1993). KCC2 renders GABA hyperpolarizing during
Intracellular carbonic anhydrase activity and neuronal maturation. Nature 397, 251255.
its role in GABA-induced acidosis in isolated rat Rivera, C., Voipio, J., Thomas-Crusells, J., Li, H.,
hippocampal pyramidal neurones. Acta Physiol Emri, Z., Sipila, S., Payne, J.A., Minichiello, L.,
Scand. 148, 229231. Saarma, M., and Kaila, K. (2004). Mechanism
Plotkin, M.D., Snyder, E.Y., Hebert, S.C., and of activity-dependent downregulation of the
Delpire, E. (1997). Expression of the Na-K-2Cl neuron-specific K-Cl cotransporter KCC2. J.
cotransporter is developmentally regulated in Neurosci. 24, 46834691.
postnatal rat brains:Apossible mechanism under- Rogawski, M.A., and Loscher, W. (2004). The neu-
lying GABAs excitatory role in immature brain. robiology of antiepileptic drugs. Nat. Rev.
J Neurobiol. 33, 781795. Neurosci. 5, 553564.
70 Part I: Homeostatic Regulators

Roth-Alpermann, C., Morris, R.G., Korte, M., and Smart, S.L., Lopantsev, V., Zhang, C.L., Robbins,
Bonhoeffer, T. (2006). Homeostatic shutdown C.A., Wang, H., Chiu, S.Y., Schwartzkroin, P.A.,
of long-term potentiation in the adult hip- Messing, A., and Tempel, B.L. (1998). Deletion of
pocampus. Proc. Natl. Acad. Sci. U. S. A. 103, the K(V)1.1 potassium channel causes epilepsy
1103911044. in mice. Neuron. 20, 809819.
Ruiz, A., Campanac, E., Scott, R.S., Rusakov, D.A., Smolinsky, B., Eichler, S.A., Buchmeier, S., Meier,
and Kullmann, D.M. (2010). Presynaptic GABAA J.C., and Schwarz, G. (2008). Splice-specific
receptors enhance transmission and LTP induc- functions of gephyrin in molybdenum cofactor
tion at hippocampal mossy fiber synapses. Nat. biosynthesis. J. Biol. Chem. 283, 1737017379.
Neurosci. 13, 431438. Specht, C.G., Grunewald, N., Pascual, O., Rostgaard,
Ruusuvuori, E., Li, H., Huttu, K., Palva, J.M., N., Schwarz, G., and Triller, A. (2011). Regulation
Smirnov, S., Rivera, C., Kaila, K., and Voipio, J. of glycine receptor diffusion properties and
(2004). Carbonic anhydrase isoform VII acts as a gephyrin interactions by protein kinase C.
molecular switch in the development of synchro- EMBO J 30, 38423853.
nous gamma-frequency firing of hippocampal Spitzer, N.C. (1999). New dimensions of neuronal
CA1 pyramidal cells. J Neurosci. 24, 26992707. plasticity. Nat. Neurosci. 2, 489491.
Sanchez-Vives, M.V., Nowak, L.G., and McCormick, Spruston, N. (2008). Pyramidal neurons: Dendritic
D.A. (2000). Cellular mechanisms of long-lasting structure and synaptic integration. Nat. Rev.
adaptation in visual cortical neurons in vitro. J. Neurosci. 9, 206221.
Neurosci. 20, 42864299. Staley, K.J., and Proctor, W.R. (1999). Modulation of
Scholz, K.P., and Byrne, J.H. (1987). Long-term sen- mammalian dendritic GABA(A) receptor func-
sitization in Aplysia: Biophysical correlates in tion by the kinetics of Cl and HCO3 transport.
tail sensory neurons. Science 235, 685687. J Physiol. 519(Pt 3), 693712.
Schuchmann, S., Schmitz, D., Rivera, C., Staley, K.J., Soldo, B.L., and Proctor, W.R. (1995).
Vanhatalo, S., Salmen, B., Mackie, K., Ionic mechanisms of neuronal excitation by
Sipila, S.T., Voipio, J., and Kaila, K. (2006). inhibitory GABAA receptors. Science 269,
Experimental febrile seizures are precipitated 977981.
by a hyperthermia-induced respiratory alkalo- Stegen, M., Kirchheim, F., Hanuschkin, A.,
sis. Nat. Med. 12, 817823. Staszewski, O., Veh, R.W., and Wolfart, J. (2012).
Schulz, R., Kirschstein, T., Brehme, H., Porath, K., Adaptive intrinsic plasticity in human dentate
Mikkat, U., and Kohling, R. (2012). Network gyrus granule cells during temporal lobe epi-
excitability in a model of chronic tempo- lepsy. Cereb. Cortex. 22, 20872101.
ral lobe epilepsy critically depends on SK Stegen, M., Young, C.C., Haas, C.A., Zentner, J., and
channel-mediated AHP currents. Neurobiol. Wolfart, J. (2009). Increased leak conductance
Dis. 45, 337347. in dentate gyrus granule cells of temporal lobe
Schwarz, G., Mendel, R.R., and Ribbe, M.W. (2009). epilepsy patients with Ammons horn sclerosis.
Molybdenum cofactors, enzymes and pathways. Epilepsia. 50, 646653.
Nature 460, 839847. Streit, A.K., Derst, C., Wegner, S., Heinemann, U.,
Schwindt, P.C., Spain, W.J., and Crill, W.E. (1989). Zahn, R.K., and Decher, N. (2011). RNA editing
Long-lasting reduction of excitability by a of Kv1.1 channels may account for reduced icto-
sodium-dependent potassium current in cat neo- genic potential of 4-aminopyridine in chronic
cortical neurons. J. Neurophysiol. 61, 233244. epileptic rats. Epilepsia 52, 645648.
Seki, M., Kobayashi, C., Takahashi, N., Matsuki, Su, H., Sochivko, D., Becker, A., Chen, J., Jiang, Y.,
N., and Ikegaya, Y. (2012). Synchronized spike Yaari, Y., and Beck, H. (2002). Upregulation of a
waves in immature dentate gyrus networks. Eur. T-type Ca2+ channel causes a long-lasting modi-
J.Neurosci. 35, 673681. fication of neuronal firing mode after status epi-
Siegel, M., Marder, E., and Abbott, L.F. (1994). lepticus. J. Neurosci. 22, 36453655.
Activity-dependent current distributions in Tanner, G.R., Lutas, A., Martinez-Francois, J.R.,
model neurons. Proc. Natl. Acad. Sci. U.S. A. 91, and Yellen, G. (2011). Single K ATP channel
1130811312. opening in response to action potential firing in
Signorini, S., Liao, Y.J., Duncan, S.A., Jan, L.Y., and mouse dentate granule neurons. J. Neurosci. 31,
Stoffel, M. (1997). Normal cerebellar develop- 86898696.
ment but susceptibility to seizures in mice lack- Thoby-Brisson, M., and Simmers, J. (1998).
ing G protein-coupled, inwardly rectifying K+ Neuromodulatory inputs maintain expression
channel GIRK2. Proc. Natl. Acad. Sci. U. S. A. of a lobster motor pattern-generating network
94, 923927. in a modulation-dependent state:Evidence from
Homeostasis of Neuronal Excitability 71

long-term decentralization in vitro. J. Neurosci. of gephyrin. Proc. Natl. Acad. Sci. U. S. A 108,
18, 22122225. 379384.
Thompson, A.J., Lester, H.A., and Lummis, S.C. Tyzio, R., Minlebaev, M., Rheims, S., Ivanov, A.,
(2010). The structural basis of function in Jorquera, I., Holmes, G.L., Zilberter, Y., Ben-Ari,
Cys-loop receptors. Q. Rev. Biophys. 43, 449499. Y., and Khazipov, R. (2008). Postnatal changes
Tretter, V., Mukherjee, J., Maric, H.M., Schindelin, in somatic gamma-aminobutyric acid signal-
H., Sieghart, W., and Moss, S.J. (2012). Gephyrin, ling in the rat hippocampus. Eur. J Neurosci. 27,
the enigmatic organizer at GABAergic synapses. 25152528.
Front Cell Neurosci.6,23. Uvarov, P., Ludwig, A., Markkanen, M., Pruunsild,
Tsaur, M.L., Sheng, M., Lowenstein, D.H., Jan, P., Kaila, K., Delpire, E., Timmusk, T., Rivera, C.,
Y.N., and Jan, L.Y. (1992). Differential expres- and Airaksinen, M.S. (2007). A novel N-terminal
sion of K+ channel mRNAs in the rat brain and isoform of the neuron-specific K-Cl cotrans-
down-regulation in the hippocampus following porter KCC2. J Biol. Chem. 282, 3057030576.
seizures. Neuron. 8, 10551067. Uvarov, P., Ludwig, A., Markkanen, M., Soni, S.,
Turecek, R., and Trussell, L.O. (2001). Presynaptic Hubner, C.A., Rivera, C., and Airaksinen, M.S.
glycine receptors enhance transmitter release (2009). Coexpression and heteromerization of
at a mammalian central synapse. Nature 411, two neuronal K-Cl cotransporter isoforms in
587590. neonatal brain. J Biol. Chem. 284, 1369613704.
Turrigiano, G. (2011). Too many cooks? Intrinsic and Vacher, H., and Trimmer, J.S. (2011). Diverse roles for
synaptic homeostatic mechanisms in cortical cir- auxiliary subunits in phosphorylation-dependent
cuit refinement. Annu. Rev. Neurosci. 34, 89103. regulation of mammalian brain voltage-gated
Turrigiano, G., Abbott, L.F., and Marder, E. (1994). potassium channels. Pflugers Arch. 462, 631643.
Activity-dependent changes in the intrinsic van Welie, I., van Hooft, J.A., and Wadman, W.J.
properties of cultured neurons. Science 264, (2004). Homeostatic scaling of neuronal excit-
974977. ability by synaptic modulation of somatic
Turrigiano, G., LeMasson, G., and Marder, E. (1995). hyperpolarization-activated Ih channels. Proc.
Selective regulation of current densities under- Natl. Acad. Sci. U.S. A. 101, 51235128.
lies spontaneous changes in the activity of cul- Vibat, C.R., Holland, M.J., Kang, J.J., Putney, L.K.,
tured neurons. J. Neurosci. 15, 36403652. and ODonnell, M.E. (2001). Quantitation of
Turrigiano, G.G. (1999). Homeostatic plasticity in Na+-K+-2Cl- cotransport splice variants in
neuronal networks:The more things change, the human tissues using kinetic polymerase chain
more they stay the same. Trends Neurosci. 22, reaction. Anal. Biochem. 298, 218230.
221227. Viitanen, T., Ruusuvuori, E., Kaila, K., and Voipio, J.
Turrigiano, G.G., Leslie, K.R., Desai, N.S., (2010). The K+-Cl cotransporter KCC2 promotes
Rutherford, L.C., and Nelson, S.B. (1998). GABAergic excitation in the mature rat hippo-
Activity-dependent scaling of quantal amplitude campus. J Physiol 588, 15271540.
in neocortical neurons. Nature 391, 892896. Vithlani, M., and Moss, S.J. (2009). The role of
Turrigiano, G.G., and Nelson, S.B. (2000). Hebb and GABAAR phosphorylation in the construc-
homeostasis in neuronal plasticity. Curr. Opin. tion of inhibitory synapses and the efficacy of
Neurobiol. 10, 358364. neuronal inhibition. Biochem. Soc. Trans. 37,
Turrigiano, G.G., and Nelson, S.B. (2004). 13551358.
Homeostatic plasticity in the developing nervous Voipio, J., and Kaila, K. (2000). GABAergic excita-
system. Nat. Rev. Neurosci. 5, 97107. tion and K(+)-mediated volume transmission in
Tyagarajan, S.K., Ghosh, H., Yevenes, G.E., Imanishi, the hippocampus. Prog. Brain Res. 125, 329338.
S.Y., Zeilhofer, H.U., Gerrits, B., and Fritschy, J.M. Wake, H., Watanabe, M., Moorhouse, A.J.,
(2013). Extracellular signal-regulated kinase and Kanematsu, T., Horibe, S., Matsukawa, N., Asai,
glycogen synthase kinase 3beta regulate gephy- K., Ojika, K., Hirata, M., and Nabekura, J. (2007).
rin postsynaptic aggregation and GABAergic Early changes in KCC2 phosphorylation in
synaptic function in a calpain-dependent mech- response to neuronal stress result in functional
anism. J Biol. Chem. 288, 96349647. downregulation. J. Neurosci. 27, 16421650.
Tyagarajan, S.K., Ghosh, H., Yevenes, G.E., Wang, Z., Xu, N.L., Wu, C.P., Duan, S., and Poo,
Nikonenko, I., Ebeling, C., Schwerdel, C., M.M. (2003). Bidirectional changes in spatial
Sidler, C., Zeilhofer, H.U., Gerrits, B., Muller, dendritic integration accompanying long-term
D., and Fritschy, J.M. (2011). Regulation of synaptic modifications. Neuron 37, 463472.
GABAergic synapse formation and plastic- Waseem, T., Mukhtarov, M., Buldakova, S., Medina,
ity by GSK3{beta}-dependent phosphorylation I., and Bregestovski, P. (2010). Genetically
72 Part I: Homeostatic Regulators

encoded Cl-Sensor as a tool for monitoring of ion channel changes in temporal lobe epilepsy
Cl-dependent processes in small neuronal com- and their antiepileptic potential. Front Physiol
partments. J Neurosci. Methods 193,1423. 6,168.
Waseem, T.V., and Fedorovich, S.V. (2010). Presynaptic Xu, B., English, J.M., Wilsbacher, J.L., Stippec, S.,
glycine receptors influence plasma membrane Goldsmith, E.J., and Cobb, M.H. (2000). WNK1,
potential and glutamate release. Neurochem. Res. a novel mammalian serine/threonine protein
35, 11881195. kinase lacking the catalytic lysine in subdomain
Watanabe, M., Wake, H., Moorhouse, A.J., and II. J Biol. Chem. 275, 1679516801.
Nabekura, J. (2009). Clustering of neuronal Xu, J., Kang, N., Jiang, L., Nedergaard, M., and Kang,
K+-Cl- cotransporters in lipid rafts by tyrosine J. (2005). Activity-dependent long-term potentia-
phosphorylation. J Biol. Chem. 284, 2798027988. tion of intrinsic excitability in hippocampal CA1
Weston, A.J., and Baines, R.A. (2007). Translational pyramidal neurons. J. Neurosci. 25, 17501760.
regulation of neuronal electrical properties. Yim, M.Y., Hanuschkin, A., and Wolfart, J. (2015).
Invert. Neurosci. 7,7586. Intrinsic rescaling of granule cells restores pat-
Whiting, P., McKernan, R.M., and Iversen, L.L. tern separation ability of a dentate gyrus net-
(1990). Another mechanism for creating diver- work model during epileptic hyperexcitability.
sity in gamma-aminobutyrate type A recep- Hippocampus 25, 297308.
tors: RNA splicing directs expression of two Young, C.C., Stegen, M., Bernard, R., Muller, M.,
forms of gamma 2 phosphorylation site. Proc. Bischofberger, J., Veh, R.W., Haas, C.A., and
Natl. Acad. Sci. U.S. A. 87, 99669970. Wolfart, J. (2009). Upregulation of inward recti-
Wickenden, A.D. (2002). Potassium channels as fier K+ (Kir2) channels in dentate gyrus granule
anti-epileptic drug targets. Neuropharmacology cells in temporal lobe epilepsy. J. Physiol. 587,
43, 10551060. 42134233.
Wiegert, J.S., and Bading, H. (2011). Zhang, W., and Linden, D.J. (2003). The other side of
Activity-dependent calcium signaling and the engram:Experience-driven changes in neu-
ERK-MAP kinases in neurons: A link to struc- ronal intrinsic excitability. Nat. Rev. Neurosci. 4,
tural plasticity of the nucleus and gene transcrip- 885900.
tion regulation. Cell Calcium. 49, 296305. Zhao, C., Xu, Z., Chen, J., Yu, Z., Tong, K.L., Lo,
Winkelmann, A., Maggio, N., Eller, J., Caliskan, G., W.S., Pun, F.W., Ng, S.K., Tsang, S.Y., and Xue,
Semtner, M., Hussler, U., Jttner, R., Dugladze, H. (2006). Two isoforms of GABA(A) recep-
T., Smolinsky, B., Kowalczyk, S., Chronowska, tor beta2 subunit with different electrophysi-
E., Schwarz, G., Rathjen, F.G., Rechavi, G., Haas, ological properties: Differential expression and
C.A., Kulik, A., Gloveli, T., Heinemann, U., and genotypical correlations in schizophrenia. Mol.
Meier, J.C. (2014). Changes in neural network Psychiatry 11, 10921105.
homeostasis trigger neuropsychiatric symptoms. Zhao, C., Xu, Z., Wang, F., Chen, J., Ng, S.K., Wong,
J. Clin. Invest. 124, 696711. P.W., Yu, Z., Pun, F.W., Ren, L., Lo, W.S., Tsang,
Wittner, L., Huberfeld, G., Clemenceau, S., Eross, S.Y., and Xue, H. (2009). Alternative-splicing in
L., Dezamis, E., Entz, L., Ulbert, I., Baulac, M., the exon-10 region of GABA(A) receptor beta(2)
Freund, T.F., Magloczky, Z., and Miles, R. (2009). subunit gene: Relationships between novel iso-
The epileptic human hippocampal cornu ammo- forms and psychotic disorders. PLoS. ONE.
nis 2 region generates spontaneous interictal-like 4,e6977.
activity in vitro. Brain 132, 30323046. Zucker, R.S., and Regehr, W.G. (2002). Short-term
Wolfart, J., and Laker, D. (2015). Homeostasis or synaptic plasticity. Annu. Rev. Physiol. 64,
channelopathy? Acquired cell type-specific 355405.
PARTII

Homeostatic Control

Systems, Cells, and Organelles


5
Role ofAstrocytes inSleep and Epilepsy
J E R O M E C L A S A D O N T E A N D P H I L I P G . H AY D O N

INTRODUCTION propagate action potentials along their processes


Glia, glue in Greek, was used for the first time (Sontheimer et al., 1996; Dallerac et al., 2013).
in 1858 by German scientist Rudolf Virchow to Instead, astrocytes exhibit regulated increases
describe the connective tissue that surrounded in intracellular calcium concentration that
neuronal cells in the brain. Increasing evidence are considered a form of astrocyte excitability.
over the past 30 years suggests that astrocytes, Astrocytes express numerous membrane recep-
the most abundant glial cell type, represent much tors for neurotransmitters whose activation leads
more than a supportive tissue. Beyond the buffer- to increases in intracellular concentration of cal-
ing of extracellular potassium and the uptake and cium that can subsequently propagate through
recycling of neurotransmitters and the neuronal the astrocytic network as intercellular calcium
energy supply, astrocytes actively participate in waves. In turn, these calcium increases trigger
the processing of the information in the central the release of neuroactive substances called gli-
nervous system (CNS; for reviews see Halassa otransmitters to regulate synaptic transmission
and Haydon, 2010; Verkhratsky et al., 2011; and neuronal activity. This bidirectional commu-
Parpura et al., 2012, Ben Achour and Pascual, nication between neurons and astrocytes, termed
2012; Pirttimaki and Parri, 2013). Astrocytes dis- gliotransmision, can ultimately regulate vari-
play a highly ramified structure of thin processes ous brain functions such as sleep, breathing,
that contact neurons, blood vessels, and other sensory perception, pain, parturition, lactation,
astrocytes. Structurally, astrocytes are remark- osmoregulation, reproduction, learning, and
ably well organized. They tile in the entire brain memory (for reviews see Halassa and Haydon,
in a contiguous and nonoverlapping manner 2010; Oliet and Bonfardin, 2010; Verkhratsky
(Bushong etal., 2002; Halassa etal., 2007). Each etal., 2011; Clasadonte etal., 2011; Parpura etal.,
astrocyte disposes of its own domain in which 2012; Ben Achour and Pascual, 2012; Pirttimaki
the most distal tips of processes interdigitate and and Parri, 2013; Blutstein and Haydon, 2013).
contact with one another through gap-junction In this chapter we mainly focus on the recent
coupling to form a multicellular network called development of an astrocyte-specific transgenic
the astrocytic network (Giaume etal., 2010). The mouse that allowed the evaluation of the role of
thin branching processes from an individual glial signaling pathways in the control of sleep.
astrocyte can contact 600 dendrites and envelop Besides the role of glial cells in brain physiology,
100,000 synapses in rodents (Bushong et al., astrocytes can also contribute to the pathogen-
2002; Halassa et al., 2007). Diversity and struc- esis of neurological disorders and diseases (for
ture of astrocytes become even more complex in reviews see Sofroniew and Vinters, 2010; Parpura
humans where astrocytes can contact more than etal., 2012). We then discuss the recent evidence
1million synapses (Oberheim etal., 2009). It is that supports the critical role of astrocytes in the
also noteworthy that contact between astrocytes progressive development of one of the most com-
and neurons represent highly dynamic struc- mon brain disorders, epilepsy.
tures, since it has been shown that astrocytic
coverage of the synapse depends on neuronal C A L C I U M E X C I TA B I L I T Y
activity (Hirrlinger et al., 2004; Haber et al., INASTROCYTES
2006; Genoud etal.,2006). Unlike neurons, astrocytes do not generate action
Astrocytes express potassium and sodium potentials and, thus, they were thought to be qui-
channels, but unlike neurons they do not fire or escent. However, astrocytes display spontaneous
76 Part II: Homeostatic Control

intracellular calcium concentration increases stimuli such as whisker, limb, odor, and visual
that can reach micromolar levels and are known stimulations (Wang et al., 2006; Winship et al.,
to affect cell physiology. The development and use 2007; Dombeck etal., 2007; Petzold etal., 2008;
of calcium imaging techniques has been pivotal Schummers etal., 2008), supporting a functional
for investigating the physiology of astrocytes. The role for astrocytic excitability in brain physiology.
growth of interest in astrocytic calcium signaling In addition, astrocytic calcium signals have been
came soon after its first discovery when it was shown to be important for the control of cerebral
found that calcium increases in astrocytes could arteriole diameter (Zonta et al., 2003; Mulligan
be triggered by using the excitatory neurotrans- and MacVicar, 2004; Takano etal., 2006; Metea
mitter glutamate in cultures (Cornell-Bell etal., et al., 2006; Filosa et al., 2006; Chuquet et al.,
1990)or by evoking synaptic release of glutamate 2007; Gordon etal., 2008; Kuga etal., 2011)and
in hippocampal slices (Porter and McCarthy, the regulation of brain blood flow during optical
1996). In some circumstances, astrocytic calcium signaling (Schummers etal., 2008)and locomo-
signals may propagate to neighboring astrocytes tor behavior (Nimmerjahn etal., 2009), although
as intercellular calcium waves (Cornell-Bell etal., Takata etal. (2013) have a differentview.
1990; Bowser and Khakh, 2007). Since then, Astrocytic calcium signals have been
similar observations have been made in vivo in originally and extensively studied only in
anesthetized or freely behaving animals (Hirase the somata and thick branches due to tech-
et al., 2004; Dombeck et al., 2007; Kuga et al., nical limitations using bulk loading of
2011) and in human cortical and hippocampal membrane-permeable organic calcium indi-
slices prepared from acutely resected surgical tis- cator dyes (for a review see Tong et al., 2013).
sue (Oberheim etal., 2009, Navarrete etal.,2013). Loading of membrane-impermeable organic
The synaptic and neuronal controls of astro- calcium indicator dyes via glass pipettes has
cytic calcium originate from the fact that astro- also been used to measure calcium signals
cytes express a variety of G-protein coupled in astrocytes (Di Castro et al., 2011; Panatier
receptors for different neurotransmitters whose etal., 2011). Using this method, Di Castro etal.
activation increases the intracellular concentra- and Panatier etal. observed different patterns
tion of calcium through the release of calcium of calcium signals within astrocyte processes;
from intracellular inositol 1,4,5-trisphosphate they can be compartmentalized, localized,
(IP3)-sensitive calcium stores (for a review see focal, or expanded and can be dependent or
Perea etal., 2009). The most well-known recep- independent of neuronal activity (Di Castro
tors involved in calcium increases in astrocytes etal., 2011; Panatier etal., 2011). More recently,
are the metabotropic receptors activated by the use of cytosolic- and membrane-targeted
purines such as P2Y1, and the metabotropic glu- genetically encoded calcium indicators
tamate receptors 1 (mGluR1) and 5 (mGluR5), (GCaMPs and Lck-GCaMPs, respectively)
although for the latter, a recent study has ques- revealed a novel type of microdomain calcium
tioned its contribution to astrocytic calcium signal (Shigetomi et al., 2011). These calcium
signaling in the adult brain (Sun etal., 2013). It signals, called spotty calcium signals, are spa-
is also noteworthy that the IP3-sensitive calcium tially restricted to the membrane within the
store, although predominant, is not the only very thin astrocytic processes and are thought
source of calcium within the astrocyte. There are to contribute to basal cytosolic calcium lev-
multiple other sources of calcium that can lead els. The mechanisms underlying spotty cal-
to global increases in intracellular calcium con- cium signals differ from those for the more
centrations such as mitochondria and sodium/ classical aforementioned G-protein coupled
calcium exchangers, nonselective cation chan- receptorsmediated calcium signals; they
nels, and voltage-gated calcium channels in the involved in fact the entry of calcium from the
plasma membrane (for a review see Parpura and extracellular space through a family of nonse-
Verkhratsky,2012). lective cation channels, the transient receptor
An extremely interesting finding in the glial potential channels ankyrin 1 (Shigetomi etal.,
field was the discovery in the in vivo brain that 2011). Thus the development of new methods
astrocytic signals could differ in their spatial for studying the diversity of calcium signals
extent and in their duration during physiological within single astrocytes is and will be essential
processes such as locomotion (Nimmerjahn etal., to investigate in-depth diversity of functions
2009)and in response to a variety of physiological for astrocytes in different parts of thebrain.
Role of Astrocytes in Sleep and Epilepsy 77

ASTROCYTES RELEASE
CHEMICAL TR ANSMITTERS
T O M O D U L AT E S Y N A P T I C
AND NEURONAL ACTIVIT Y
One of the most exciting discoveries in the past
20years in the glial field has been that astrocytes
can release chemical transmitters that are able to
modulate synaptic and neuronal activity. In anal-
ogy with neurotransmitter release from neurons,
they have been termed gliotransmitters. This
discovery came from seminal works showing that
astrocytes respond with calcium elevations to
synaptically released neurotransmitters (Porter
and McCarthy, 1996; see previous discussion)
and that astrocytes could also release transmit-
ters (Parpura et al., 1994). Thus the realization
of this bidirectional communication between
neurons and astrocytes led to the concept of the
tripartite synapse (Araque etal., 1999), which not
only consists of pre- and postsynaptic elements
but also of an astrocytic element that responds to FIGURE 5.1: Proposed model of the modula-
presynaptic transmitter release and feeds back via tion of brain functions by astrocytic purines.
gliotransmitter release to affect synaptic activity. Astrocytes release ATP through a SNARE-dependent
The most-studied gliotransmitters are glu- exocytosis or via the gap-junction connexin 43 hemichan-
tamate, D-serine, gamma-aminobutyric acid nels (Cx43), volume-activated anion channels (VAAC), or
(GABA), adenosine triphosphate (ATP), and pore-forming P2X7 receptors. In the extracellular space
adenosine. Calcium-dependent exocytosis rep- (ECS), ATP is converted to adenosine by a combination
resents the better characterized pathways of gli- of extracellular enzymes. Ecto-nucleoside triphosphate
otransmitter release (for reviews see Gucek etal., diphosphohydrolases (E-NTPDases) and ecto-nucleotide
2012; Zorec et al., 2012). In culture, astrocytes pyrophosphatase/phosphodiesterases (E-NPPs) metabo-
express the soluble N-ethylmaleimide-sensitive lize ATP and adenosine diphosphate (ADP) to adenosine
fusion protein receptor (SNARE) complex, monophosphate (AMP), whereas alkaline phosphatases
which is colocalized with small vesicles posi- (ALPs) and ecto-5-nucleotidase (CD73) metabolize AMP
tive for vesicular glutamate transporters (Bezzi to adenosine. Adenosine can be taken up into the intra-
et al., 2004; Zhang et al., 2004; Montana et al., cellular space (ICS) of the astrocyte through the bidirec-
2004), ATP-storing vesicles (Coco et al., 2003; tional ectonucleotide transporters (ENTs). This passive
Zhang et al., 2007; Sawada et al., 2008), and transport of adenosine depends on the activity of the
D-serine-containing vesicles (Mothet et al., intracellular enzyme adenosine kinase (ADK) that elimi-
2005). In addition, ultrastructural studies have nates adenosine via phosphorylation to AMP and drives
shown that astrocytic processes contain small the flux of adenosine into the astrocyte. In the ICS of the
synaptic-like vesicles with a diameter of 30 to astrocyte, adenosine can also be converted to inosine by
40 nM (Bezzi et al., 2004; Jourdain et al., 2007; the enzyme adenosine deaminase (ADA). The accumula-
Martineau et al., 2013). Importantly, these vesi- tion of astrocytic adenosine in the ECS leads to the tonic
cles contain glutamate and D-serine and are activation of the neuronal adenosine A1 receptor, result-
clustered in close proximity to synapses (Bezzi ing in the modulation of synaptic transmission and traf-
et al., 2004; Jourdain et al., 2007; Martineau ficking of NMDA receptors at the cell surface. In a more
et al., 2013). Convincing evidence for astro- integrated system, this contributes to the modulation of
cytic vesicular release of ATP is provided by the cortical network activity, sleep homeostasis, cognitive
dominant-negative SNARE (dnSNARE) mouse functions, and epilepsy. These brain functions attributed
in which exocytosis in astrocytes is selectively to astrocytic purines are supported by a series of experi-
inhibited and results in a reduction in ATP release mental studies in which the dominant-negative form of the
following astrocyte stimulation (Pascual et al., SNARE complex (dnSNARE) was genetically expressed
2005; see following discussion for more details into the astrocytes to disrupt the SNARE-dependent exo-
and Figure 5.1). Other gliotransmitter release cytosis of ATP. See text for additional details.
78 Part II: Homeostatic Control

mechanisms have been proposed, including for the impairment of spatial working memory
membrane transporters, volume-activated anion and NMDA-dependent hippocampal long-term
channels, ligand-gated P2X7 channels, and con- depression (LTD) in vivo induced by an acute
nexin/pannexin hemichannels (for a review see exposure of exogenous cannabinoids.
Malarkey and Parpura, 2008)and more recently A hallmark of astrocytic glutamate release
the two-pore domain potassium channels and is the generation of slow inward currents (SICs)
the bestrophin anion channels (Lee et al., 2010; in postsynaptic neurons from slice preparations.
Woo et al., 2012; Han et al., 2013). Although SICs were first identified in the thalamic neurons
numerous studies have demonstrated the role of where they were correlated with spontaneous
gliotransmitters in synaptic transmission, there calcium spikes in astrocytes (Parri et al., 2001).
is still debate and controversy about the mecha- Since then, SICs have been detected in many
nism by which they are released, whether or not it regions of the CNS such as the cortex (Ding etal.,
is calcium-dependent, and whether it has physio- 2007), hippocampus (Fellin etal., 2004), nucleus
logical significance (Fiacco etal., 2007; Petravicz accumbens (D Ascenzo et al., 2007), olfactory
etal., 2008; Agulhon etal., 2010; Hamilton and bulb (Kozlov etal., 2006), brainstem (Reyes-Haro
Attwell, 2010; Wang et al., 2013). Rather than etal., 2010), spinal cord (Bardoni etal., 2010; Nie
debating the relevance of each of these mecha- etal., 2010), and, more recently, human cortical
nisms, we provide a synopsis of recent work sup- and hippocampal slices prepared from acutely
porting the functional role of gliotransmitters in resected surgical tissue (Navarrete et al., 2013).
synaptic and neuronal physiology. Typically, SICs are due to a calcium-dependent
release of astrocytic glutamate that preferentially
Release ofGlutamate activates extrasynaptic NMDA receptors con-
Glutamate was one of the first gliotransmit- taining the NR2B subunit (Fellin et al., 2004).
ters identified in the early 1990s (Parpura etal., SICs can occur spontaneously or be synaptically
1994). Since then, numerous studies have shown evoked (Parri et al., 2001; Fellin et al., 2004).
that astrocytic glutamate released through a Although the physiological role of SICs is still
calcium-dependent mechanism exerts many unknown or even questioned by some (Fiacco
effects on synaptic transmission and neu- etal., 2007), they can induce neuronal depolari-
ronal excitability. By acting on presynaptic zation and synchronize firing of CA1 pyramidal
glutamatergic receptors, including mGluR1, cells over short distances of 100m (Fellin etal.,
mGluR2/3, mGluR5, kainate receptors, and 2004), suggesting that SICs could represent a
N-methyl-D-aspartate (NMDA) receptors, astro- source of excitation during epileptic discharges
cytic glutamate strengthens or weakens inhibi- as discussed later in this chapter. Interestingly,
tory and excitatory synapses and consequently studies conducted in spinal cord slices have
participates in synaptic plasticity (Pasti et al., shown that SICs could be triggered by pharma-
1997; Kang etal., 1998; Liu etal., 2004a, 2004b; cological blockade of glutamate uptake with the
Fiacco et al., 2004; Perea and Araque, 2007; nontransportable competitive glutamate trans-
Jourdain et al., 2007; Navarrete and Araque, porter blocker D-Threo--benzyloxyaspartate
2010; Bonansco etal., 2011). Although all of these (Nie et al., 2010), suggesting that changes in
studies were mainly performed in acute brain ambient glutamate concentrations could rep-
slice preparations, only a few have been carried resent a key component that contributes to the
out in vivo to elucidate the role of astrocytic regulation of this bidirectional glutamatergic
glutamate in synaptic plasticity. For example, interaction between neurons and astrocytes.
Navarrete etal. (2012) demonstrated that astro- However, the prevalence of this phenomenon in
cytic glutamate was involved in the generation other brain areas and its (patho)physiological
of in vivo hippocampal long-term potentiation implications remain to be determined.
(LTP) mediated through activation of neuronal
mGluRs following stimulation of the cholinergic Release ofD-Serine
septo-hippocampal pathway. Asimilar plasticity Convincing ultrastructural studies indicate that
has been observed in the cortex in vivo involv- astrocytes contain all the machinery that is neces-
ing astrocytic muscarinic acetylcholine recep- sary for the calcium-dependent vesicular release
tors and neuronal NMDA receptors (Takata of D-serine (Mothet etal., 2005; Martineau etal.,
etal., 2011). Finally, Han etal. (2012) showed that 2013). This amino acid is produced within astro-
astroglial cannabinoid receptors were responsible cytes through the conversion of L- to D-serine
Role of Astrocytes in Sleep and Epilepsy 79

by the specific enzyme serine racemase (Mothet astrocytes is not very well described. Some have
et al., 2000; Mothet et al., 2005). Acting as the proposed a calcium-dependent mechanism
natural substrate for glycine binding sites on involving the anion bestrophin anion channels in
the NMDA receptor, D-serine is considered an the cerebellum (Lee etal., 2010). However, in this
endogenous co-agonist of the NMDA recep- case, bestrophin anion channels mediate tonic
tor and thus can potentially modulate synap- inhibition instead of SOCs (Lee etal., 2010). The
tic plasticity (Mothet et al., 2000; Mothet et al., physiological role of SOCs remains completely
2005). Indeed, in the supraoptic nucleus of the unknown, although it has been suggested that,
hypothalamus of rodents, changes in the glial like SICs, they could play an important role under
coverage of synapses dependent on physiologi- pathological conditions such as epileptiform dis-
cal signals affect extracellular levels of D-serine charges (Jimenez-Gonzalez etal., 2011; Le Meur
and consequently can lead either to LTP or LTD et al., 2012). Interestingly, a recent study has
in acute brain slices (Panatier etal., 2006). This shown that SOCs recorded in thalamic slices from
metaplastacity is critical for the modulation of a genetic rat model of absence epilepsy (genetic
excitability of magnocellular neurosecretory absence epilepsy rats from Strasbourg) displayed
cells during neuroendocrine processes such as a reduced amplitude compared with control ani-
lactation and parturition (Panatier etal., 2006). mals (Pirttimaki etal., 2013). The smaller ampli-
Moreover, in situ studies manipulating serine tude of SOCs seems to result from a malfunction
racemase activity and using calcium clamp of of the astrocytic GABA-transporter GAT-1 in
astrocytes (Henneberger et al., 2010) or attenu- these animals, suggesting that, like SICs, SOCs
ating basal calcium levels in astrocytes through can be influenced by the ambient GABA concen-
inhibition of transient receptor potential chan- trations. However, it remains difficult to ascribe
nels ankyrin 1 channels (Shigetomi et al., 2013; a precise pathological significance to the smaller
see previous discussion) have revealed that astro- amplitude of SOCs in these epileptic animals.
cytic D-serine regulated LTP within the hip-
pocampus by targeting preferentially synaptic ReleaseofATP
NMDA receptors (Papouin etal., 2012). Arole for Astrocytic ATP was initially proposed as an
astrocyte-derived D-serine in cortical plasticity extracellular messenger and a primary signal
has also been suggested (Takata etal.,2011). for the propagation of calcium waves through
the astrocytic network by acting directly on
Release ofGABA purinergic receptors (Guthrie et al., 1999; for a
Despite early reports showing that astrocytes can review see Giaume et al., 2010). Although dif-
accumulate, synthesize, and release GABA, the ferent mechanisms of ATP release from astro-
idea that this major inhibitory transmitter of the cytes have been proposed (Figure 5.1), including
mammal CNS could also be a gliotransmitter has volume-activated anion channels, gap-junction
not been widely supported (for reviews see Angulo connexin 43 hemichannels, and pore-forming
etal., 2008; Velez-Fort etal., 2012). Several stud- P2X7 receptors, convincing morphological and
ies mainly performed in acute brain slices have biochemical evidence suggests that astrocytes
shown that astrocytes from the olfactory bulb, may preferentially use calcium-dependent exo-
thalamus, and hippocampus inhibit neighboring cytosis to release ATP (for a review see Gucek
neurons by releasing GABA, which generates slow et al., 2012). Additionally, Pascual et al. (2005)
outward currents (SOCs) due to the activation of have generated an inducible transgenic mouse
neuronal GABA A receptors (Kozlov et al., 2006; in which only astrocytes express a dnSNARE
Jimenez-Gonzalez etal., 2011; Le Meur etal., 2012). domain of vesicle-associated membrane pro-
Interestingly, these SOCs share some similarities tein 2 to suppress exocytosis of gliotransmitters.
with NMDA receptor-mediated SICs caused by Bioluminescence imaging demonstrated that this
the release of glutamate from astrocyte. Indeed, molecular genetic manipulation led to reduced
SOCs can occur simultaneously in neighboring extracellular ATP, providing further support that
neurons and provide a source of synchronized ATP release from astrocytes is mediated through
inhibition by targeting extrasynaptic GABAA a vesicular-dependent mechanism (Pascual
receptors (Kozlov etal., 2006; Jimenez-Gonzalez etal.,2005).
etal., 2011). Although SICs are mediated through Once ATP is released from astrocytes, it can
an exocytotic release of astrocytic glutamate, the affect synaptic and neuronal activity through
mechanism by which GABA is released from its direct action on purinergic receptors. For
80 Part II: Homeostatic Control

example, in the paraventricular nucleus of the extracellular concentration of adenosine depends


hypothalamus of dehydrated rats, withdrawal on the activity of adenosine kinase (ADK), an
of the glial coverage surrounding magnocellular intracellular enzyme mainly expressed in astro-
neurosecretory cells influences extracellular lev- cytes that converts adenosine to AMP (Figure
els of ATP released from astrocytes in response 5.1). This conversion creates a concentration gra-
to adrenergic input (Gordon etal., 2005). During dient that drives the influx of adenosine into the
this process, ATP has been shown to directly cell through bidirectional equilibrative nucleo-
activate P2X7 receptors located on magnocellu- tide transporters (Figure5.1).
lar neurosecretory cells to enhance membrane In situ studies have shown that adenosine
insertion of alpha-amino-3-hydroxy-5-methyl- derived from astrocyte-released ATP controls
4-isoxazolepropionic acid receptors and thereby synaptic and neuronal activity. In the retina, for
cell excitability (Gordon etal., 2005). This mech- example, extracellular adenosine, which derives
anism is critical for facilitating the secretion of from ATP released by the Mller glial cells,
the two neurohormones, oxytocin and vasopres- hyperpolarizes neurons and inhibits their firing
sin, involved in the control of osmoregulation through the activation of A1 receptors (Newman,
(Gordon etal., 2005). Interestingly, further stud- 2003). In addition, using pharmacology in the
ies indicated that this physiological process also astrocyte dnSNARE mouse, seminal studies have
involved an mGluR1-mediated rise in calcium in discovered that the SNARE-mediated release of
astrocytes (Gordon et al., 2009). Another study ATP represents an important source of extracel-
from Gourine et al. (2010) using in situ and in lular adenosine (Figure 5.1), which suppresses
vivo approaches has shown that astrocytes in the excitatory synaptic transmission through the
brainstem contributes to the regulation of venti- activation of presynaptic A1 receptors (Pascual
lation through a pH-induced vesicular release of et al., 2005; Halassa et al., 2009; Schmitt et al.,
ATP that in turn increases activity of medullary 2012). Although this astrocytic signaling path-
chemo-receptor neurons. way is supported by other studies (Zhang etal.,
2003; Serrano etal., 2006), it has been suggested
Adenosine Derived From that SNARE-mediated release of ATP could
Astrocyte- ReleasedATP instead increase excitatory synaptic transmis-
Besides its direct actions through purinergic sion by acting specifically on presynaptic A2A
receptors, ATP can also exert indirect effects receptors (Panatier et al., 2011). Subsequently,
on its rapid conversion to adenosine by a com- direct measurements of adenosine in hipp-
bination of extracellular enzymes (Figure 5.1). pocampal slices with adenosine biosensors have
Ecto-nucleoside triphosphate diphosphohy- confirmed that astrocytic dnSNARE expression
drolases and ecto-nucleotide pyrophosphatase/ reduces extracellular levels of adenosine (Schmitt
phosphodiesterases metabolize ATP and adeno- etal., 2012), supporting the idea that one source
sine diphosphate to adenosine monophosphate of extracellular adenosine is SNARE-mediated
(AMP), and alkaline phosphatases (ALPs) and release of ATP that is hydrolyzed in the extracel-
ecto-5-nucleotidase metabolize AMP to adeno- lular space to adenosine (Figure5.1).
sine (Zimmermann, 2000). Subsequently, adeno- In addition to exerting presynaptic inhibition
sine can exert numerous effects in the brain that via A1 receptors, adenosine derived from astro-
are mediated through the activation of four sub- cytic ATP can also exert postsynaptic actions to
types of G-protein-coupled adenosine receptors regulate the trafficking of NMDA receptors in
(A1, A2A, A2B, and A3; for a review see Fredholm cortical and hippocampal pyramidal neurons
etal., 2005). It is noteworthy that the breakdown (Figure 5.1; Fellin etal., 2009; Deng etal., 2011;
of ATP to adenosine in the extracellular space is Clasadonte etal., 2013). Using the dnSNARE mice
not the only source of adenosine. Adenosine can along with biochemical, pharmacological, and
be directly released from neurons and astrocytes electrophysiological approaches, further studies
through a passive mechanism involving a family demonstrated that the tonic activation of post-
of equilibrative nucleotide transporters (Boison, synaptic A1 receptors leads to a delayed Src fam-
2013; Figure 5.1). This direct release of adenosine ily kinase-dependent tyrosine phosphorylation of
occurring typically during hypoxic conditions the NMDA receptor subunits NR2A and NR2B
has been recently debated (Martin et al., 2007; that is required for their endocytosis (Deng etal.,
Parkinson etal., 2009; Lovatt etal., 2012; Zhang 2011). In the dnSNARE animals, the reduced
etal., 2012; Wall and Dale, 2013). In this case, the extracellular adenosine tone leads to a reduced
Role of Astrocytes in Sleep and Epilepsy 81

cell surface expression of the NMDA receptors (for a review see Brown etal., 2012). In the follow-
that thereby decreases the NMDA current (Fellin ing section, we focus on recent studies that have
et al., 2009; Deng et al., 2011; Clasadonte et al., investigated the role of astrocyte-derived adeno-
2013). These results are furthermore consistent sine in sleep-related behavior.
with earlier studies showing that dnSNARE mice
exhibit a reduced NMDA receptor-dependent Astrocytes Modulate Brain Activity
LTP at the hippocampal synapses (Pascual etal., atthe CircuitLevel
2005). It is important to note that this regulation The brain displays distinct network dynamics in
of postsynaptic NMDA receptor trafficking by different states of anesthesia, wakefulness, and
astrocytic adenosine is a slow process. Indeed, sleep. During non-rapid eye movement (NREM)
pharmacological studies performed in cortical sleep and certain forms of anesthesia, cortical
slices by Deng etal. (2011) have shown that the local field potential patterns are characterized
reduction of NMDA receptor trafficking was by the large-amplitude slow oscillations (< 1 Hz),
phenocopied in wild-type littermates (WT) only whereas small-amplitude and rapid fluctuations
when slices were incubated with the A1 receptor appear during the two other vigilance states
antagonist 8-cyclopentyl-1,3-dimethylxanthine rapid eye movement (REM) sleep and wakeful-
(CPT) for at least 3 hours, and rescued in ness (Steriade, 1997). The cortical slow oscilla-
dnSNARE animals only when slices were incu- tions detected in vivo with local field potential
bated with the A1 receptor agonist 2-Chloro-N6- reflect the activity of a vast network of pyrami-
cyclopentyladenosine for at least 1 hour. Thus dal neurons that oscillate between a depolarized
astrocytes can regulate NMDA receptors function up-state and a hyperpolarized down-state with
over different timescales. Release of glutamate a high level of synchrony (Steriade etal., 2006).
and D-serine by astrocytes provides the poten- While recording cortical local field potential in
tial for second-to-second changes in the NMDA vivo in anesthetized mice, it was found that the
receptor current, while the adenosine-dependent expression of dnSNARE in astrocytes caused
pathway that regulates NMDA receptor traffick- a striking reduction in the power of slow oscil-
ing represents a much slower process. By regulat- lations compared to that from WT littermates
ing NMDA receptor trafficking, astrocytes could (Fellin etal., 2009; Schmitt etal., 2012). In addi-
contribute to homeostatic synaptic plasticity that tion, local disinhibition of the circuits by acute
works over long timescales (hours to days) such blockade of the A1 receptors with CPT increased
as during sleep/wake cycles (Vyazovskiy et al., the power of cortical slow oscillations in WT
2008) in which astrocyte-derived adenosine is mice while this effect was abolished in dnSNARE
known to play important roles (Halassa et al., mice, confirming a reduced extracellular level
2009), as described previously. Additionally, we of adenosine in these transgenic animals (Fellin
will see later in this chapter that this particular et al., 2009; Schmitt et al., 2012). Importantly,
astrocytic signaling pathway is critical for the the SNARE-dependent A1 receptor modulation
development of temporal lobe epilepsy(TLE). of both cortical slow oscillations and synaptic
transmission was dependent on the time of the
ROLE OFASTROCYTES day and more specifically dependent on the time
INSLEEP spent in wakefulness (Schmitt etal., 2012). These
Because adenosine is known to be a powerful results provide the first in vivo evidence that
modulator of sleep (for a review see Basheer etal., astrocyte-derived adenosine modulates brain
2004), several studies have investigated whether activity at the circuit level and is potentially
astrocyte-derived adenosine could contribute to involved in the control of sleep-related behavior.
the regulation of sleep physiology (for a review
see Blutstein and Haydon, 2013). It is known that Astrocytes Modulate Sleep
adenosine levels rise during wakefulness; sleep Homeostasis
deprivation leads to adenosine elevations, and In line with the aforementioned study dem-
infusion of adenosine promotes sleep while infu- onstrating that astrocytic adenosine is critical
sion of adenosine receptors antagonists such as for the control of circuit function in the cortex,
caffeine promotes wakefulness. Although the Halassa et al. (2009) performed studies to ask
functions of sleep are still unresolved, recent whether this astrocyte-derived adenosine con-
progress has been made in deciphering the brain tributes to sleep homeostasis. Sleep is modulated
mechanisms that control sleep and wakefulness by both the circadian oscillator that controls the
82 Part II: Homeostatic Control

timing of sleep and wakefulness and the sleep et al., 2009). Accordingly, it has been demon-
homeostat that integrates the time awake and strated that the reduced power of cortical slow
promotes the sleep drive. The sleep drive, also oscillations observed in dnSNARE animals
called sleep pressure, can be monitored with cor- could be phenocopied in WT animals when both
tical electroencephalography (EEG) by measur- A1 and NMDA receptors were acutely inhibited
ing the power of slow-wave activity (SWA:delta with CPT and the NMDA receptor antagonist
frequency range 0.54 Hz) during NREM sleep. D-()-2-amino-5-phosphonopentanoate (D-AP5),
It is commonly accepted that sleep pressure and respectively (Fellin et al., 2009).
the power of SWA per se increases in proportion The idea that astrocytic adenosine controls
to the time spent awake and decreases during sleep homeostasis is supported by other stud-
sleep (Borbly and Achermann, 2005). Thus, by ies. For example, reduced SWA during NREM
combining EEG with electromyogram (EMG) sleep have also been previously described in con-
recordings in mice to monitor the three differ- ditional knockout mice lacking A1 receptors in
ent vigilance states NREM, REM, and wakeful- neurons (Bjorness et al, 2009). Mice, in which
ness, Halassa etal. (2009) found that dnSNARE the two enzymes ecto-5-nucleotidase and ADK
expression in astrocytes reduced the power of known to regulate adenosine metabolism (Figure
SWA during NREM sleep at the onset of the sleep 5.1) have been genetically manipulated to lower
period while it was normally elevated in WT mice adenosine tone in the brain, also display sleep
after a natural period of prolonged wakefulness. phenotype (Zielinski et al., 2012; Palchykova
In addition, astrocytic dnSNARE expression etal., 2010). In addition, patients bearing a genetic
attenuated the increased in SWA during NREM mutation of the intracellular adenosine deami-
sleep in response to acute sleep deprivation of 6 nase, an enzyme involved in the inactivation of
hours compared with WT mice (Figures 5.2Ai, adenosine (Figure 5.1), display an increased sleep
5.2Aii, and 5.2B) and prevented the compensa- pressure and present a more consolidated sleep at
tory increase in sleep time that normally occurs night (Bachmann etal.,2012).
after sleep deprivation (Halassa et al., 2009;
Figure 5.2C). Importantly, the dnSNARE sleep Astrocytes, Sleep Loss, and
phenotype was entirely phenocopied in WT ani- Memory Consolidation
mals by chronic intracerebroventricular deliv- An important function of sleep is offline pro-
ery of the A1 receptor antagonist CPT (Halassa cessing of information encountered during the
etal., 2009). Although these results suggest that day and consolidation of memory. Conversely,
the SNARE-dependent release of adenosine from accumulation of sleep pressure after prolonged
astrocytes represents a major component of the wakefulness is associated with substantial
homeostatic control of sleep (Figure 5.1), some impairments in cognitive functions (McCoy and
experimental discrepancies must be noted. The Strecker, 2011; Yoo etal., 2007). Given that astro-
chronic inhibition of A1 receptors that reduces cytes play a major role in the homeostatic con-
the power of SWA and thereby mimics the trol of sleep, Halassa etal. (2009) investigated the
dnSNARE phenotype (Halassa etal., 2009)is in contribution of these glial cells in the cognitive
apparent contradiction with the fact that acute deficits induced by sleep loss. While high sleep
inhibition of A1 receptors increases the power pressure in WT mice was associated with poor
of cortical slow oscillations (Fellin et al., 2009; performances in memory recognition tests fol-
Schmitt et al., 2012). These differences can be lowing acute sleep deprivation, the lack of accu-
explained by the fact that the effects of acute and mulated sleep pressure in dnSNARE animals
chronic inhibitions of A1 receptors are mediated prevented the effects of sleep loss on the consoli-
by two distinct mechanisms. Acute inhibition dation of recognition memory. In addition, the
of A1 receptors attenuates the power of cortical chronic infusion of the A1 receptor antagonist
slow oscillations by causing a rapid local disinhi- CPT in WT mice mimicked the dnSNARE phe-
bition of the circuits (Fellin etal., 2009; Schmitt notype by preventing the deleterious effects of
etal., 2012). On the other hand, long-term inhi- sleep deprivation on memory recognition.
bition of A1 receptors have been shown to cause Similarly, another study performed by
a delayed reduction in NMDA receptor activity Florian etal. (2011) demonstrated that dnSNARE
(Deng et al., 2011) and thus could explain why animals exhibited better performances in spatial
dnSNARE animals displayed a reduced power memory tests than WT animals after sleep dep-
of both cortical slow oscillations (Fellin et al., rivation. This lack of alteration in the consolida-
2009; Schmitt et al., 2012) and SWA (Halassa tion of spatial memory was consistent with the
Role of Astrocytes in Sleep and Epilepsy 83

(a)(i) WT
Baseline (ZT7-8) Recovery after SD (ZT7-8)

EEG
5V
EMG
1s

(a)(ii) dnSNARE
Baseline (ZT7-8) Recovery after SD (ZT7-8)

EEG
5V
EMG
1s

(b) Recovery after SD (c)


Baseline
260
WT Recovery
Normalized Power

220 * * dnSNARE
60 N.S.
(0.51.5 Hz)

**

(% Recording Time)
Total Sleep Time
180 *
40

140 20

100 0
6 8 10 12 WT dnSNARE
Time (Zeitgeber)

FIGURE 5.2: Gliotransmission is essential for sleep pressure accumulation. (Ai) Representative
EEG/EMG recordings in non-rapid eye movement (NREM) sleep from wild-type (WT) mice during baseline day
(left) and recovery day after 6 hours of sleep deprivation (SD, right). Recordings during the baseline and recovery
days are shown at the same time point between the zeitgeber times 7 and 8 (ZT78). Note that the amplitude of the
EEG slow-wave activity (SWA) in WT mice increases during recovery after SD, compared to baseline. (Aii) Same
experiments as in (Ai) but performed with dnSNARE mice. Note that the amplitude of EEG SWA in dnSNARE
mice does not increase during recovery after SD, compared to baseline. (B)Analysis of the normalized power of
SWA during NREM sleep indicated that SWA was decreased in dnSNARE mice (n=7) following SD when com-
pared with WT littermates (n=8) (ANOVA:p < 0.001, F=7.91; post hoc test, *p < 0.05). (C)SD increased the total
sleep time in WT (n=8) but not in dnSNARE animals (n=9) during an 18-hour recovery period compared with
a baseline period (Students paired t test:**p < 0.01; N.S.=nonsignificant).
(B) and (C)adapted from Halassa etal. (2009).

fact that dnSNARE mice did not show a decrease has a potent and rapid antidepressant action in
in the late phase of hippocampal LTP after sleep severely depressed individuals (for a review see
deprivation when compared with WT mice. Hemmeter etal., 2010). Some have even suggested
Further, the chronic treatment of WT mice with that insomnia, a common symptom of depres-
CPT in this study also mimicked the dnSNARE sion, might represent an endogenous adaptive
phenotype. Overall, these studies suggest that mechanism to counteract underlying depres-
astrocyte-derived adenosine contributes to the sive mood rather than just be part of the pathol-
impairment of memory consolidation following ogy (Adrien, 2002). However, the mechanisms
sleeploss. underlying antidepressant effects of sleep dep-
rivation are still relatively unknown. Given that
Astrocytes, Sleep Loss, astrocytes modulate sleep homeostasis through
and Depression A1 receptor signaling (Halassa et al., 2009), a
There are several links between depression and recent study has investigated the potential role
sleep. Both monopolar and bipolar depression are of glial cells in the beneficial effects of sleep
associated with sleep disturbances. Paradoxically, deprivation on depression (Hines et al., 2013).
it has been observed that acute sleep deprivation In this study, the authors first confirmed that
84 Part II: Homeostatic Control

depressed animals that were sleep-deprived for whether astrocytes contribute to the increased
12 hours displayed significantly less depressive sleep pressure during systemic infections and
symptoms than control depressed mice. Then, whether this is a result of changes in adenosine
using the dnSNARE animals to disrupt astro- signaling. Astrocytic dnSNARE expression pre-
cytic adenosine signaling, they found that this vented the increase in both NREM SWA power
genetic manipulation prevented the antidepres- and NREM sleep time that normally occur in
sant effect of sleep deprivation on depressive-like WT mice during the first hours following intra-
behaviors. Additionally, they demonstrated that peritoneal injections of lipopolysaccharide. In
the antidepressant effects were potentially due to addition, intracerebroventricular delivery of
a sleep deprivationinduced increase in extracel- CPT to block A1 receptors in WT mice mimicked
lular levels of adenosine acting specifically on A1 the dnSNARE phenotype that is consistent with
receptors. Indeed, intracerebroventricular infu- the role that astrocytic adenosine and A1 recep-
sion of the A1 receptor agonist 2-Chloro-N6- tors play in the regulation of sleep homeostasis.
cyclopentyladenosine in WT mice mimicked This work indicates that astrocytes, in addition
the effects of sleep deprivation on depression to playing a pivotal role in the CNS immune
phenotypes. Further, similarly to dnSNARE ani- and inflammatory responses by releasing sev-
mals, A1 receptor knockout mice were resistant eral relevant immunologically factors (Volterra
to the antidepressant effects of sleep depriva- and Meldolesi, 2005; Farina etal., 2007), are also
tion (Hines etal., 2013). Consistent with another critical for the control of sleep pressure during
study in which the role of astrocyte-derived ATP systemic infections by releasing adenosine that
in depressive-like behaviors has been identified subsequently acts on A1 receptors.
(Cao etal., 2013), these results provide a mecha-
nistic insight into how sleep deprivation pro- ROLE OFASTROCYTES
duces its antidepressant effects. INEPILEPSY
Besides their important role in maintaining CNS
Astrocytes and Sleeping Behavior functions under physiologic conditions, growing
During Inflammatory Response evidence suggest that astrocytes can also contrib-
It is known that activation of the immune sys- ute to the development of brain disorders such
tem induces an increase in sleep pressure (SWA) as epilepsy (for reviews see Wetherington etal.,
during NREM sleep and thereby an increase 2008; Seifert etal., 2010; Clasadonte and Haydon,
in time spent in NREM sleep. This increased 2012). Epilepsy represents one of the most com-
sleeping behavior is part of several behavioral mon neurological disorders, affecting 1% of the
changes called sickness which often occur dur- population worldwide. The clinical manifesta-
ing systemic infections (for a review see Rohleder tion of epilepsy is the occurrence of recurring,
etal., 2012). However, the mechanisms by which unprovoked spontaneous seizures caused by the
the inflammatory response promotes sleep drive hypersynchronous discharges of a population of
are not well understood. Interestingly, several neurons mainly in the hippocampus that spread
studies have shown an increase of extracellular across the brain through the recruitment of other
concentrations of ATP and its derivative adeno- neuronal populations (Spencer, 2002; Avoli etal.,
sine in the brain during the development of the 2002). Numerous studies have identified a variety
systemic inflammatory response (Krueger, 2008; of mechanisms by which astrocytes could con-
Gourine et al., 2007). Because astrocytes are a tribute to the generation of seizures (for reviews
major source of the adenosine that can regulate see Wetherington etal., 2008; Seifert etal., 2010).
sleep homeostasis, a recent study has suggested In the following sections, we summarize only
that these glial cells could contribute to the the recent works that have investigated the role
induction of sleep behavior during inflammatory of reactive astrocytosis and gliotransmission in
responses (Nadjar etal., 2013). In this study, sys- epilepsy.
temic infections in WT mice were mimicked by
peripheral lipopolysaccharide treatment. Using Reactive Astrocytosis and Epilepsy
EEG/EMG recordings, the authors first con- Reactive astrocytosis (also known as reactive
firmed that intraperitoneal injections of lipopol- astrogliosis) is often associated with TLE in
ysaccharide in WT mice induced an increase humans (Cohen-Gadol et al., 2004) and animal
in both NREM SWA power and NREM sleep models of epilepsy (Borges et al., 2003; Shapiro
time. Then they used the dnSNARE mice to ask et al., 2008). How or why this process occurs is
Role of Astrocytes in Sleep and Epilepsy 85

poorly understood. Reactive astrocytosis is gen- several works in human and experimental epi-
erally observed under pathological conditions lepsy in which a relationship was found between
and is typically characterized by morphological the downregulation of GS and hippocampal
changes in astrocytes, which increase in size and reactive astrocytosis (Eid et al., 2004; Hammer
number (for reviews see Sofroniew, 2009; Robel et al., 2008). Furthermore, it has been reported
et al., 2011). In fact, reactive astrocytes display that mutation of the GS gene in humans resulted
an increase in expression of glial cytoskeletal in seizure development (Haberle et al., 2005,
proteins such as the glial fibrillary acidic protein 2006). Finally, studies in rats have also shown
(GFAP) and vimentin that are therefore used as that chronic hippocampal infusion of methio-
hallmarks of the epileptic brain (Wilhelmsson nine sulfoximine blocks GS-induced progressive
etal., 2004; Pekny and Nilsson, 2005). These reac- development of seizures along with neuropatho-
tive changes in astrocytes are also accompanied logical changes like those seen in human TLE
by the loss of astrocytic domain organization (Eid etal., 2008; Wang etal.,2009).
(Oberheim et al., 2008) and generation of new Although these experimental results suggest
astrocytes from stem cells (Borges et al., 2006). that loss of GS induces seizures by causing an
It is noteworthy that these overall morphologi- increase in extracellular glutamate, other stud-
cal changes in astrocytes during epilepsy occur ies have tested the hypothesis that loss of GS may
together with neuronal loss and synaptic rear- contribute to epilepsy by causing a depletion of
rangements within the hippocampus in a process GABA in inhibitory synaptic terminals. In sup-
called hippocampal sclerosis (Borges etal., 2003; port of this hypothesis, studies performed in
de Lanerolle et al., 2012). In addition, evidence CA1 hippocampal slices demonstrated that selec-
from studies in human epileptic tissue reports tive blockade of GS with methionine sulfoximine
that reactive astrocytes undergo striking changes altered GABAergic synaptic transmission (Liang
in the expression of specific enzymes, such as glu- etal., 2006)without affecting glutamatergic syn-
tamine synthetase (GS; Eid etal., 2012)and ADK aptic transmission (Kam et al., 2007), suggest-
(Boison, 2012). Based on recent evidence, we next ing that the glial glutamine-glutamate-GABA
discuss how these changes in astrocyte function cycle represents the major contributor to synap-
may contribute to the increased neuronal excit- tic GABA release regulating inhibitory synaptic
ability found in epileptic tissue. strength. Subsequently, an elegant study per-
formed by Ortinski et al. (2010) demonstrated
Downregulation ofGS that a higher titer viral transduction of astro-
During Epilepsy cytes with enhanced green fluorescent protein
GS found in astrocytes is the only enzyme known via injections of adeno-associated virus from
to date that is capable of converting glutamate serotype 2/5 (AAV2/5) into the mice hippocam-
and ammonia to glutamine in the mammalian pus led to reactive astrocytosis. The mechanisms
brain. This reaction is important, because it underlying AAV2/5-induced reactive astrocyto-
allows ammonia detoxification in the brain and sis are unknown. However, because this reactiv-
also provides a continuous supply of glutamine, ity in astrocytes was relatively local and occurred
which is necessary for the synthesis of gluta- in the absence of alterations in adjacent neurons
mate and GABA in neurons. Furthermore, rapid and microglia, the authors suggested that it
metabolism of glutamate via GS is a prerequisite was likely a consequence of direct interactions
for efficient glutamate clearance from the extra- between the virus itself and the astrocyte, rather
cellular space (for a review see Eid et al., 2012). than a consequence of inflammatory processes
Given that, it is likely that any alterations in the involving microglial activation. Thus AAV2/5,
glutamine-glutamate-GABA cycle could affect which has a strong astrocytic tropism, can be
the extracellular concentrations of glutamate used as a selective tool to generate reactive astro-
and GABA and consequently neuronal network cytosis isolated from other pathologic processes.
excitability. Initial clinical studies performed in In agreement with the idea that reactive astro-
humans with TLE have shown that the accumula- cytosis leads to a reduced expression of GS, the
tion of extracellular glutamate was due to a slow- authors found a pronounced downregulation of
ing in the conversion of glutamate to glutamine, GS associated with increased expression in glial
leading to the hypothesis that GS deficiency fibrillary acidic protein and vimentin. Then, by
contributes to the development of epilepsy (Otis using hippocampal slice preparations, they tested
and Jahr, 1998). This idea was then supported by the hypothesis that GS deficits, and therefore the
86 Part II: Homeostatic Control

reduced glutamine pool, could decrease inhibi- be responsible for the increased neuronal excit-
tory synaptic transmission. Interestingly, elec- ability during seizures. In support of this notion,
trophysiological recordings of CA1 pyramidal a series of studies from Boisons group identified
neurons located in the vicinity of enhanced green ADK as one of the major molecular link between
fluorescent proteinpositive areas indicated that reactive astrocytosis and neuronal dysfunc-
spontaneous and evoked GABAergic synaptic tion in epilepsy (for a review see Boison, 2012).
transmission was significantly reduced while First, ADK was found to be upregulated in reac-
glutamatergic synaptic transmission remained tive astrocytes in human and experimental epi-
unaltered. In addition, treatment of the hip- lepsy causing extracellular adenosine deficiency
pocampal slices with glutamine rescued the (Fedele etal., 2005; Aronica etal., 2011; Li etal.,
deficit in synaptic inhibition, providing further 2008, 2012). Subsequently, it has been found
proof that this GABA depletion was caused by that transgenic overexpression of ADK in mice
neuronal glutamine starvation. Finally, using induced spontaneous recurrent seizures while
voltage-sensitive dye-imaging techniques along mice with a genetic forebrain-selective reduction
with electrical stimulation of temporoammonic of ADK or receiving intrahippocampal implants
pathways, the authors demonstrated that reactive of ADK-deficient stem cellderived neuronal
astrocytosis was associated with network hyper- precursors were resistant to seizure develop-
excitability, which was subsequently attenuated ment (Li etal., 2008). These findings indicate that
when hippocampal slices were treated with glu- astroglial ADK represents a promising target for
tamine. These results suggest that GS deficits the prevention of epileptogenesis.
associated with reactive astrocytosis could con-
tribute to elevated seizure susceptibility during GLIOTR ANSMISSION
epileptic conditions by causing a loss of synaptic AND EPILEPSY
inhibition. Based on the aforementioned observa-
tions that astrocytes release glutamate in a
Upregulation ofADK During Epilepsy calcium-dependent manner and are able to con-
Under physiological conditions, the ambient trol neuronal NMDA receptors trafficking, it is
level of adenosine is controlled by a metabolic not hard to envisage that these glial cells could
clearance through astrocytes (Boison, 2013). contribute to epileptogenesis. In the following
Whereas adenosine can be released from neu- sections, we discuss the recent studies that have
rons or derived from the extracellular cleavage investigated the potential role of gliotransmis-
of ATP, the astrocyte-based enzyme ADK elimi- sion in epilepsy in situ and invivo.
nates adenosine via phosphorylation to AMP
(Figure 5.1). Because astrocytes express equili- Astrocytic Calcium-Dependent
brative nucleotide transporters, it is the meta- Release of Glutamate in Epilepsy
bolic clearance through ADK that drives the flux A direct role of gliotransmission in the genera-
of adenosine into astrocytes, which thereby form tion of epileptiform activity in situ came from
a sink for the metabolic clearance of adenosine demonstration that through calcium-dependent
(Figure 5.1). Because adenosine has been found release of glutamate, astrocytes in the hippocam-
to be elevated in patients following seizures, it pus can directly excite a group of neighboring
has been hypothesized that adenosine released CA1 pyramidal neurons in a synchronized fash-
during a seizure mediates seizure arrest and pos- ion via extrasynaptic NMDA receptor-mediated
tictal refractoriness (During and Spencer, 1992). SICs (Fellin et al., 2004). Subsequently, several
The A1 receptor-mediated functions are largely studies in brain slices (Fellin etal., 2006)and in
responsible for the anticonvulsant and neuro- anaesthetized animals (Tian et al., 2005) have
protective activity of adenosine (Fredholm etal., shown that calcium oscillations were increased
2005). Indeed, it is known that activation of A1 during chemically induced epileptiform activ-
receptors leads to a global reduction in neuronal ity. Importantly, these calcium increases in
excitability via inhibition of presynaptic gluta- astrocytes were dampened by several known
mate release (Masino et al., 2002) and postsyn- anticonvulsant drugs (Tian et al., 2005). The
aptic membrane hyperpolarization (Arrigoni calcium-dependent glutamate release from
et al., 2006). Given that, it is plausible that any astrocytes and consequently the activation of
alterations in ADK activity that consequently extrasynaptic NMDA receptors were then found
lead to changes in adenosine homeostasis could to contribute to neuronal death in the cortex
Role of Astrocytes in Sleep and Epilepsy 87

and hippocampus during the days that followed in dnSNARE animals that persisted during epi-
chemically induced status epilepticus in mice lepsy (Clasadonte et al., 2013) (Figures 5.3Ci,
(Ding et al., 2007). Finally, a recent work from 5.3Cii, and 5.3Ciii). Given that, it is likely that
Gmez-Gonzalo et al. (2010) demonstrated a this reduced activity of synaptic NMDA recep-
direct role of astrocytes in focal epilepsy. The tors, which is expected to reduce neuronal excit-
authors found that astrocytes in cortical slices ability, could mediate the effects of astrocytic
displayed massive intracellular calcium increases dnSNARE expression on epilepsy. To test this
in response to a surge of neuronal activity and by hypothesis, we used an in situ model of epilepsy
signaling back to neurons they lower the thresh- and found that the frequency of epileptiform dis-
old for the generation of focal seizures. Combined charges recorded in CA1 hippocampal slices was
with the observations that reactive astrocytes reduced by astrocytic dnSNARE expression sim-
exhibit increased expression of mGluRs dur- ilarly to results obtained on seizures in vivo. We
ing epilepsy (Aronica etal., 2000; Tang and Lee, then observed that blocking NMDA receptors
2001; Steinhauser and Geifert, 2002), which may with D-AP5 mimicked the dnSNARE phenotype
potentiate calcium-dependent glutamate release, in WT slices. These results were subsequently
these results suggest that a hyperexcitable neu- replicated in WT mice in vivo by using chronic
ronal network may arise in the brain resulting intracerebroventricular infusion of D-AP5.
from enhanced glutamatergic gliotransmission Additionally, we noticed that D-AP5 treat-
and trigger a focal site of seizure initiation. ment remained ineffective on seizure activity in
dnSNARE animals, providing further proof that
Astrocytic Control ofSynaptic NMDA the effects of astrocytic dnSNARE expression
Receptor Trafficking and Epilepsy were mediated through reduced activity of syn-
Although initial studies suggesting a role of aptic NMDA receptors (Clasadonte etal.,2013).
astrocytes in the generation of epileptiform activ- Because it is known that long-term behavioral
ity were mainly performed in situ, a direct role of abnormalities are often associated with epilepsy
gliotransmission in epileptogenesis in vivo was (Swinkels et al., 2005; Mller et al., 2009), we
identified only recently (Clasadonte etal., 2013). then investigated whether astrocytic dnSNARE
Using dnSNARE mice along with electrophysi- expression could prevent the development of
ological, behavioral, and histological approaches, these behavioral defects (Clasadonte etal., 2013).
we have demonstrated that genetic impairment Consistent with previous studies (Mller et al.,
of the SNARE complex in astrocytes led to a 2009), we found that exploratory habits of WT
hypofunction of postsynaptic NMDA receptors mice with epilepsy were dramatically affected in
and ameliorated the development of epilepsy the open field test. Epileptic WT mice displayed
that includes progressive increase in seizure fre- a striking increase in duration of stops between
quency, hippocampal sclerosis, and behavioral subsequent locomotor excursions. Interestingly,
abnormalities. these behavioral defects were significantly atten-
By using the mouse pilocarpine model of uated in epileptic dnSNARE mice (Figures5.3Di,
epilepsy along with continuous video-EEG 5.3Dii5.3Diii).
recordings that commenced 5 days after In the same study (Clasadonte et al., 2013),
pilocarpine-induced status epilepticus and con- we subsequently compared the degree of hip-
tinued for as long as 150 days, we found that pocampal sclerosis between WT and dnSNARE
astrocytic dnSNARE expression delayed the animals 8 months after pilocarpine treatment.
occurrence of the first spontaneous recurrent Immunostaining against the neuronal epitope
seizure (Figure 5.3A), reduced seizure sever- NeuN indicated a significant greater neuro-
ity, and attenuated the progressive increase protection in the hilus of the dentate gyrus
in seizure frequency (Figure 5.3B) that typi- from dnSNARE mice compared to WT mice
cally occurs in human TLE (Clasadonte et al., (Figures 5.3Ei and 5.3Eii), consistent with our
2013). In accordance with our previous stud- previous studies demonstrating that pharmaco-
ies showing that astrocytic dnSNARE expres- logical blockade of gliotransmission prevented
sion leads to a reduced expression of synaptic neuronal loss in the hippocampus after induction
NMDA receptors in cortical cells (Deng et al., of status epilepticus (Ding etal., 2007). In addi-
2011), whole-cell patch-clamp recording of CA1 tion, immunostaining against the glial markers
pyramidal neurons indicated a decrease in min- glial fibrillary acidic protein and vimentin showed
iature and evoked postsynaptic NMDA currents a striking attenuation in reactive astrocytosis in
(a) (b) (c)(i) (c)(iii)

(c)(ii)

(d)(i) (d)(ii) (d)(iii)

(e)(i) (f)(i) (g)(i)

(e)(ii) (f)(ii) (g)(ii)

FIGURE5.3: Gliotransmission modulates pathophysiological consequences of temporal lobe epi-


lepsy. (A)Kaplan-Meier analysis of the time of spontaneous recurrent seizure appearance after pilocarpine-induced
status epilepticus (SE) showing a delayed seizure onset in dnSNARE mice compared to wild-type (WT) mice (n=6
per genotype; log rank test, *p < 0.05). (B)The average number of seizures per day for each 1-month block was
attenuated from 2 to 5months after SE in dnSNARE mice (n=68) compared to WT mice (n=46; ANOVA:geno-
type, F=14.893, p=0.001; time, F=5.410, p=0.002; genotype time interaction, F=2.664, p=0.048; post hoc test,
*p < 0.05). Top insets represent spontaneous recurrent seizures detected in the cortical EEG of WT and dnSNARE
mice several months after SE. (Ci) Representative whole-cell patch-clamp recordings of NMDA and alpha-amino-
3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) currents in CA1 pyramidal neurons obtained from SE WT
and SE dnSNARE mice 5months after pilocarpine treatment. Note the reduced amplitude of NMDA current in SE
dnSNARE mice compared to SE WT mice. (Cii) AMPA/NMDA ratio was increased in SE dnSNARE mice (9 cells
from 4 animals) compared to SE WT mice (13 cells from 5 animals; Students t test, ***p < 0.001) 5months after
Role of Astrocytes in Sleep and Epilepsy 89

the dentate gyrus (Figures 5.3Fi, 5.3Fii, 5.3Gi, and Frenguelli, 2004; Boison, 2012) and neu-
and 5.3Gii) as well as in the CA1CA3 areas roprotective effects (Gomes et al., 2011), one
(Clasadonte et al. 2013). Because changes in would have expected that the reduction of
expression of astroglial proteins such as GS and extracellular adenosine acting on A1 recep-
ADK are often associated with reactive astrocyto- tors in dnSNARE animals (Pascual etal., 2005;
sis (see previous discussion), it is likely that these Halassa etal., 2009; Schmitt etal., 2012)would
molecular alterations are also dampened by astro- have worsened pathophysiological conse-
cytic dnSNARE expression during epilepsy, pro- quences of epilepsy. This discrepancy could
viding consequently further protection against a be due to the difference in the timescales in
dysregulation in the neuronal network excitabil- which the processes take place. Indeed, it has
ity, though this remains to be tested. been shown that short- and long-term inacti-
As mentioned, astrocytic glutamate acting vations of adenosine receptors have diametri-
on neuronal NMDA receptors could potentially cally opposite effects on pathophysiological
contribute to seizure generation and brain consequences of epilepsy (Georgiev etal., 1993;
damage. However, the inability to measure Von Lubitz etal., 1994; Johansson etal., 1996;
dnSNARE-dependent changes in glutamate Rigoulot et al., 2003). This pharmacological
levels, measured with microelectrode biosen- phenomenon termed effect inversion has been
sors in hippocampal slices (Clasadonte et al., questioned by Jacobson etal. (1996). Consistent
2013), makes it unlikely that the nonprogres- with the anticonvulsant role of adenosine, acute
sive development of epilepsy, including seizure administration of the A1 receptor antagonist
occurrence, neuronal damage, and behavioral CPT decreases the threshold to NMDA-induced
defects, discovered in dnSNARE animals was seizures and increases seizure-induced brain
a consequence of an impaired release of gluta- damage (Von Lubitz et al., 1994). In con-
mate from astrocytes. Since adenosine acting trast, chronic treatment with CPT, which
on A1 receptors is primarily known to exert most likely mimics the dnSNARE phenotype,
anticonvulsant (Dunwiddie, 1999; Etherington decreases seizure susceptibility and provides

FIGURE5.3 Continued
pilocarpine treatment. (Ciii) Pictures showing CA1 pyramidal neurons filled with biocytin (red) in SE WT and
SE dnSNARE mice during AMPA/NMDA ratio measurements. The presence of green fluorescent protein (GFP)
confirms that the dnSNARE transgene was expressed in astrocytes (green) of dnSNARE animals (Pascual etal.,
2005). (Di) Representative 30 min of track plots of control-no SE and SE-experienced WT mice placed in an open
field 8months after pilocarpine treatment. (Dii) Same experiments as in (Di) but performed with control-no SE
and SE-experienced dnSNARE mice. (Diii) The average duration of time stopped over the 30 min test period was
increased in SE WT mice but not in SE dnSNARE mice (WT no SE, n=8; WT SE, n=7; dnSNARE no SE, n=7;
dnSNARE SE, n=8; ANOVA followed by post hoc test, *p < 0.05 when WT SE is compared with WT no SE). (Ei)
Representative hippocampal sections from control-no SE and SE-experienced WT and dnSNARE mice stained
with an antibody directed against the neuron-specific epitope NeuN 8months after pilocarpine treatment. Striking
neuronal cell loss was observed in the hilus (h)of the dentate gyrus (DG) in SE WT mice but not in SE dnSNARE
mice. (Eii) Average neuronal density in the hilus of the DG (WT no SE, n=3 animals; WT SE, n=7 animals;
dnSNARE no SE, n=5 animals; dnSNARE SE, n=4 animals; ANOVA followed by post hoc test, **p < 0.01, ***p <
0.001; ns=nonsignificant). (Fi) Same hippocampal sections as in (Ei) but stained with an antibody directed against
the astrocyte-specific epitope glial fibrillary acidic protein (GFAP). GFAP expression was increased in the hilus
(h)of the DG in SE WT mice but not in SE dnSNARE. (Fii) Average GFAP area in the hilus of the DG (WT no SE,
n=3 animals; WT SE, n=7 animals; dnSNARE no SE, n=5 animals; dnSNARE SE, n=4 animals; ANOVA fol-
lowed by post hoc test, *p < 0.05; ns). (Gi) Representative hippocampal sections stained with an antibody directed
against the astrocyte-specific epitope vimentin 8 months after pilocarpine treatment. Vimentin expression was
dramatically increased in the hilus (h) of the DG in SE WT mice but not in SE dnSNARE mice. Samples were
obtained from another group of WT and dnSNARE mice that either did (SE) or did not enter SE (control-no SE).
(Gii) Average vimentin area in the hilus of the DG (WT no SE, n=11 animals; WT SE, n=11 animals; dnSNARE
no SE, n=9 animals; dnSNARE SE, n=7 animals; ANOVA followed by post hoc test, *p < 0.05, **p < 0.01, ***p <
0.001; ns). Average scale bars for (Ciii), (Ei), (Fi) and (Gi) represent 100m.
Adapted from Clasadonte etal. (2013).
90 Part II: Homeostatic Control

neuroprotection (Von Lubitz et al., 1994). The References


mechanism underlying the observed protection Adrien, J. (2002). Neurobiological bases for the rela-
of long-term inactivation of A1 receptors may tion between sleep and depression. Sleep Med.
rest at the level of second messenger systems Rev. 6, 341351.
coupled to these receptors. In agreement with Agulhon, C., Fiacco, T.A., and McCarthy, K.D.
this idea, we have found that one of the molecu- (2010). Hippocampal short- and long-term plas-
lar consequences of prolonged inactivation of ticity are not modulated by astrocyte Ca2+ sig-
A1 receptors is the reduction of NMDA recep- naling. Science 327, 12501254.
tor surface trafficking (Deng etal., 2011). Thus Angulo, M.C., Le Meur, K., Kozlov, A.S., Charpak,
we suggest that this long-term process mediates S., and Audinat, E. (2008). GABA, a forgotten
the beneficial effects of astrocytic dnSNARE gliotransmitter. Prog. Neurobiol. 86, 297303.
Araque, A., Parpura, V., Sanzgiri, R.P., and Haydon, P.G.
expression on pathophysiological consequences
(1999). Tripartite synapses:Glia, the unacknowl-
of epilepsy.
edged partner. Trends Neurosci. 22, 208215.
By combining a model of epilepsy that more
Aronica, E., van Vliet, E.A., Mayboroda, O.A.,
closely mimics the complex feature of TLE
Troost, D., da Silva, F.H., and Gorter, J.A. (2000).
in patients with astrocyte-specific molecular
Upregulation of metabotropic glutamate recep-
genetics, these findings revealed that astro- tor subtype mGluR3 and mGluR5 in reactive
cytes, by controlling neuronal NMDA receptor astrocytes in a rat model of mesial temporal lobe
activity, contribute to the progressive devel- epilepsy. Eur. J.Neurosci. 12, 23332344.
opment of TLE (Figure 5.1), including seizure Aronica, E., Zurolo, E., Iyer, A., de Groot, M., Anink,
occurrence, brain damage, and behavioral J., Carbonell, C., van Vliet, E.A., Baayen, J.C.,
deficits. These results provide further evidence Boison, D., and Gorter, J.A. (2011). Upregulation
to support the idea that astrocytes represent a of adenosine kinase in astrocytes in experimen-
potential therapeutic target for the treatment of tal and human temporal lobe epilepsy. Epilepsia
epilepsy. 52, 16451655.
Arrigoni, E., Chamberlin, N.L., Saper, C.B., and
CONCLUDING REMARKS McCarley, R.W. (2006). Adenosine inhibits basal
In this chapter we summarized the efforts that forebrain cholinergic and noncholinergic neu-
have been made over past decades to under- rons in vitro. Neuroscience 140, 403413.
stand the mechanisms underlying neuron- Avoli, M., DAntuono, M., Louvel, J., Kohling, R.,
to-astrocyte communication in the brain. Biagini, G., Pumain, R., DArcangelo, G., and
Although there remain some disparities Tancredi, V. (2002). Network and pharmaco-
between areas mainly due to the lack of tools logical mechanisms leading to epileptiform syn-
that allow the selective manipulation of astro- chronization in the limbic system in vitro. Prog.
cytic signals, recent work employing the trans- Neurobiol. 68, 167207.
Bachmann, V., Klaus, F., Bodenmann, S., Schafer, N.,
genic dnSNARE mouse line has been beneficial
Brugger, P., Huber, S., Berger, W., and Landolt,
to discern the role of astrocytes in more inte-
H.P. (2012). Functional ADA polymorphism
grated systems during physiological and patho-
increases sleep depth and reduces vigilant atten-
logical conditions. As examples, we discussed
tion in humans. Cereb. Cortex 22, 962970.
the role of astrocytes in sleep-related behavior
Bardoni, R., Ghirri, A., Zonta, M., Betelli, C., Vitale,
as well as in the etiology of epilepsy. The rela-
G., Ruggieri, V., Sandrini, M., and Carmignoto,
tionship between sleep and epilepsy has been G. (2010). Glutamate-mediated astrocyte-to-
recognized since antiquity and demonstrates neuron signalling in the rat dorsal horn. J.
a delicate association of brain physiology and Physiol. 588, 831846.
dysfunction. For example, sleep and sleep dep- Basheer, R., Strecker, R.E., Thakkar, M.M., and
rivation affect the distribution and frequency McCarley, R.W. (2004). Adenosine and sleep-
of seizures in humans. Consequently, it would wake regulation. Prog. Neurobiol. 73, 379396.
be very interesting in the future to investi- Ben Achour, S. and Pascual, O. (2012). Astrocyte-
gate whether glia represents the cellular link neuron communication: Functional conse-
between sleep and epilepsy. Much more work quences. Neurochem. Res. 37, 24642473.
still needs to be done to understand how gli- Bezzi, P., Gundersen, V., Galbete, J.L., Seifert, G.,
otransmission contributes to brain physiology Steinhauser, C., Pilati, E., and Volterra, A. (2004).
and pathology. In a common effort, this will Astrocytes contain a vesicular compartment that
ultimately lead to the development of new ther- is competent for regulated exocytosis of gluta-
apeutics for treating brain disorders. mate. Nat. Neurosci. 7, 613620.
Role of Astrocytes in Sleep and Epilepsy 91

Bjorness, T.E., Kelly, C.L., Gao, T., Poffenberger, V., development of temporal lobe epilepsy. Proc.
and Greene, R.W. (2009). Control and function Natl. Acad. Sci. USA 110, 1754017545.
of the homeostatic sleep response by adenosine Clasadonte, J., and Haydon, P.G. (2012). Astrocytes
A1 receptors. J. Neurosci. 29, 12671276. and epilepsy. In Jaspers Basic Mechanisms of the
Blutstein, T., and Haydon, P.G. (2013). The Epilepsies, J.L.Noebels, M.Avoli, M.A.Rogawski,
Importance of astrocyte-derived purines in the R.W.Olsen, and A.V.Delgado-Escueta, eds. (New
modulation of sleep. Glia 61, 129139. York: Oxford University Press), pp. 591605.
Boison, D. (2012). Adenosine dysfunction in epi- Clasadonte, J., Sharif, A., Baroncini, M., and Prevot,
lepsy. Glia 60, 12341243. V. (2011). Gliotransmission by prostaglandin
Boison, D. (2013). Adenosine kinase: Exploitation e(2): A prerequisite for GnRH neuronal func-
for therapeutic gain. Pharmacol. Rev. 65, tion? Front Endocrinol. (Lausanne)2, 91.
906943. Coco, S., Calegari, F., Pravettoni, E., Pozzi, D.,
Bonansco, C., Couve, A., Perea, G., Ferradas, C.A., Taverna, E., Rosa, P., Matteoli, M., and
Roncagliolo, M., and Fuenzalida, M. (2011). Verderio, C. (2003). Storage and release of ATP
Glutamate released spontaneously from astro- from astrocytes in culture. J. Biol. Chem. 278,
cytes sets the threshold for synaptic plasticity. 13541362.
Eur. J.Neurosci. 33, 14831492. Cohen-Gadol, A.A., Pan, J.W., Kim, J.H., Spencer,
Borbly, A.A., and Achermann, P. (2005). Sleep D.D., and Hetherington, H.H. (2004). Mesial
homeostasis and models of sleep regulation. temporal lobe epilepsy:Aproton magnetic reso-
In Principles and Practice of Sleep Medicine, nance spectroscopy study and a histopathologi-
M.H.Kryger, T.Roth, and W.C.Dement, eds. cal analysis. J. Neurosurg. 101, 613620.
(Philadelphia:Elsevier Saunders), pp. 405417. Cornell-Bell, A.H., Finkbeiner, S.M., Cooper, M.S.,
Borges, K., Gearing, M., McDermott, D.L., Smith, and Smith, S.J. (1990). Glutamate induces cal-
A.B., Almonte, A.G., Wainer, B.H., and cium waves in cultured astrocytes: Long-range
Dingledine, R. (2003). Neuronal and glial glial signaling. Science 247, 470473.
pathological changes during epileptogenesis Dallerac, G., Chever, O., and Rouach, N. (2013).
in the mouse pilocarpine model. Exp. Neurol. How do astrocytes shape synaptic transmis-
182, 2134. sion? Insights from electrophysiology. Front Cell
Borges, K., McDermott, D., Irier, H., Smith, Y., and Neurosci. 7, 159.
Dingledine, R. (2006). Degeneration and prolif- DAscenzo, M., Fellin, T., Terunuma, M., Revilla-
eration of astrocytes in the mouse dentate gyrus Sanchez, R., Meaney, D.F., Auberson, Y.P., Moss,
after pilocarpine-induced status epilepticus. S.J., and Haydon, P.G. (2007). mGluR5 stimu-
Exp. Neurol. 201, 416427. lates gliotransmission in the nucleus accumbens.
Bowser, D.N., and Khakh, B.S. (2007). Vesicular ATP Proc. Natl. Acad. Sci. USA 104, 19952000.
is the predominant cause of intercellular calcium de Lanerolle, N.C., Lee, T.S., and Spencer, D.D. (2012).
waves in astrocytes. J. Gen. Physiol 129, 485491. Histopathology of human epilepsy. In Jaspers
Brown, R.E., Basheer, R., McKenna, J.T., Strecker, Basic Mechanisms of the Epilepsies, J.L.Noebels,
R.E., and McCarley, R.W. (2012). Control of sleep M.Avoli, M.A.Rogawski, R.W.Olsen, and
and wakefulness. Physiol. Rev. 92, 10871187. A.V.Delgado-Escueta, eds. (New York: Oxford
Bushong, E.A., Martone, M.E., Jones, Y.Z., and University Press), pp. 387404.
Ellisman, M.H. (2002). Protoplasmic astrocytes Deng, Q., Terunuma, M., Fellin, T., Moss, S.J., and
in CA1 stratum radiatum occupy separate ana- Haydon, P.G. (2011). Astrocytic activation of
tomical domains. J. Neurosci. 22, 183192. A1 receptors regulates the surface expression of
Cao, X., Li, L.P., Wang, Q., Wu, Q., Hu, H.H., Zhang, NMDA receptors through a Src kinase depen-
M., Fang, Y.Y., Zhang, J., Li, S.J., Xiong, W.C., dent pathway. Glia 59, 10841093.
Yan, H.C., Gao, Y.B., Liu, J.H., Li, X.W., Sun, L.R., Di Castro, M.A., Chuquet, J., Liaudet, N.,
Zeng, Y.N., Zhu, X.H., and Gao, T.M. (2013). Bhaukaurally, K., Santello, M., Bouvier, D., Tiret,
Astrocyte-derived ATP modulates depressive- P., and Volterra, A. (2011). Local Ca2+ detection
like behaviors. Nat. Med. 19, 773777. and modulation of synaptic release by astrocytes.
Chuquet, J., Hollender, L., and Nimchinsky, E.A. Nat. Neurosci. 14, 12761284.
(2007). High-resolution in vivo imaging of the Ding, S., Fellin, T., Zhu, Y., Lee, S.Y., Auberson, Y.P.,
neurovascular unit during spreading depression. Meaney, D.F., Coulter, D.A., Carmignoto, G.,
J. Neurosci. 27, 40364044. and Haydon, P.G. (2007). Enhanced astrocytic
Clasadonte, J., Dong, J., Hines, D.J., and Haydon, Ca2+ signals contribute to neuronal excito-
P.G. (2013). Astrocyte control of synaptic toxicity after status epilepticus. J. Neurosci. 27,
NMDA receptors contributes to the progressive 1067410684.
92 Part II: Homeostatic Control

Dombeck, D.A., Khabbaz, A.N., Collman, F., Fiacco, T.A., Agulhon, C., Taves, S.R., Petravicz, J.,
Adelman, T.L., and Tank, D.W. (2007). Imaging Casper, K.B., Dong, X., Chen, J., and McCarthy,
large-scale neural activity with cellular resolu- K.D. (2007). Selective stimulation of astrocyte
tion in awake, mobile mice. Neuron 56, 4357. calcium in situ does not affect neuronal excit-
Dunwiddie, T.V. (1999). Adenosine and suppression atory synaptic activity. Neuron 54, 611626.
of seizures. Adv. Neurol. 79, 10011010. Fiacco, T.A., and McCarthy, K.D. (2004). Intracellular
During, M.J. and Spencer, D.D. (1992). Adenosine:A astrocyte calcium waves in situ increase the fre-
potential mediator of seizure arrest and postictal quency of spontaneous AMPA receptor currents
refractoriness. Ann. Neurol. 32, 618624. in CA1 pyramidal neurons. J. Neurosci. 24,
Eid, T., Behar, K., Dhaher, R., Bumanglag, A.V., 722732.
and Lee, T.S. (2012). Roles of glutamine synthe- Filosa, J.A., Bonev, A.D., Straub, S.V., Meredith, A.L.,
tase inhibition in epilepsy. Neurochem. Res. 37, Wilkerson, M.K., Aldrich, R.W., and Nelson,
23392350. M.T. (2006) Local potassium signaling couples
Eid, T., Ghosh, A., Wang, Y., Beckstrom, H., Zaveri, neuronal activity to vasodilation in the brain.
H.P., Lee, T.S., Lai, J.C., Malthankar-Phatak, Nat Neurosci. 9, 13971403.
G.H., and de Lanerolle, N.C. (2008). Recurrent Florian, C., Vecsey, C.G., Halassa, M.M., Haydon,
seizures and brain pathology after inhibition P.G., and Abel, T. (2011). Astrocyte-derived
of glutamine synthetase in the hippocampus in adenosine and A1 receptor activity contribute to
rats. Brain 131, 20612070. sleep loss-induced deficits in hippocampal syn-
Eid, T., Thomas, M.J., Spencer, D.D., Runden- aptic plasticity and memory in mice. J. Neurosci.
Pran, E., Lai, J.C., Malthankar, G.V., Kim, J.H., 31, 69566962.
Danbolt, N.C., Ottersen, O.P., and de Lanerolle, Fredholm, B.B., Chen, J.F., Cunha, R.A.,
N.C. (2004). Loss of glutamine synthetase in the Svenningsson, P., and Vaugeois, J.M. (2005).
human epileptogenic hippocampus: Possible Adenosine and brain function. Int. Rev.
mechanism for raised extracellular glutamate in Neurobiol. 63, 191270.
mesial temporal lobe epilepsy. Lancet 363, 2837. Genoud, C., Quairiaux, C., Steiner, P., Hirling, H.,
Etherington, L.A., and Frenguelli, B.G. (2004). Welker, E., and Knott, G.W. (2006). Plasticity of
Endogenous adenosine modulates epileptiform astrocytic coverage and glutamate transporter
activity in rat hippocampus in a receptor sub- expression in adult mouse cortex. PLoS Biol.
type-dependent manner. Eur. J. Neurosci. 19, 4, e343.
25392550. Georgiev, V., Johansson, B., and Fredholm, B.B.
Farina, C., Aloisi, F., and Meinl, E. (2007). Astrocytes (1993). Long-term caffeine treatment leads to
are active players in cerebral innate immunity. a decreased susceptibility to NMDA-induced
Trends Immunol. 28, 138145. clonic seizures in mice without changes in
Fedele, D.E., Gouder, N., Guttinger, M., Gabernet, L., adenosine A1 receptor number. Brain Res. 612,
Scheurer, L., Rulicke, T., Crestani, F., and Boison, 271277.
D. (2005). Astrogliosis in epilepsy leads to over- Giaume, C., Koulakoff, A., Roux, L., Holcman, D.,
expression of adenosine kinase, resulting in sei- and Rouach, N. (2010). Astroglial networks: a
zure aggravation. Brain 128, 23832395. step further in neuroglial and gliovascular inter-
Fellin, T., Gomez-Gonzalo, M., Gobbo, S., actions. Nat Rev Neurosci. 11, 8799.
Carmignoto, G., and Haydon, P.G. (2006). Gomes, C.V., Kaster, M.P., Tom, A.R., Agostinho,
Astrocytic glutamate is not necessary for the P.M., and Cunha, R.A. (2011). Adenosine recep-
generation of epileptiform neuronal activ- tors and brain diseases: neuroprotection and
ity in hippocampal slices. J. Neurosci. 26, neurodegeneration. Biochim. Biophys. Acta.
93129322. 1808, 13801399.
Fellin, T., Halassa, M.M., Terunuma, M., Succol, F., Gmez-Gonzalo, M., Losi, G., Chiavegato, A.,
Takano, H., Frank, M., Moss, S.J., and Haydon, Zonta, M., Cammarota, M., Brondi, M., Vetri, F.,
P.G. (2009). Endogenous nonneuronal modula- Uva, L., Pozzan, T., de Curtis, M., Ratto, G.M.,
tors of synaptic transmission control cortical Carmignoto, G. (2010). An excitatory loop with
slow oscillations in vivo. Proc. Natl. Acad. Sci. astrocytes contributes to drive neurons to sei-
USA 106, 1503715042. zure threshold. PLoS Biol. 8, e1000352.
Fellin, T., Pascual, O., Gobbo, S., Pozzan, T., Haydon, Gordon, G.R., Baimoukhametova, D.V., Hewitt,
P.G., and Carmignoto, G. (2004). Neuronal S.A., Rajapaksha, W.R., Fisher, T.E., and Bains,
synchrony mediated by astrocytic glutamate J.S. (2005). Norepinephrine triggers release of
through activation of extrasynaptic NMDA glial ATP to increase postsynaptic efficacy. Nat.
receptors. Neuron 43, 729743. Neurosci. 8, 10781086.
Role of Astrocytes in Sleep and Epilepsy 93

Gordon, G.R., Choi, H.B., Rungta, R.L., Ellis- Hamilton, N.B., and Attwell, D. (2010). Do astro-
Davies, G.C., and MacVicar, B.A. (2008). Brain cytes really exocytose neurotransmitters? Nat.
metabolism dictates the polarity of astrocyte Rev. Neurosci. 11, 227238.
control over arterioles. Nature 7223, 745749. Hammer, J., Alvestad, S., Osen, K.K., Skare, O.,
Gordon, G.R., Iremonger, K.J., Kantevari, S., Ellis- Sonnewald, U., and Ottersen, O.P. (2008).
Davies, G.C., MacVicar, B.A., and Bains J.S. Expression of glutamine synthetase and glu-
(2009). Astrocyte-mediated distributed plastic- tamate dehydrogenase in the latent phase and
ity at hypothalamic glutamate synapses. Neuron chronic phase in the kainate model of temporal
64, 391403. lobe epilepsy. Glia 56, 856868.
Gourine, A.V., Dale, N., Llaudet, E., Poputnikov, Han, J., Kesner, P., Metna-Laurent, M., Duan, T.,
D.M., Spyer, K.M., and Gourine, V.N. (2007). Xu, L., Georges, F., Koehl, M., Abrous, D.N.,
Release of ATP in the central nervous system Mendizabal-Zubiaga, J., Grandes, P., Liu, Q., Bai,
during systemic inflammation: real-time mea- G., Wang, W., Xiong, L., Ren, W., Marsicano,
surement in the hypothalamus of conscious rab- G., and Zhang, X. (2012). Acute cannabinoids
bits. J. Physiol. 585, 305316. impair working memory through astroglial CB1
Gourine, A.V., Kasymov, V., Marina, N., Tang, F., receptor modulation of hippocampal LTD. Cell
Figueiredo, M.F., Lane, S., Teschemacher, A.G., 148, 10391050.
Spyer, K.M., Deisseroth, K., and Kasparov, S. Han, K.S., Woo, J., Park, H., Yoon, B.J., Choi, S., and
(2010). Astrocytes control breathing through pH- Lee, C.J. (2013). Channel-mediated astrocytic
dependent release of ATP. Science 329, 571575. glutamate release via Bestrophin-1 targets syn-
Gucek, A., Vardjan, N., and Zorec, R. (2012). aptic NMDARs. Mol. Brain6, 4.
Exocytosis in astrocytes:Transmitter release and Hemmeter, U.M., Hemmeter-Spernal, J., and Krieg,
membrane signal regulation. Neurochem. Res. J.C. (2010). Sleep deprivation in depression.
37, 23512363. Expert. Rev. Neurother. 10, 11011115.
Guthrie, P.B., Knappenberger, J., Segal, M., Bennett, Henneberger, C., Papouin, T., Oliet, S.H., and
M.V., Charles, A.C., and Kater, S.B. (1999). ATP Rusakov, D.A. (2010). Long-term potentiation
released from astrocytes mediates glial calcium depends on release of D-serine from astrocytes.
waves. J. Neurosci. 19, 520528. Nature 463, 232236.
Haber, M., Zhou, L., and Murai, K.K. (2006). Hines, D.J., Schmitt, L.I., Hines, R.M., Moss, S.J.,
Cooperative astrocyte and dendritic spine and Haydon, P.G. (2013). Antidepressant effects
dynamics at hippocampal excitatory synapses. J. of sleep deprivation require astrocyte-dependent
Neurosci. 26, 88818891. adenosine mediated signaling. Transl. Psychiatry
Haberle, J., Gorg, B., Rutsch, F., Schmidt, E., Toutain, 3, e212.
A., Benoist, J.F., Gelot, A., Suc, A.L., Hohne, Hirase, H., Qian, L., Bartho, P., and Buzsaki, G.
W., Schliess, F., Haussinger, D., and Koch, H.G. (2004). Calcium dynamics of cortical astrocytic
(2005). Congenital glutamine deficiency with networks in vivo. PLoS Biol. 2, E96.
glutamine synthetase mutations. N. Engl. J.Med. Hirrlinger, J., Hulsmann, S., and Kirchhoff, F.
353, 19261933. (2004). Astroglial processes show spontaneous
Haberle, J., Gorg, B., Toutain, A., Rutsch, F., Benoist, motility at active synaptic terminals in situ. Eur.
J.F., Gelot, A., Suc, A.L., Koch, H.G., Schliess, F., J.Neurosci. 20, 22352239.
and Haussinger, D. (2006). Inborn error of amino Jacobson, K.A., von Lubitz, D.K., Daly, J.W., and
acid synthesis: Human glutamine synthetase Fredholm, B.B. (1996). Adenosine receptor
deficiency. J. Inherit. Metab. Dis. 29, 352358. ligands: Differences with acute versus chronic
Halassa, M.M., Fellin, T., Takano, H., Dong, J.H., and treatment. Trends Pharmacol. Sci. 17, 108113.
Haydon, P.G. (2007). Synaptic islands defined by Jimenez-Gonzalez, C., Pirttimaki, T., Cope, D.W.,
the territory of a single astrocyte. J. Neurosci. 27, and Parri, H.R. (2011). Non-neuronal, slow
64736477. GABA signalling in the ventrobasal thalamus
Halassa, M.M., Florian, C., Fellin, T., Munoz, J.R., targets delta-subunit-containing GABA(A)
Lee, S.Y., Abel, T., Haydon, P.G., and Frank, M.G. receptors. Eur. J.Neurosci. 33, 14711482.
(2009). Astrocytic modulation of sleep homeo- Johansson, B., Georgiev, V., Kuosmanen, T., and
stasis and cognitive consequences of sleep loss. Fredholm, B.B. (1996). Long-term treatment with
Neuron 61, 213219. some methylxanthines decreases the susceptibil-
Halassa, M.M., and Haydon, P.G. (2010). Integrated ity to bicuculline-and pentylenetetrazol-induced
brain circuits: Astrocytic networks modulate seizures in mice: Relationship to c-fos expres-
neuronal activity and behavior. Annu. Rev. sion and receptor binding. Eur. J. Neurosci. 8,
Physiol 72, 335355. 24472458.
94 Part II: Homeostatic Control

Jourdain, P., Bergersen, L.H., Bhaukaurally, K., (2012). Neuronal adenosine release, and not
Bezzi, P., Santello, M., Domercq, M., Matute, astrocytic ATP release, mediates feedback inhi-
C., Tonello, F., Gundersen, V., and Volterra, A. bition of excitatory activity. Proc. Natl. Acad. Sci.
(2007). Glutamate exocytosis from astrocytes USA 109, 62656270.
controls synaptic strength. Nat. Neurosci. 10, Malarkey, E.B., and Parpura, V. (2008). Mechanisms
331339. of glutamate release from astrocytes. Neurochem.
Kam, K., and Nicoll, R. (2007). Excitatory synap- Int. 52, 142154.
tic transmission persists independently of the Martin, E.D., Fernandez, M., Perea, G., Pascual, O.,
glutamate-glutamine cycle. J. Neurosci. 27, Haydon, P.G., Araque, A., and Cena, V. (2007).
91929200. Adenosine released by astrocytes contributes to
Kang, J., Jiang, L., Goldman, S.A., and Nedergaard, hypoxia-induced modulation of synaptic trans-
M. (1998). Astrocyte-mediated potentiation of mission. Glia 55, 3645.
inhibitory synaptic transmission. Nat. Neurosci. Martineau, M., Shi, T., Puyal, J., Knolhoff, A.M.,
1, 683692. Dulong, J., Gasnier, B., Klingauf, J., Sweedler,
Kozlov, A.S., Angulo, M.C., Audinat, E., and J.V., Jahn, R., and Mothet, J.P. (2013). Storage and
Charpak, S. (2006). Target cell-specific modu- uptake of D-serine into astrocytic synaptic-like
lation of neuronal activity by astrocytes. Proc. vesicles specify gliotransmission. J. Neurosci. 33,
Natl. Acad. Sci. USA 103, 1005810063. 34133423.
Krueger, J.M. (2008). The role of cytokines in sleep Masino, S.A., Diao, L., Illes, P., Zahniser, N.R.,
regulation. Curr. Pharm. Des. 14, 34083416. Larson, G.A., Johansson, B., Fredholm, B.B.,
Kuga, N., Sasaki, T., Takahara, Y., Matsuki, N., and and Dunwiddie, T.V. (2002). Modulation of hip-
Ikegaya, Y. (2011). Large-scale calcium waves pocampal glutamatergic transmission by ATP
traveling through astrocytic networks in vivo. J. is dependent on adenosine a(1) receptors. J.
Neurosci. 31, 26072614. Pharmacol. Exp. Ther. 303, 356363.
Le Meur, K., Mendizabal-Zubiaga, J., Grandes, P., and McCoy, J.G., and Strecker, R.E. (2011). The cognitive
Audinat, E. (2012). GABA release by hippocam- cost of sleep lost. Neurobiol. Learn. Mem. 96,
pal astrocytes. Front. Comput. Neurosci.6, 59. 564582.
Lee, S., Yoon, B.E., Berglund, K., Oh, S.J., Park, H., Metea, M.R., and Newman, E.A. (2006). Glial cells
Shin, H.S., Augustine, G.J., and Lee, C.J. (2010). dilate and constrict blood vessels: A mecha-
Channel-mediated tonic GABA release from nism of neurovascular coupling. J. Neurosci. 26,
glia. Science 330, 790796. 28622870.
Li, T., Lytle, N., Lang, G.Q., Sandau, U.S., and Boison, Montana, V., Ni, Y., Sunjara, V., Hua, X., and
D. (2012). Local disruption of glial adenosine Parpura, V. (2004). Vesicular glutamate trans-
homeostasis in mice associates with focal elec- porter-dependent glutamate release from astro-
trographic seizures: A first step in epileptogen- cytes. J. Neurosci. 24, 26332642.
esis? Glia 60, 8395. Mothet, J.P., Parent, A.T., Wolosker, H., Brady, R.O.,
Li, T., Ren, G., Lusardi, T., Wilz, A., Lan, J.Q., Jr., Linden, D.J., Ferris, C.D., Rogawski, M.A.,
Iwasato, T., Itohara, S., Simon, R.P., and Boison, and Snyder, S.H. (2000). D-serine is an endog-
D. (2008). Adenosine kinase is a target for the enous ligand for the glycine site of the N-methyl-
prediction and prevention of epileptogenesis in D-aspartate receptor. Proc. Natl. Acad. Sci. USA
mice. J. Clin. Invest 118, 571582. 97, 49264931.
Liang, S.L., Carlson, G.C., and Coulter, D.A. (2006). Mothet, J.P., Pollegioni, L., Ouanounou, G.,
Dynamic regulation of synaptic GABA release by Martineau, M., Fossier, P., and Baux, G. (2005).
the glutamate-glutamine cycle in hippocampal Glutamate receptor activation triggers a cal-
area CA1. J. Neurosci. 26, 85378548. cium-dependent and SNARE protein-dependent
Liu, Q.S., Xu, Q., Arcuino, G., Kang, J., and release of the gliotransmitter D-serine. Proc.
Nedergaard, M. (2004a). Astrocyte-mediated Natl. Acad. Sci. USA 102, 56065611.
activation of neuronal kainate receptors. Proc. Mller, C.J., Groticke, I., Bankstahl, M., and Loscher,
Natl. Acad. Sci. US A 101, 31723177. W. (2009). Behavioral and cognitive alterations,
Liu, Q.S., Xu, Q., Kang, J., and Nedergaard, M. spontaneous seizures, and neuropathology
(2004b). Astrocyte activation of presynaptic developing after a pilocarpine-induced status
metabotropic glutamate receptors modulates epilepticus in C57BL/6 mice. Exp. Neurol. 219,
hippocampal inhibitory synaptic transmission. 284297.
Neuron Glia Biol. 1, 307316. Mulligan, S.J., and MacVicar, B.A. (2004). Calcium
Lovatt, D., Xu, Q., Liu, W., Takano, T., Smith, N.A., transients in astrocyte endfeet cause cerebrovas-
Schnermann, J., Tieu, K., and Nedergaard, M. cular constrictions. Nature 431, 195199.
Role of Astrocytes in Sleep and Epilepsy 95

Nadjar, A., Blutstein, T., Aubert, A., Laye, S., and Panatier, A., Vallee, J., Haber, M., Murai, K.K.,
Haydon, P.G. (2013). Astrocyte-derived adenos- Lacaille, J.C., and Robitaille, R. (2011). Astrocytes
ine modulates increased sleep pressure during are endogenous regulators of basal transmission
inflammatory response. Glia 61, 724731. at central synapses. Cell 146, 785798.
Navarrete, M., and Araque, A. (2010). Papouin, T., Ladepeche, L., Ruel, J., Sacchi, S.,
Endocannabinoids potentiate synaptic transmis- Labasque, M., Hanini, M., Groc, L., Pollegioni,
sion through stimulation of astrocytes. Neuron L., Mothet, J.P., and Oliet, S.H. (2012). Synaptic
68, 113126. and extrasynaptic NMDA receptors are gated
Navarrete, M., Perea, G., Fernandez, d.S., Gomez- by different endogenous coagonists. Cell 150,
Gonzalo, M., Nunez, A., Martin, E.D., and 633646.
Araque, A. (2012). Astrocytes mediate in vivo Parkinson, F.E., Xiong, W., Zamzow, C.R., Chestley,
cholinergic-induced synaptic plasticity. PLoS T., Mizuno, T., and Duckworth, M.L. (2009).
Biol. 10, e1001259. Transgenic expression of human equilibrative
Navarrete, M., Perea, G., Maglio, L., Pastor, J., Garcia nucleoside transporter 1 in mouse neurons. J.
de Sola, R., and Araque, A. (2013). Astrocyte Neurochem. 109, 562572.
calcium signal and gliotransmission in human Parpura, V., Basarsky, T.A., Liu, F., Jeftinija, K.,
brain tissue. Cereb. Cortex 23, 12401246. Jeftinija, S., and Haydon, P.G. (1994). Glutamate-
Newman, E.A. (2003). Glial cell inhibition of neurons mediated astrocyte-neuron signalling. Nature
by release of ATP. J. Neurosci. 23, 16591666. 369, 744747.
Nie, H., Zhang, H., and Weng, H.R. (2010). Parpura, V., Heneka, M.T., Montana, V., Oliet, S.H.,
Bidirectional neuron-glia interactions triggered Schousboe, A., Haydon, P.G., Stout, R.F., Jr., Spray,
by deficiency of glutamate uptake at spinal sen- D.C., Reichenbach, A., Pannicke, T., Pekny, M.,
sory synapses. J. Neurophysiol. 104, 713725. Pekna, M., Zorec, R., and Verkhratsky, A. (2012).
Nimmerjahn, A., Mukamel, E.A., and Schnitzer, M.J. Glial cells in (patho)physiology. J. Neurochem.
(2009). Motor behavior activates Bergmann glial 121, 427.
networks. Neuron 62, 400412. Parpura, V., and Verkhratsky, A. (2012). Homeostatic
Oberheim, N.A., Takano, T., Han, X., He, W., Lin, function of astrocytes:Ca(2+) and Na(+) signal-
J.H., Wang, F., Xu, Q., Wyatt, J.D., Pilcher, W., ling. Transl. Neurosci. 3, 334344.
Ojemann, J.G., Ransom, B.R., Goldman, S.A., Parri, H.R., Gould, T.M., and Crunelli, V. (2001).
and Nedergaard, M. (2009). Uniquely hominid Spontaneous astrocytic Ca2+ oscillations in situ
features of adult human astrocytes. J. Neurosci. drive NMDAR-mediated neuronal excitation.
29, 32763287. Nat. Neurosci. 4, 803812.
Oberheim, N.A., Tian, G.F., Han, X., Peng, W., Pascual, O., Casper, K.B., Kubera, C., Zhang, J.,
Takano, T., Ransom, B., and Nedergaard, M. Revilla-Sanchez, R., Sul, J.Y., Takano, H., Moss,
(2008). Loss of astrocytic domain organization S.J., McCarthy, K., and Haydon, P.G. (2005).
in the epileptic brain. J. Neurosci. 28, 32643276. Astrocytic purinergic signaling coordinates syn-
Oliet, S.H., and Bonfardin, V.D. (2010). Morphological aptic networks. Science 310, 113116.
plasticity of the rat supraoptic nucleuscellular Pasti, L., Volterra, A., Pozzan, T., and Carmignoto,
consequences. Eur. J.Neurosci. 32, 19891994. G. (1997). Intracellular calcium oscillations in
Ortinski, P.I., Dong, J., Mungenast, A., Yue, C., astrocytes:Ahighly plastic, bidirectional form of
Takano, H., Watson, D.J., Haydon, P.G., and communication between neurons and astrocytes
Coulter, D.A. (2010). Selective induction of astro- in situ. J. Neurosci. 17, 78177830.
cytic gliosis generates deficits in neuronal inhibi- Pekny, M. and Nilsson, M. (2005). Astrocyte activa-
tion. Nat. Neurosci. 13, 584591. tion and reactive gliosis. Glia 50, 427434.
Otis, T.S., and Jahr, C.E. (1998). Anion currents and Perea, G., and Araque, A. (2007). Astrocytes poten-
predicted glutamate flux through a neuronal glu- tiate transmitter release at single hippocampal
tamate transporter. J. Neurosci. 18, 70997110. synapses. Science 317, 10831086.
Palchykova, S., Winsky-Sommerer, R., Shen, H.Y., Perea, G., Navarrete, M., and Araque, A. (2009).
Boison, D., Gerling, A., and Tobler, I. (2010). Tripartite synapses:Astrocytes process and con-
Manipulation of adenosine kinase affects sleep trol synaptic information. Trends Neurosci. 32,
regulation in mice. J. Neurosci. 30, 1315713165. 421431.
Panatier, A., Theodosis, D.T., Mothet, J.P., Touquet, Petravicz, J., Fiacco, T.A., and McCarthy, K.D.
B., Pollegioni, L., Poulain, D.A., and Oliet, S.H. (2008). Loss of IP3 receptor-dependent Ca2+
(2006). Glia-derived D-serine controls NMDA increases in hippocampal astrocytes does not
receptor activity and synaptic memory. Cell 125, affect baseline CA1 pyramidal neuron synaptic
775784. activity. J. Neurosci. 28, 49674973.
96 Part II: Homeostatic Control

Petzold, G.C., Albeanu, D.F., Sato, T.F., and Murthy, Shigetomi, E., Jackson-Weaver, O., Huckstepp, R.T.,
V.N. (2008). Coupling of neural activity to blood ODell, T.J., and Khakh, B.S. (2013). TRPA1 chan-
flow in olfactory glomeruli is mediated by astro- nels are regulators of astrocyte basal calcium lev-
cytic pathways. Neuron 58, 897910. els and long-term potentiation via constitutive
Pirttimaki, T., Parri, H.R., and Crunelli, V. (2013). D-serine release. J. Neurosci. 33, 1014310153.
Astrocytic GABA transporter GAT-1 dysfunc- Shigetomi, E., Tong, X., Kwan, K.Y., Corey, D.P.,
tion in experimental absence seizures. J. Physiol. and Khakh, B.S. (2011). TRPA1 channels regu-
591, 823833. late astrocyte resting calcium and inhibitory
Pirttimaki, T.M. and Parri, H.R. (2013). Astrocyte synapse efficacy through GAT-3. Nat. Neurosci.
plasticity: Implications for synaptic and neuro- 15, 7080.
nal activity. Neuroscientist 19, 604615. Sofroniew, M.V. (2009). Molecular dissection of reac-
Porter, J.T., and McCarthy, K.D. (1996). Hippocampal tive astrogliosis and glial scar formation. Trends
astrocytes in situ respond to glutamate released Neurosci. 32, 638647.
from synaptic terminals. J. Neurosci. 16, Sofroniew, M.V. and Vinters, H.V. (2010).
50735081. Astrocytes: Biology and pathology. Acta
Reyes-Haro, D., Mller, J., Boresch, M., Pivneva, Neuropathol. 119, 735.
T., Benedetti, B., Scheller, A., Nolte, C., and Sontheimer, H., Black, J.A., and Waxman, S.G. (1996).
Kettenmann, H. (2010). Neuron-astrocyte inter- Voltage-gated Na+ channels in glia:Properties and
actions in the medial nucleus of the trapezoid possible functions. Trends Neurosci. 19, 325331.
body. J. Gen. Physiol 135, 583594. Spencer, S.S. (2002). Neural networks in human
Rigoulot, M-A., Leroy, C., Koning, E., Ferrandon, epilepsy:Evidence of and implications for treat-
A., and Nehlig, A. (2003). Prolonged low-dose ment. Epilepsia 43, 219227.
caffeine exposure protects against hippocampal Steinhauser, C., and Seifert, G. (2002). Glial mem-
damage but not against the occurrence of epi- brane channels and receptors in epilepsy:Impact
lepsy in the lithium-pilocarpine model in the rat. for generation and spread of seizure activity. Eur.
Epilepsia 44, 529535. J.Pharmacol. 447, 227237.
Robel, S., Berninger, B., and Gotz, M. (2011). The Steriade, M. (1997). Synchronized activities of cou-
stem cell potential of glia:Lessons from reactive pled oscillators in the cerebral cortex and thala-
gliosis. Nat. Rev. Neurosci. 12, 88104. mus at different levels of vigilance. Cereb. Cortex
Rohleder, N., Aringer, M., and Boentert, M. (2012). 7, 583604.
Role of interleukin-6 in stress, sleep, and fatigue. Steriade, M. (2006). Grouping of brain rhythms in
Ann. NY Acad. Sci. 1261, 8896. corticothalamic systems. Neuroscience 137,
Sawada, K., Echigo, N., Juge, N., Miyaji, T., Otsuka, 10871106.
M., Omote, H., Yamamoto, A., and Moriyama, Sun, W., McConnell, E., Pare, J.F., Xu, Q., Chen,
Y. (2008). Identification of a vesicular nucleo- M., Peng, W., Lovatt, D., Han, X., Smith, Y., and
tide transporter. Proc. Natl. Acad. Sci. USA 105, Nedergaard, M. (2013). Glutamate-dependent
56835686. neuroglial calcium signaling differs between
Schmitt, L.I., Sims, R.E., Dale, N., and Haydon, P.G. young and adult brain. Science 339, 197200.
(2012). Wakefulness affects synaptic and network Swinkels, W.A., Kuyk, J., van Dyck, R., and
activity by increasing extracellular astrocyte- Spinhoven, P. (2005). Psychiatric comorbidity in
derived adenosine. J. Neurosci. 32, 44174425. epilepsy. Epilepsy Behav. 7, 3750.
Schummers, J., Yu, H., and Sur, M. (2008). Tuned Takano, T., Tian, G.F., Peng, W., Lou, N., Libionka,
responses of astrocytes and their influence W., Han, X., and Nedergaard, M. (2006).
on hemodynamic signals in the visual cortex. Astrocyte-mediated control of cerebral blood
Science 320, 16381643. flow. Nat. Neurosci. 9, 260267.
Seifert, G., Carmignoto, G., and Steinhauser, C. Takata, N., Mishima, T., Hisatsune, C., Nagai, T.,
(2010). Astrocyte dysfunction in epilepsy. Brain Ebisui, E., Mikoshiba, K., and Hirase, H. (2011).
Res. Rev. 63, 212221. Astrocyte calcium signaling transforms cholin-
Serrano, A., Haddjeri, N., Lacaille, J.C., and ergic modulation to cortical plasticity in vivo. J.
Robitaille, R. (2006). GABAergic network activa- Neurosci. 31, 1815518165.
tion of glial cells underlies hippocampal hetero- Takata, N., Nagai, T., Ozawa, K., Oe, Y., Mikoshiba,
synaptic depression. J. Neurosci. 26, 53705382. K., and Hirase, H. (2013). Cerebral blood flow
Shapiro, L.A., Wang, L., and Ribak, C.E. (2008). modulation by Basal forebrain or whisker stimu-
Rapid astrocyte and microglial activation fol- lation can occur independently of large cytosolic
lowing pilocarpine-induced seizures in rats. Ca2+ signaling in astrocytes. PLoS One 8(6),
Epilepsia 49(Suppl 2), 3341. e66525.
Role of Astrocytes in Sleep and Epilepsy 97

Tang, F.R., and Lee, W.L. (2001). Expression of Wilhelmsson, U., Li, L., Pekna, M., Berthold, C.H.,
the group II and III metabotropic glutamate Blom, S., Eliasson, C., Renner, O., Bushong,
receptors in the hippocampus of patients with E., Ellisman, M., Morgan, T.E., and Pekny, M.
mesial temporal lobe epilepsy. J. Neurocytol. 30, (2004). Absence of glial fibrillary acidic protein
137143. and vimentin prevents hypertrophy of astrocytic
Tian, G.F., Azmi, H., Takano, T., Xu, Q., Peng, W., Lin, processes and improves post-traumatic regen-
J., Oberheim, N., Lou, N., Wang, X., Zielke, H.R., eration. J. Neurosci. 24, 50165021.
Kang, J., and Nedergaard, M. (2005). An astro- Winship, I.R., Plaa, N., and Murphy, T.H. (2007).
cytic basis of epilepsy. Nat. Med. 11, 973981. Rapid astrocyte calcium signals correlate with
Tong, X., Shigetomi, E., Looger, L.L., and Khakh, neuronal activity and onset of the hemodynamic
B.S. (2013). Genetically encoded calcium indi- response in vivo. J. Neurosci. 27, 62686272.
cators and astrocyte calcium microdomains. Woo, D.H., Han, K.S., Shim, J.W., Yoon, B.E., Kim,
Neuroscientist 19, 274291. E., Bae, J.Y., Oh, S.J., Hwang, E.M., Marmorstein,
Velez-Fort, M., Audinat, E., and Angulo, M.C. A.D., Bae, Y.C., Park, J.Y., and Lee, C.J. (2012).
(2012). Central role of GABA in neuron-glia TREK-1 and Best1 channels mediate fast and
interactions. Neuroscientist 18, 237250. slow glutamate release in astrocytes upon GPCR
Verkhratsky, A., Parpura, V., and Rodriguez, J.J. activation. Cell 151, 2540.
(2011). Where the thoughts dwell:The physiology Yoo, S.S., Hu, P.T., Gujar, N., Jolesz, F.A., and Walker,
of neuronal-glial diffuse neural net. Brain Res. M.P. (2007). A deficit in the ability to form new
Rev. 66, 133151. human memories without sleep. Nat. Neurosci.
Volterra, A. and Meldolesi, J. (2005). Astrocytes, 10, 385392.
from brain glue to communication elements:The Zhang, D., Xiong, W., Chu, S., Sun, C., Albensi, B.C.,
revolution continues. Nat. Rev. Neurosci. 6, and Parkinson, F.E. (2012). Inhibition of hippo-
626640. campal synaptic activity by ATP, hypoxia or oxy-
Von Lubitz, D.K., Paul, I.A., Ji, X.D., Carter, M., and gen-glucose deprivation does not require CD73.
Jacobson, K.A. (1994). Chronic adenosine A1 PLoS One 7, e39772.
receptor agonist and antagonist:Effect on recep- Zhang, J.M., Wang, H.K., Ye, C.Q., Ge, W., Chen,
tor density and N-methyl-D-aspartate induced Y., Jiang, Z.L., Wu, C.P., Poo, M.M., and Duan,
seizures in mice. Eur. J.Pharmacol. 253, 9599. S. (2003). ATP released by astrocytes mediates
Vyazovskiy, V.V., Cirelli, C., Pfister-Genskow, M., glutamatergic activity-dependent heterosynaptic
Faraguna, U., and Tononi, G. (2008) Molecular suppression. Neuron 40, 971982.
and electrophysiological evidence for net synap- Zhang, Q., Fukuda, M., Van Bockstaele, E., Pascual,
tic potentiation in wake and depression in sleep. O., and Haydon, P.G. (2004). Synaptotagmin
Nat. Neurosci. 11, 200208. IV regulates glial glutamate release. Proc. Natl.
Wall, M.J., and Dale, N. (2013). Neuronal transporter Acad. Sci. USA 101, 94419446.
and astrocytic ATP exocytosis underlie activity- Zhang, Z., Chen, G., Zhou, W., Song, A., Xu, T., Luo,
dependent adenosine release in the hippocam- Q., Wang, W., Gu, X.S., and Duan, S. (2007).
pus. J. Physiol. 591, 38533871. Regulated ATP release from astrocytes through
Wang, F., Smith, N.A., Xu, Q., Goldman, S., Peng, lysosome exocytosis. Nat. Cell Biol. 9, 945953.
W., Huang, J.H., Takano, T., and Nedergaard, M. Zielinski, M.R., Taishi, P., Clinton, J.M., and Krueger,
(2013). Photolysis of caged Ca2+ but not recep- J.M. (2012). 5-Ectonucleotidase-knockout mice
tor-mediated Ca2+ signaling triggers astrocytic lack non-REM sleep responses to sleep depriva-
glutamate release. J. Neurosci. 33, 1740417412. tion. Eur. J.Neurosci. 35, 17891798.
Wang, X., Lou, N., Xu, Q., Tian, G.F., Peng, W.G., Zimmermann, H. (2000). Extracellular metabo-
Han, X., Kang, J., Takano, T., and Nedergaard, M. lism of ATP and other nucleotides. Naunyn
(2006). Astrocytic Ca2+ signaling evoked by sen- Schmiedebergs Arch. Pharmacol. 362, 299309.
sory stimulation in vivo. Nat. Neurosci. 9, 816823. Zonta, M., Angulo, M.C., Gobbo, S., Rosengarten, B.,
Wang, Y., Zaveri, H.P., Lee, T.S., and Eid, T. (2009). Hossmann, K.A., Pozzan, T., and Carmignoto,
The development of recurrent seizures after con- G. (2003). Neuron-to-astrocyte signaling is cen-
tinuous intrahippocampal infusion of methio- tral to the dynamic control of brain microcircu-
nine sulfoximine in rats: A video-intracranial lation. Nat. Neurosci. 6, 4350.
electroencephalographic study. Exp. Neurol. Zorec, R., Araque, A., Carmignoto, G., Haydon,
220, 293302. P.G., Verkhratsky, A., and Parpura, V. (2012).
Wetherington, J., Serrano, G., and Dingledine, R. Astroglial excitability and gliotransmission:An
(2008). Astrocytes in the epileptic brain. Neuron appraisal of Ca2+ as a signalling route. ASN
58, 168178. Neuro. 4, e00080.
6
AstrocyteNeuron Interactions
A NGUS M.BROW N A ND BRUCE R .R A NSOM

INTRODUCTION neuronal membrane can result in large devia-


The diversity and complexity of brain function tions from baseline levels in the interstitial fluid
in part derives from the close association of het- (Ransom, 2009). The most recognized of these is
erogeneous cell types separated by a narrow gap the effect of neuronal activity on [K+]o, which can
filled with interstitial fluid. Such an arrangement increase from 3 mM to 12 mM under physiologi-
optimizes rapid cell-to-cell communication and cal conditions (Ransom etal., 1992). pHo can also
facilitates homeostatic control of the compart- be affected, the resulting interstitial acidification
ments present within the central nervous system being dependent on the degree of neuronal activ-
(CNS). There are two main types of cell present in ity (Ransom et al., 1992). Synaptic activity can
the brain: neurons, the classic electrically excit- result in elevated [glutamate]o at synapses, which,
able cell, and glia, of which we focus on astro- given the potential excitotoxic function of gluta-
cytes. Broadly speaking, the function of neurons mate, can have grave consequences if not rapidly
is communication with other neurons, and the sequestered (Choi, 1992). Such alterations in ion
function of astrocyte cells is to maintain an opti- concentrations also have effects on osmolarity
mal environment for neuronal activity. Thus the of the various compartments, thus intercellular
dogmatic view of brain function is of neuronal water transport is controlled by astrocytes (see
communication mediated by electrical signal- later discussion). The interstitial space is also the
ing. Such signaling relies on energy-dependent site of energy substrate transfer between astro-
maintenance of transmembrane concentrations cytes and neurons, with astrocyte-to-neuron
of sodium (Na+) and potassium (K+; Bear et al., metabolic interactions being increasingly real-
2007). The transmembrane separation of ions is ized as a vital component of CNS function.
essential for the action potentials that underlie
neuronal activity, but such activity results in dis- I O N H O M E O S TA S I S
ruption of these gradients by (a) disturbing the
equilibrium of ion distribution both in cells and [K+]o
in the interstitium, which (b) requires homeo- The transmembrane ion gradients that are pre-
static mechanisms to restore ionic equilibrium sent across neuronal and glial cell membranes
for optimal neural function (Dienel, 2009). Given are vital for reliable electrical communication
the close proximity of cells to each other, it is easy throughout the CNS. In general, cell membranes
to appreciate how activity in one cell type can are more permeable to K+ than to Na+ at rest, thus
affect neighboring cells via the interstitial fluid. the resting membrane potential approaches the K+
This interdependent relationship lies at the heart reversal potential (Hille, 2001). Indeed, astrocytes
of astrocyte neuron interactions. In the past are exclusively permeable to K+, thus the resting
decade or so, the diversity of roles assumed by membrane potential equals the K+ reversal poten-
astrocytes has burgeoned to the extent that full tial, resulting in the astrocyte membrane potential
coverage is beyond the scope of this chapter, thus responding in a manner predicted by the Nernst
we focus on a few key functions of astrocytes. equation to elevations in [K+]o (Kuffler & Nicholls,
Neuronal activity in the form of action poten- 1966). Excitation of neurons and axons in the
tials and synaptic activity disturbs the ionic form of action potentials are a result of transient
composition of interstitial space. Given the con- increases in Na+ permeability followed by K+ per-
strictive dimensions of the interstitial space, rela- meability. The increased Na+ permeability occurs
tively minor changes in ion movements across via voltage-dependent opening of Na+ channels
AstrocyteNeuron Interactions 99
leading to a Na+ influx into the cells, followed by AQP4 aquaporin water channel was knocked out,
opening of K+ channels leading to an efflux of K+ activity-evoked elevations in interstitial K+ were
from the cell (Hille, 2001). Given the large extra- attenuated compared to wild type, indicative of
cellular [Na+] and the limited movements of Na+ decreased water uptake into astrocytes. In addi-
into cells required to produce an action potential tion, there was enhanced gap-junction coupling
(see Hille, 2001, for detailed calculations), there is of astrocytes and enhanced K+ redistribution
a negligible effect on [Na+]o resulting from action (Strohschein etal.,2011).
potentials. However, given the longer duration of
opening of K+ channels, the low [K+]o, and the small pHo
interstitial space, which acts to concentrate ions, Astrocytes are an important site for regulation of
there is a significant increase in [K+]o as a result pH. Such regulation is important as alterations in
of action potential conduction (Ransom et al., pH of astrocytes can lead to changes in the pH
2000). Although the increase in [K+]o is apparent of the interstitium, which in turn can affect the
in response to a single action potential, it is repeti- pH of neurons given the constricted interstitial
tive activity that results in significant, long-lasting space separating astrocytes and neurons. pH
elevations in [K+]o. An interesting feature of physi- is of course a measurement of hydrogen, [H+],
ologically evoked [K+]o elevations is that there expressed on a logarithmic scale to counter the
is a ceiling level above which the [K+]o will not nanomolar concentrations of H+ in living tissue
increase due to the buffering capacity of the astro- (pH=log10[H+]). The expression of pH and the
cytes (Ransom et al., 1985). Large elevations of limited range encountered under physiological
[K+]o would lead to neuronal depolarization, thus conditions can suggest that [H+] does not vary a
the transmembrane ion gradients must be rap- great deal, but relevant physiological concentra-
idly restored for optimal neuronal function. One tions are from 10 to 300 nM (Rose & Ransom,
of the first roles of astrocyte to be described was 1998). Such a range of [H+] has very important
in buffering increases in [K+]o (Kofuji & Newman, consequences as proteins and enzymes are
2004). Astrocytes take up K+ via three separate exquisitely sensitive to pH with changes of as lit-
mechanisms:the Na+K+ATPase, which dominates tle as 0.05 units having large effects on function
at modest elevations of K+; the NKCCl cotrans- (Stryer, 1995). From data acquired in multiple
porter, which dominates at higher concentrations mammalian astrocytes, a broad generalization
(Hertz et al., 2013); and the inwardly rectifying can be made that pHi of astrocytes contains up
K+ channel (Kofuji & Newman, 2004). The astro- to 0.2 pH units more acid than the interstitial pH
cytic Na+K+ATPase has a Km value of up to 15 mM, (Rose & Ransom, 1998). This implies an obvious
significantly higher that its neuronal counterpart difference in transmembrane [H+] and a reversal
and a value that makes it ideally suited to buff- potential for H+ of about 10 mV. Given a rest-
ering the elevations of K+ routinely encountered ing membrane potential of 85 mV or so, there
under physiological conditions (Verkhratsky &
Butt, 2007). Since astrocytes are interconnected
via gap junctions into syncitia, the K+ accumulated Subretinal space
during buffering activity can be redistributed via
the syncitium and released at distant sites in order
to maintain low [K+]o, a process known as spatial
buffering (Newman, 1995). Within individual
astrocytes, such as Muller cells in the retina, a
specialized form of buffering occurs known as K+ IPL
siphoning. Muller cells span the width of the ret- Endfoot
ina but express K+ channels in a polarized man-
Vitreous
ner. The Kir2.1 inwardly rectifying channels are
expressed at the inner and outer plexifrom layers FIGURE 6.1: Schematic illustration of K+
where there is significant synaptic activity and siphoning in a retinal Muller cells. Inwardly
thus release of K+ from neural elements. As such, rectifying Kir2.1 channels (filled circle) are expressed
the Kir2.1 channels are ideally located to take up in the inner plexiform layer (IPL, grey area) to absorb
K+. The Muller cells express Kir4.1 channels at the K+ released during synaptic activity. K+ is distributed
endfoot and vitreous borders where they release K+ within the Muller cell and is ejected via Kir4.1 channels
into the vitreous humor and the blood (Kofuji & (open circles) located on the endfoot into the vitreous
Newman, 2004; Figure 6.1). In mice where the humor and the subretinalspace.
100 Part II: Homeostatic Control

is a clear outward movement of H+ creating an shuttling, as lactate is cotransported with H+,


inward electrochemical gradient for H+ across the or as a result of depolarization causing inward
astrocyte membrane. There are a variety of mech- transport of the Na+/HCO3 cotransporter. It is
anisms in which pHi of astrocytes is regulated, hypothesized that neuronal-induced elevation
and these can be divided into Na+-dependent in [K+]o ultimately leads to a negative feedback
and Na+-independent mechanisms. Into the loop that limits neuronal activity. The elevated
former category are the Na+/H+ exchanger, [K+]o causes depolarization of the astrocyte mem-
Na+/HCO3 contransport, and Na+-dependent brane, which in turns leads to inward transport
Cl/HCO3 exchanger, and into the latter lie of HCO3 via the Na+/HCO3 cotransporter. The
Cl/HCO3 exchange and the H+ pump (Rose & resulting astrocyte alkalization causes an acidi-
Ransom, 1998). The differences in astrocyte fication of the interstitium, which in turn leads
transmembrane Na+ concentrations provide the to depression of neural activity by an as yet
electrochemical gradient for select ions to couple unknown mechanism (Rose & Ransom,1998).
their movements relative to the Na+ gradient. The
Na+/H+ exchanger is present in all mammalian NEUROTRANSMITTER
cells. It is electroneutral with a 1:1 stochiometry H O M E O S TA S I S
in which influx of 1 Na+ is countered by the efflux A key function of astrocytes is the recovery and
of a H+, resulting in intracellular alkalization. cycling of the excitatory neurotransmitter glu-
The Na+/H+ exchanger plays an important role tamate at synapses. Glutamate, a nonessential
in setting the baseline pHi, a role it shares with amino acid, is the dominant excitatory neuro-
the HCO3 exchangers and transporters. Na+/ transmitter in the CNS and is estimated to be
HCO3 cotransport is exclusive to glial cells and present at 80% of excitatory CNS synapses (Bear
absent from neurons (Verkhratsky & Butt, 2007). et al., 2007). Given the potential excitotoxic
The cotransporter is electrogenic due to the sto- effects of uncontrolled glutamate levels in the
chiometry of transport of 1 Na+ per 2 HCO3 in brain, the blood-brain barrier excludes glutamate
astrocytes. The resulting reversal potential of70 present in the systemic circulation from the brain
to 80 mV is close to the resting membrane parenchyma, thus glutamate must be manufac-
potential and dictates that membrane depolariza- tured within the brain. The source of glutamate is
tion would lead to inward HCO3 transport and ultimately glucose, with glutamate derived from
alkalization, whereas membrane hyperpolariza- the tricarboxylic acid substrate -ketoglutyrate
tion would lead to HCO3 efflux and intracellular via actions of the enzyme glutamate dehydro-
acidification (Pappas & Ransom, 1994). The elec- genase, a reaction that occurs in the presynaptic
trogenic nature of the cotransporter is important terminal (McKenna et al., 2006). Once gluta-
as it implies that membrane potential will influ- mate is secreted from the presynaptic terminal
ence transport and, perhaps more important and subsequently released from the postsynaptic
with regard to function, that activation of the receptors, it is taken up into astrocytes, rather
cotransporter will alter membrane potential. The than back into the presynaptic terminal. This is
Na+-dependent Cl/HCO3 exchanger uses the facilitated by glutamate transporters present on
Na+ gradient to transport Cl into the cell and the astrocyte membrane, which use the trans-
HCO3 out, whereas the Cl/HCO3 exchanger is membrane Na+ gradient to drive glutamate into
independent of the Na+ gradient. Since the acid the astrocyte (i.e., from a compartment of low
extrusion mechanisms described earlier employ glutamate concentration, the synaptic cleft, to
the Na+ gradient, these ultimately require adeno- a compartment of high glutamate concentra-
sine triphosphate (ATP) to fuel the Na+K+ATPase tion, the astrocyte). Once inside the astrocyte
that maintains the uneven distribution of trans- the glutamate is converted to glutamine by the
membrane Na+. The H+ pump (H+ ATPase) also ATP-dependent enzyme glutamine synthase and
requires ATP to fuel pumping of H+ out of thecell. released back into the extracellular space, from
which it is taken up by the presynaptic termi-
Activity-Induced pH Changes nal. The glutamine, once inside the presynaptic
and Regulation terminal, is converted to glutamate by glutami-
Depolarization of astrocytes, as would occur as a nase and is subsequently repackaged for vesicu-
consequence of elevated interstitial K+ resulting lar release (McKenna etal., 2006). This sequence
from increased neuronal activity, causes alkali- constitutes the glutamateglutamine cycle, and it
zation in astrocytes (Pappas & Ransom, 1994). confers several advantages on the neuron. First,
This may result from astrocyte-to-neuron lactate it shifts the metabolic burden from the neuron to
AstrocyteNeuron Interactions 101

the astrocyte, thus sparing neuronal ATP, which given that the direction of glutamate movements
could be a vital neuroprotective mechanism dur- is determined by the Na+ concentration (Attwell,
ing periods of energy deprivation. Second, the 2000). The resulting extracellular glutamate has
glutamine shuttled from the astrocyte to the direct access to Ca2+-permeable receptors, result-
neuron is inert as a neurotransmitter; that is, ing in toxic Ca2+ influx into neurons triggering
it does not bind to glutamate receptors and is apoptosis and necrosis (Choi,1992).
thus incapable of contributing to excitotoxicity
(Figure6.2). E N E R G Y M E TA B O L I S M
A consequence of energy deprivation, as can Interest was reignited in the dormant field of
occur during stroke or anoxia or aglycemia as brain glycogen research by several studies. These
separate insults, is a decrease in ATP production, were predominantly tissue culture studies, the
such that the tissue demand for ATP exceeds sup- conclusions of which could be extrapolated to
ply. The ATP requirements fall under two main more intact systems. The first study to suggest a
categories:signaling, involving action potentials link between astrocytic glycogen and function
and synaptic potentials, and maintenance of resulted from experiments in which hypotha-
the hyperpolarized resting membrane potential lamic neurons were cultured with glial cells either
(Attwell & Laughlin, 2001). It is the latter that has present or absent from the culture. Neurons that
the most far-reaching consequences under condi- were seeded in the company of glial cells survived
tions of energy deprivation, as there is insufficient for longer periods of time than neurons seeded
ATP to fuel the Na+K+ATPase that maintains the in the absence of glial cells. The clear conclusion
transmembrane ion gradients (i.e., low [K+]i and was that coculturing neurons with glia confers a
high [Na+]o.) Thus, as a result, the ion concentra- benefit of an unknown type, possibly metabolic,
tions tend to equilibrate across the cell membrane on the neurons, prolonging their survival in the
such that the resting membrane potential depo- artificial culture environment (Whatley et al.,
larizes. The equaling of the Na+ concentration 1981). A subsequent study asked a more direct
across the cell membrane has grave consequences question: What is the manner of protection
for cell survival as the Na+-coupled glutamate offered by the glial cells in cocultures of corti-
transporters can result in an efflux of glutamate cal neurons? The conclusion from that study was
from the astrocyte into the interstitial space, that it is not the presence per se of the glial cells
(astrocytes) that protects the neurons but rather
the glycogen contained within the astrocytes.
In cocultures in which the glycogen content of
the astrocytes was decreased, neurons did not
survive as long as when the astrocytes displayed
gln gln
ATP robust glycogen content (Swanson & Choi, 1993).
GL
pre GS That glycogen can protect neurons is not surpris-
glu
Na+
ADP ing, given its role in the liver and skeletal muscle
glu glu as an energydepot.
post
In the more intact central model of white
matter, the rodent optic nerve, studies have high-
lighted the basic role of glycogen as a central
energy store/buffer. In an in vitro model of the rat
astrocyte
optic nerve, where the tissue is maintained in a
superfusion chamber held at 37C, bubbled with
95% oxygen (O2) 5% CO2, and superfused with
FIGURE 6.2: The glutamateglutamine cycle. artificial cerebrospinal fluid containing 10 mM
Glutamate (glu) released from the presynaptic termi- glucose, the stimulus-evoked compound action
nal is cleared from the synaptic cleft via Na+-coupled potential (CAP) can be maintained for many
glutamate transporters present on the astrocyte mem- hours. If the preparation is exposed to anoxia
brane (open circle). The glutamate is converted to glu- (withdrawal of O2), the CAP falls to zero within
tamine (gln) in the astrocyte by glutamine synthase 5 minutes (Stys etal., 1991). However, in the pres-
(GS), a reaction that requires adenosine triphosphate ence of O2, but in the absence of glucose, the CAP
(ATP). The glutamine is then shuttled back to the fails after 20 to 30 minutes (Ransom & Fern,
presynaptic terminal for conversion to glutamine by 1997). This delay in CAP failure in the absence
glutaminase(GL). of exogenously applied glucose strongly suggests
102 Part II: Homeostatic Control

that an endogenous energy reserve is present in


the tissue that is capable of supporting axon con-
duction, albeit for a limited period of time. The
most likely candidate is glycogen. The presence
of glycogen in the CNS has been known for dec-
ades, and it has the intriguing feature of being
present only in astrocytes in the adult (Cataldo &
Broadwell, 1986). To investigate whether gly-
cogen was the energy reserve that supported
function, parallel experiments were carried out FIGURE 6.3: Glycogen in the astrocyte fuels axon
in which the CAP was recorded in the absence function. Glucose is transported from the blood vessel
of glucose and glycogen content measured. The to astrocytes via glucose transporters (closed circles).
results indicated that after switching to zero glu- In astrocytes glycogen is formed from glucose via gly-
cose, while the CAP is maintained, the glycogen cogen synthase (GS) and is converted, ultimately, to
content starts to fall, and once the glycogen level lactate by glycogen phosphorylase (GP). The lactate is
in the tissue reaches its nadir, the CAP starts to shuttled from the astrocyte to the neuron via monocar-
fail and from that point falls rapidly to zero. This boxylate transporters (MCTs, open circles), where it is
temporal correlation between glycogen content oxidatively metabolized.
and CAP conduction closely associates the gly-
cogen present in the tissue with the ability of the
nerve to conduct action potentials (Figure 6.3) reasonably be viewed as a consequence of iatro-
and leads to the prediction that the larger the gly- genic insulin therapy, and thus its occurrence has
cogen content of the tissue, the longer the CAP is been a significant clinical emergency only since
maintained. This was tested by altering the gly- the introduction of exogenously applied insulin
cogen content of the tissue prior to the period of to treat type 1 diabetics (Frier & Fisher, 2007).
aglycemia. Incubating the nerves in 1 mM nor- However, it should be noted that the condition of
epinephrine decreased glycogen content, whereas insulinomas would also produce such effects.
incubating the tissue in artificial cerebrospinal We studied the ability of glycogen to support
fluid containing 25 mM glucose elevated gly- axon conduction in our optic nerve preparation
cogen content. Exposing nerves to aglycemia by imposing on the tissue a high-frequency train
after increasing or decreasing the glycogen con- of stimuli of 100 Hz. What we found was that 100
tent led to an increase or decrease, respectively, Hz stimulus for 4 minutes caused a significant
in the latency to CAP failure (Wender et al., decrease in glycogen content, even in the pres-
2000). Similar experiments were carried out in ence of normoglycemic concentrations (10 mM)
a mouse model, and qualitatively similar results of glucose. These results imply that, under con-
were found. Glycogen content decreased from ditions where tissue energy demand exceeds glu-
the onset of aglycemia and took 20 minutes to cose supply, glycogen is metabolized to provide
reach its nadir, at which point the CAP failed. supplemental energy substrate to support axon
Augmentation of glycogen content increased conduction, a clear example of glycogen support-
CAP latency during aglycemia, whereas attenu- ing physiological function (Brown etal.,2003).
ation of glycogen content increased CAP latency
(Brown etal.,2003). L A C TAT E T R A N S F E R F R O M
In addition to glycogens ability to support ASTROCYTE TOAXON
axonal conduction during periods of glucose The means by which glycogen supports function
withdrawal, the ability of glycogen to support has also been investigated. Clearly, glycogen is too
physiological function was studied. Although large a molecule to be transported intercellularly,
glucose withdrawal is an extremely useful thus it must be metabolized in the astrocytes to
experimental maneuver with which to study a conduit that is transported from the astrocyte
a substrates ability to support function in the to the neuron/axon (Dringen et al., 1993). The
absence of glucose, it is artificial in that in vivo most likely candidate for this conduit is lactate.
blood glucose levels would be hypoglycemic In tissue culture studies, astrocyte cultures were
rather than aglycemic, although it has been cal- shown to release lactate but not glucose (Dringen
culated that blood glucose levels in the brain can etal., 1995). This is an important point as astro-
approach zero during severe bouts of hypoglyce- cytes lack the enzyme glucose-6-phosphatase
mia (Frier & Fisher, 2007). Indeed, aglycemia can and thus cannot convert glucose-6-phosphate to
AstrocyteNeuron Interactions 103

glucose, as occurs in the liver. Thus once glucose in [K+]o (see previous discussion), thus K+ can
is taken up by astrocytes and is phosphorylated be proposed as a universal signaling molecule
to glucose-6-phosphate, the glucose is com- indicative of increased neuronal activity. The
mitted down a metabolic pathway that will not [K+]o activates a Na+-coupled bicarbonate trans-
result in glucose formation. Lactate is produced porter on astrocyte membranes whose activa-
by the action of the enzyme lactate dehydro- tion results in increased transport of bicarbonate
genase on the substrate pyruvate, a reversible and intracellular alkalization of the astrocyte.
reaction. The equilibrium between lactate and This in turn activates the enzyme soluble adenyl
pyruvate depends on the energy status of the cyclase, which converts ATP to cyclic AMP. The
cell and the presence of lactate dehydrogenase cyclic AMP then promotes glycogenolysis via
isoforms (Stryer, 1995). In astrocytes, the lactate glycogen phosphorylase and results in glycogen
dehydrogenase isoform that converts pyruvate to metabolism to lactate, which is then transported
lactate is present. In mouse optic nerve, we have out of the astrocyte to the neuron (Pellerin &
shown that lactate can support the CAP in the Magistretti,1994).
absence of exogenously applied glucose (Brown
et al., 2001), supporting the notion that lactate G LY C O G E N A N D M E M O R Y
delivered to neurons from astrocytes can support In a recent study on the rodent brain, a role
axon function. The lactate movement across cell for glycogen in supporting the acquisition of
membranes is accomplished via the actions of memories was described (Choi etal., 2012). The
membrane-bound monocarboxylate transport- experiments entailed introducing rats into a box,
ers (MCT), which cotransport H+ with the lac- from which another box was readily accessible
tate. The MCT that preferentially moves lactate through a door. If the rats entered the second
out of cells is the MCT1 subtype, and it is located box, they were given a shock through the floor
on astrocytes, whereas the MCT2, which takes up of the box, which acted as an aversive condition-
lactate into cells, is located on neurons. If lactate ing response. The rats were removed from the
does support axon conduction as a consequence box and then reintroduced into the box from
of glycogen metabolism, then interrupting lactate periods of between 1 hour and 7days later. The
transfer from astrocytes to axons should result in latency for the rats to enter the second box was
a loss of axon conduction as seen by a decrease in used as an index of their memory: the longer
the CAP area. This was tested in a rodent prepa- the latency, the better the rat had retained that
ration by introducing compounds that inhibit memory. Injecting 1,4-dideoxy-1,4-imino-D-
lactate transport at the MCT (d-lactate, CIN), arabinitol (DAB), an inhibitor of glycogen phos-
resulting in reduced CAParea. phorylase, into the hippocampi of the rat resulted
in a dose-dependent decrease in the latency,
E L E VAT E D [ K + ] O S I G N A L S indicative of decreased memory function. The
A S T R O C Y T E T O M E TA B O L I Z E DAB had to be injected before the conditioning
G LY C O G E N stimuli in order to be effective. The lactate con-
It is timely to consider the signaling mechanism centration ([lac]o) in the extracellular space in
by which increased neuronal activity is sensed by the hippocampi was elevated in the hour or so
astrocytes. In the mid-1990s, the astrocyte neu- after the conditioning stimuli, whereas in rats
ron lactate shuttle hypothesis was proposed, in injected with DAB no elevation in [lac]o was seen.
which uptake of glutamate into astrocytes was Injecting the rats with lactate at the same time
coupled to glucose uptake into astrocytes. This that they were injected with DAB attenuated the
hypothesis has proved to be controversial and effects of DAB, indicating that circumventing the
clearly does not apply to areas of the brain devoid contribution of glycogen-derived lactate by direct
of glutamatergic synapses, or white matter, which introduction of lactate in the extracellular space
accounts for 50% of the adult human brain vol- restored the animals memory. Finally, using
ume. Amore recent hypothesis is more attractive molecular biology techniques to downregulate
as it applies to the entire CNS (and possibly the expression of the monocarboxylate transporter 1
peripheral nervous system). In a study predomi- (MCT1), which takes up glycogen-derived lactate
nantly using astrocyte cultures, a key role for into the neurons, decreased memory acquisition
elevated [K+]o was identified as the primary sign- (Suzuki et al., 2011). These data strongly sug-
aling mechanism between neurons and astro- gest that glycogen is required for acquisition of
cytes. It is universally accepted that increased memories, where it is metabolized in astrocytes
neuronal activity results in localized elevations and then the resulting glycogen-derived lactate is
104 Part II: Homeostatic Control

transported out of the astrocyte and into astro- several primary conclusions can be drawn. First,
cytes via the MCT1. This is the first description of AQP4 is essential for water movement across
glycogen playing a vital role in a key physiologi- astrocyte cell membranes; in knockout animals
cal brain function. the water transport is one-seventh that of control
conditions. Similarly, AQP4 plays a vital role in
A S T R O C Y T E S FA C I L I TAT E water transport across the blood-brain barrier
C S F C I R C U L AT I O N A N D (Papadopoulos & Verkman, 2013). The aqua-
CLEANSING OFBR AINECS porin member AQP1 is expressed on the apical
In addition to comprising different cell types membrane of chorioid plexus cells and facili-
(neurons and glia, of which there are subdivi- tates water movement from the chorioid plexus
sions of each), the CNS is compartmentalized blood supply into the ventricles (Papadopoulos &
as follows. The cellular compartments com- Verkman, 2013). A Na+K+ATPase on the apical
prising the cell bodies of the neurons and glia membrane creates the Na+ gradient that pro-
are surrounded by the fluid-filled interstitium. motes water movement (Nielsen etal., 1993). CSF
The brain parenchyma itself is enclosed within absorption occurs via AQP4 channels expressed
three meninges: the pia mater, arachnoid, and on ependymal cells, which facilitates move-
dura. The cerebrospinal fluid (CSF) is contained ment of water into the venous drainage system
within the subarachnoid space (between the (Ransom, 2009). The AQP4 channels present on
dura and arachnoid) and the ventricular system astrocyte endfeet that abut the blood vessels are
(Suzuki etal., 2011). The CSF interacts with the a low-resistance pathway for water movement
interstitial fluid across two borders, the pial glial between the blood and the interstitial fluid and
membrane and ependymal cells, which line the additionally provide a route for clearance of sol-
ventricles. The pial glial membrane is present utes from the interstitium to the venous drain-
at the loosely fenestrated epithelial cells lining age. Experiments using fluorescent tracers and
blood vessels supplying the brain via the Virchow AQP4 knockout mice confirmed the presence
Robin space. The CSF is produced by the choroid of the pathway and emphasized its importance
plexus cells that line the ventricles, and the posi- in the drainage of clearance of large solutes,
tive pressure resulting from continual produc- including beta-amyloid, from the parenchyma
tion is assumed to be the main force that drives (Papadopoulos & Verkman, 2013). AQP4 chan-
CSF flow. The water content of the interstitium nels and voltage-dependent Kir4.1 ion channels
and the CSF varies, with the CSF being more are colocalized to the perivascular and subpial
hypotonic than the interstitium due to lack of endfeet at synapses. Increased synaptic activity
proteins and cholesterol (Ransom, 2009). From results in elevated interstitial [K+], which is trans-
the point of view of water distribution, it should ported into astrocytes via the Kir4.1 ion channel.
be appreciated that there is continual change in This creates osmotic pressure across the astrocyte
compartmental osmolarity due to neural activ- membrane and drives water accumulation in
ity. For example, the elevated [K+] and glutamate astrocytes, which is subsequently redistributed
in the interstitium that accompany neuronal via the astrocytic syncitium (Strohschein et al.,
activity will cause increases in osmolarity that 2011; Iliff etal.,2012).
must be compensated for. Thus water move-
ment between compartments is a very important BLOOD VESSEL DIAMETER
process to ensure optimal brain function. The Functional hyperemia, first proposed over a
AQP4 member of the aquaporin water channel century ago (Verkhratsky & Butt, 2007), is the
family is expressed on astrocyte membranes. Its process by which neuronal actively is coupled
expression is polarized with primary expression to localized blood flow, controlled by blood ves-
on endfeet surrounding blood vessels, astrocyte sel diameter. The mechanism(s) of this process,
processes abutting the glia limitans, and ependy- which has recently been elucidated, displays a
mal cells (Ransom, 2009). The function of aqua- degree of metabolic flexibility such that the alter-
porins is to facilitate water movement across ations in blood vessel diameter are determined
cell membranes commensurate with alterations by factors that are indicative of the energy status
in water content. The role of AQP4 has derived of the tissue. One of the first studies to investi-
mainly from knockout experiments where func- gate in detail the role of astrocytes in controlling
tion in genetically engineered mice lacking the localized arteriole diameter demonstrated that
AQP4 was measured. From these experiments elevated [Ca2+]i in astrocytes evoked arteriole
AstrocyteNeuron Interactions 105

dilation via the actions of prostaglandin E2 (Roy that contained mitochondria, clearly signal-
& Sherrington, 1890). However, the same mecha- ing an energy-dependent role for these objects.
nism, namely elevated astrocytic [Ca 2+]i, was also Functionally the human astrocytes resembled
shown to result in arteriole constriction (Zonta rodent astrocytes in that they supported intra-
et al., 2003). The apparent contradictory effects cellular Ca 2+ waves and responded to the pres-
can be explained by the degree of oxygena- ence of neurotransmitters by generating Ca 2+, but
tion of the tissue (Mulligan & MacVicar, 2004). the waves travelled four times faster than their
Under conditions of low O2, there is an increase rodent counterparts (Oberheim et al., 2009).
in interstitial lactate, which blocks the uptake of The implications of these findings will almost
prostaglandin into astrocytes. The prostaglan- certainly be far reaching, with the most obvious
din then acts to dilate blood vessels to deliver conclusion that astrocytes can communicate via
increased blood (and O2) to the tissue. However, the extended processes over long distances.
in well-oxygenated tissue, interstitial lactate lev- A recent study has investigated the function
els are low and the effects of arachindoic acid of human astrocytes further by creating chimeric
derived 20-HETE dominate, and result in arte- mice that possess both native murine astro-
riole constriction. cytes as well as transplanted human astrocytes,
that maintain their human morphology, and by
HUMAN AND RODENT extension that express their unique human func-
A S T R O C Y T E S M AY B E tions. The chimeric mice displayed improvements
Q U A L I TAT I V E LY D I F F E R E N T in the key psychological parameters of learning
The dogmatic view of astrocytes as relatively and memory. Avariety of tests were carried out,
small cells that oversee an individual nonover- and in all cases the chimeric mice performed bet-
lapping domain was shattered by a recent report ter than wild-type mice (Oberheim etal., 2009).
that described both morphological and func- Such studies, while in their infancy, are intrigu-
tional features of human astrocytes (Gordon ing and prompt the key questions regarding what
et al., 2008). Cortical tissue was obtained dur- function(s) or property(s) of the astrocytes confer
ing surgery to remove focal lesions sites in the the human performance on thesemice.
hippocampus of patients suffering from intrac-
table epilepsy. Immunohistological analysis of CONCLUSION
the tissue using glial fibrillary acidic protein Astrocytes are essential partners in the optimal
and diolistic staining techniques revealed that functioning of neurons. Akey aspect of this rela-
human astrocytes vary dramatically from their tionship is the astrocyteneuron interactions
rodent cousins. The most immediate difference via the interstitial fluid. The constricted inter-
was in the size of the astrocytes. Whereas rodent stitial space leads to close proximity of the cells
astrocytes cover a field extending about 56 m in such that any deviation from equilibrium of ions
diameter, the human astrocytes covered a field affects neighboring cells. Thus the interstitial
over twice (142 m) as wide. It is calculated that fluid is an ideal means by which cells can commu-
the human astrocyte domain encompasses up nicate. The ionic disruptions caused by increased
to 2million synapses. More important from the neuronal activity (e.g., increased [K+]o, decreased
point of view of cell-to-cell communication, the pH, and increased [glutamate]o) are all buffered
domains of the human astrocytes overlapped, by astrocytes, as are the subsequent changes in
whereas those of the rodent did not, strongly sug- osmolarity resulting from these perturbations.
gesting astrocyte-to-astrocyte communication Of increasing importance are the metabolic
in the human. In addition, the dendrites of the interactions between neurons and astrocytes in
human astrocyte were much finer, and thus the the form of how neurons signal increased activity
dendritic field of the human astrocytes had to astrocytes and how the astrocytes respond to
many orders of magnitude more surface area such activity.
than that of the rodent astrocyte. However, the
most startling discovery was the presence of long References
processes that emerged from the somas of the Attwell D. (2000). Brain uptake of glutamate:Food
human astrocytes. These processes could be up for thought. J Nutr 130, 10231025.
to 1mm long and traversed cortical layers, hence Attwell D & Laughlin SB. (2001). An energy budget
their description as interlaminar astrocytes. The for signaling in the grey matter of the brain. J
process expressed regularly spaced varicosities Cereb Blood Flow Metab. 21, 11331145.
106 Part II: Homeostatic Control

Bear MF, Connors BW & Paradiso MA. (2007). Kofuji P & Newman EA. (2004). Potassium buffering
Neuroscience: Exploring the brain. Philadelphia: in the central nervous system. Neuroscience 129,
Lippincott Williams and Watkins. 10451056.
Brown AM, Baltan Tekkk S & Ransom BR. (2003). Kuffler SW & Nicholls JG. (1966). The physiology of
Glycogen regulation and functional role in neuroglial cells. Ergeb Physiol 57, 190.
mouse white matter. J Physiol 549, 501512. McKenna MC, Gruetter R, Sonnewald U,
Brown AM, Wender R & Ransom BR. (2001). Waagepetersen HS & Schousboe A. (2006).
Metabolic substrates other than glucose support Energy metabolism of the brain. In: Siegel GJ,
axon function in central white matter. J Neurosci Albers RW, Brady ST & Price D, L., eds. Basic
Res 66, 839843. neurochemistry. San Diego: Academic Press,
Cataldo AM & Broadwell RD. (1986). Cytochemical 531558.
identification of cerebral glycogen and glucose- Mulligan SJ & MacVicar BA. (2004). Calcium tran-
6-phosphatase activity under normal and exper- sients in astrocyte endfeet cause cerebrovascular
imental conditions. I. Neurons and glia. J Elec constrictions. Nature 431, 195199.
Micro Tech 3, 413437. Newman E. (1995). Glial cell regulation of extracel-
Choi DW. (1992). Excitotoxic cell death. J Neurobiol lular potassium. In:Ransom B & Kettenmann B,
23, 12611276. eds. Neuroglia. Oxford:Oxford University Press,
Choi HB, Gordon GR, Zhou N, Tai C, Rungta RL, 717731.
Martinez J, Milner TA, Ryu JK, McLarnon JG, Nielsen S, Smith BL, Christensen EI & Agre P. (1993).
Tresguerres M, Levin LR, Buck J & MacVicar BA. Distribution of the aquaporin CHIP in secretory
(2012). Metabolic communication between astro- and resorptive epithelia and capillary endothelia.
cytes and neurons via bicarbonate-responsive Proc Natl Acad Sci USA 90, 72757279.
soluble adenylyl cyclase. Neuron 75, 10941104. Oberheim NA, Takano T, Han X, He W, Lin JH,
Dienel GA. (2009). Energy metabolism in the brain. Wang F, Xu Q, Wyatt JD, Pilcher W, Ojemann
In:Byrne JH & Roberts JL, eds. From molecules to JG, Ransom BR, Goldman SA & Nedergaard
networks:An introduction to cellular and molec- M. (2009). Uniquely hominid features of adult
ular neuroscience. 2nd ed. NewYork:Academic human astrocytes. J Neurosci 29, 32763287.
Press, 49110. Papadopoulos MC & Verkman AS. (2013). Aquaporin
Dringen R, Gebhardt R & Hamprecht B. (1993). water channels in the nervous system. Nat Rev
Glycogen in astrocytes:Possible function as lac- Neurosci 14, 265277.
tate supply for neighboring cells. Brain Res 623, Pappas CA & Ransom BR. (1994). Depolarization-
208214. induced alkalinization (DIA) in rat hippocampal
Dringen R, Peters H, Wiesinger H & Hamprecht B. astrocytes. J Neurophysiol 72, 28162826.
(1995). Lactate transport in cultured glial cells. Pellerin L & Magistretti PJ. (1994). Glutamate uptake
Dev Neurosci 17, 6369. into astrocytes stimulates aerobic glycoly-
Frier BM & Fisher BM. (2007). Hypoglycaemia in sis:Amechanism coupling neuronal activity to
clinical diabetes. NewYork:JohnWiley. glucose utilization. Proc Natl Acad Sci USA 91,
Gordon GR, Choi HB, Rungta RL, Ellis-Davies GC & 1062510629.
MacVicar BA. (2008). Brain metabolism dictates Ransom B. (2009). The neuronal microenvironment.
the polarity of astrocyte control over arterioles. In:Boron WF & Boulpaep EL, eds. Medical phys-
Nature 456, 745749. iology. Philadelphia:Saunders Elsevier, 289309.
Hertz L, Xu J, Song D, Du T, Yan E & Peng L. (2013). Ransom BR & Fern R. (1997). Does astrocytic glyco-
Brain glycogenolysis, adrenoceptors, pyruvate gen benefit axon function and survival in CNS
carboxylase, Na(+), K(+)-ATPase and Marie white matter during glucose deprivation? Glia
E. Gibbs pioneering learning studies. Front 21, 134141.
Integr Neurosci7, 20. Ransom BR, Walz W, Davis PK & Carlini WG.
Hille B. (2001). Ionic channels of excitable mem- (1992). Anoxia-induced changes in extracellular
branes. Sunderland, MA:Sinauer Associates. K+ and pH in mammalian central white matter. J
Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Cereb Blood Flow Met 12, 593602.
Gundersen GA, Benveniste H, Vates GE, Deane Ransom BR, Yamate CL & Connors BW. (1985).
R, Goldman SA, Nagelhus EA & Nedergaard M. Activity-dependent shrinkage of extracellular
(2012). A paravascular pathway facilitates CSF space in rat optic nerve:Adevelopmental study. J
flow through the brain parenchyma and the Neurosci 5, 532545.
clearance of interstitial solutes, including amy- Ransom CB, Ransom BR & Sontheimer H.
loid beta. Sci Transl Med 4, 147ra111. (2000). Activity-dependent extracellular K+
AstrocyteNeuron Interactions 107

accumulation in rat optic nerve:The role of glial for long-term memory formation. Cell 144,
and axonal Na+ pumps. J Physiol 522, 427442. 810823.
Rose CR & Ransom BR. (1998). pH regulation in Swanson RA & Choi DW. (1993). Glial glycogen
mammalian glia. In:Kaila K & Ransom BR, eds. stores affect neuronal survival during glucose
pH and brain function. New York: Wiley-Liss, deprivation in vitro. J Cereb Blood Flow Met 13,
253275. 162169.
Roy CS & Sherrington CS. (1890). On the regulation Verkhratsky A & Butt A. (2007). Glial neurobiology.
of the blood-supply of the brain. J Physiol 11, NewYork:Wiley.
85108. Wender R, Brown AM, Fern R, Swanson RA, Farrell
Strohschein S, Huttmann K, Gabriel S, Binder DK, K & Ransom BR. (2000). Astrocytic glycogen
Heinemann U & Steinhauser C. (2011). Impact of influences axon function and survival during
aquaporin-4 channels on K+ buffering and gap glucose deprivation in central white matter. J
junction coupling in the hippocampus. Glia 59, Neurosci 20, 68046810.
973980. Whatley SA, Hall C & Lim L. (1981). Hypothalamic
Stryer L. (1995). Biochemistry. New York: W.H. neurons in dissociated cell culture:The mecha-
Freeman. nism of increased survival times in the pres-
Stys PK, Waxman SG & Ransom BR. (1991). Na+-Ca 2+ ence of non-neuronal cells. J Neurochem 36,
exchanger mediates Ca 2+ influx during anoxia in 20522056.
mammalian central nervous system white mat- Zonta M, Angulo MC, Gobbo S, Rosengarten B,
ter. Anna Neurol 30, 375380. Hossmann KA, Pozzan T & Carmignoto G.
Suzuki A, Stern SA, Bozdagi O, Huntley GW, Walker (2003). Neuron-to-astrocyte signaling is central
RH, Magistretti PJ & Alberini CM. (2011). to the dynamic control of brain microcircula-
Astrocyte-neuron lactate transport is required tion. Nat Neurosci 6, 4350.
7
Homeostatic Synaptic Scaling
atCentral Synapses
N I R A J S . D E S A I A N D E L I S A B E T H C . WA L C O T T

DEVELOPMENT AND because the presynaptic neurons are themselves


LEARNING developing, synaptic properties change substan-
Nearly every physiological system requires tially over developmental time, and landmark
some method of homeostatic regulationsome developmental events, like eye opening, abruptly
means of regulating its internal environment shift input. In the absence of implausibly exten-
and maintaining stable performance despite sive genetic instructions, it is difficult to imagine
external perturbations (Cannon, 1932). This is how developing neural circuits can function, or
perhaps especially true of the nervous system, even avoid destructive epileptic events, without
which not only faces the same challenges to sta- tight feedback control. Experiments, both in
bility as other systems (e.g., protein degradation vitro and in vivo, have confirmed this intuition,
and turnover, environmental perturbations) but demonstrating that development of the neocor-
also must, while maintaining stability, change tex and other brain structures is guided by sen-
many of its fundamental properties over time sory and motor experience and by neural activity
and in response to experience so as to allow (Berardi et al., 2000; Hensch, 2004; Knudsen,
individuals to learn and remember (Marder and 2004; Hengen etal., 2013; Keck etal.,2013).
Goaillard, 2006). Stability and change are not The homeostatic requirements imposed by
strictly opposing qualities, but neither are they learning processes are less obvious, and much
complementary, and the need to balance the two of what we understand about them comes from
and keep them from interfering with each other theoretical and computational studies rather
makes homeostatic regulation of the nervous sys- than experimental ones (Miller and Mackay,
tem an extraordinarily complicated problem. To 1994; Abbott and Nelson, 2000; Turrigiano and
make the problem somewhat more tractable, it is Nelson, 2000; Nelson and Turrigiano, 2008). The
common practice to break it up into two distinct starting point for most of these studies is Hebbs
but related (and still difficult) problems:develop- rule, which holds that a synapse connecting one
ment and learning. neuron to another should be strengthened if the
The developmental problem arises to a con- firing of the first neuron repeatedly and persis-
siderable extent because of the rapidity and tently leads to firing of the second (Dayan and
complexity of synapse development (Waites Abbott, 2005). Many variations on this rule have
etal., 2005; Collin and van den Heurval, 2013). been proposed over the years, and Hebbian mod-
In mammalian neocortex, the number of syn- els of synaptic plasticity have come to dominate
apses a typical neuron receives changes by sev- biologically based theories of learning. One com-
eral orders of magnitude in the weeks just before mon feature of this work is the understanding
and just after birth. Some of these synapses are that Hebbian plasticity is best complemented by
excitatory, others inhibitory, and still others homeostatic control (Abbott and Nelson, 2000).
neuromodulatory; some arise from neighboring The intuition here is simple and appealing:con-
neurons, others from neurons millimeters away; sider two neurons (A and B) connected by a syn-
some are located close to the cell body, while oth- apse. If the firing of Adrives B to fire, the synapse
ers are far along the dendritic arbor. The synaptic between them will be strengthened; this will
drive coming through these synapses fluctuates make it more likely that Awill drive B to fire in
dramatically over the course of development, the future; this will lead to more strengthening;
Homeostatic Synaptic Scaling at Central Synapses 109
this will lead to more driving; and so on. Absent cortical systems, though it must be noted that
some kind of constraint or negative feedback, closely related phenomena have been examined
Hebbian plasticity can lead to situations of using invertebrate and spinal systems (Davis
runaway synaptic potentiation because of such and Bezprozvanny, 2001; Davis, 2006; Pratt
positive feedback loops. Moreover, even without and Aizenman, 2007; Rich and Wenner, 2007;
positive feedback, synaptic modifications pro- Wenner, 2011; Frank, 2014; Wenner,2014).
duced by learning processes still likely necessi-
tate homeostatic control. Computational studies PHENOMENA
of neural networks with biologically constrained The field of homeostatic synaptic scaling has
parameters indicate that, without stringent (and been expanding rapidly, and recently numer-
unlikely) parameter fine tuning, such networks ous detailed and excellent reviews of many of its
are highly unstable and prone to extreme regimes aspects have appeared (Davis, 2006; Turrigiano,
of high or low activity (Lazar et al., 2009; Watt 2008; Pozo and Goda, 2010; Wenner, 2011;
and Desai, 2010; Tetzlaff etal., 2011, 2012). Only Turrigiano, 2012; Vitureira et al., 2012; Wang,
including mechanisms of homeostatic control 2012; Davis, 2013; Lazarevic etal., 2013; Vitureira
can produce reasonable network behavior. and Goda, 2013; Chen etal., 2014; Lee etal., 2014;
What form should those mechanisms take? Pribiag and Stellwagen, 2014; Siddoway et al.,
This is a question that has been actively and 2014; Thalhammer and Cingolani, 2014; Whitt
explicitly investigated for the better part of three etal., 2014). In this section we limit ourselves to
decades, in the context of both development a concise description of the basic phenomena,
and learning. Several answers have been offered including some of what is known about its molec-
(Bienenstock etal., 1982; Turrigiano, 1999; Desai, ular underpinnings.
2003; Abraham, 2008; Nelson and Turrigiano, Synaptic scaling has been studied in many
2008; Watt and Desai, 2010; Turrigiano, 2011; ways:in the neocortex, hippocampus, and spinal
Vitureira et al., 2012; Chen et al., 2013; Davis, cord; in cell cultures, slice cultures, brain slices,
2013; Chistiakova etal., 2014). One is that a neu- and the intact brain; from young animals and
rons intrinsic excitability, as determined by its mature animals; in infragranular, granular, and
complement of voltage- and calcium-gated ion supragranular cortical layers; and after various
channels, should be modulated so as to coun- types of pharmacological manipulation and sen-
terbalance shifts in synaptic input. Another is sory deprivation. As one might imagine, these
that synapse number should vary as a function experiments have yielded complex and (occa-
of overall activity. A third is that the rules that sionally) contradictory results.
govern Hebbian plasticity should themselves
incorporate homeostatic features, an idea called Excitatory Synapses
metaplasticity. But the idea that has perhaps Pyramidal neurons. By far the majority of exper-
received the most attention is synaptic scaling. imental studies of synaptic scaling have focused
In its simplest form, synaptic scaling holds that on plasticity of excitatory synapses onto excitatory
the strengths of all of a neurons excitatory syn- neurons. As of this writing, there are dozens of
apses should be scaled up (or down) as the neu- examples of strengthening (or weakening) of glu-
rons average firing rate goes down (or up; Lissin tamatergic synapses following prolonged reduc-
etal., 1998; OBrien etal., 1998; Turrigiano etal., tions (or increases) in activity levels in networks
1998). Since its introduction more than 15years of neurons. One of the earliest efforts (Turrigiano
ago, synaptic scaling has been extensively stud- etal., 1998)still serves as an exemplar. This study
ied experimentallyin cell cultures, slice cul- used dissociated cultures of neocortical neu-
tures, acute brain slices, and in vivoand its rons. Such cultures are useful for several rea-
simplest form has been elaborated to include sons:individual neurons can be easily visualized
contributions from inhibitory synapses and to and identified morphologically; the environment
allow it to act on different spatial scales and time- in which neurons develop is under experimen-
scales (Turrigiano, 2008; Pozo and Goda, 2010; tal control; and, perhaps most important, cul-
Davis, 2013). Many of these findings have been tures exhibit pronounced spontaneous spiking
further examined using computational mod- and synaptic activity, with firing rates of several
els (van Rossum etal., 2000; Renart etal., 2003; spikes per second. Pharmacological manipula-
Savin et al., 2010; Tetzlaff et al., 2013). In this tions of spontaneous activity can consequently
chapter we provide a succinct overview of much be used to explore activity-dependent plasticity.
of the work to date, focusing on evidence from Adopting this strategy, Turrigiano etal. reduced
110 Part II: Homeostatic Control

activity for one or more days using the sodium But the canonical story is incomplete, and
channel blocker tetrodotoxin or the -amino- portions may well be wrong. Indeed, every part
3-hydroxy-5-methyl-4-isoxazolepropionic acid has been challenged to some degree. The biggest
(AMPA) receptor antagonist CNQX, or increased challenge has been the discovery of a form of syn-
activity for one or more days using the GABA A aptic scaling that acts rapidly and locally, possibly
antagonist bicuculline, and then characterized even at individual synapses (Ju etal., 2004; Sutton
excitatory synaptic strengths by measuring the et al., 2004, 2006; Aoto et al., 2008; Hou et al.,
amplitude and frequency of miniature excitatory 2008; Soden and Chen, 2010; Chen etal., 2014).
postsynaptic currents (mEPSCs), which were car- This form depends not only on action potential
ried by AMPA receptors. They found that mEPSC firing but on synaptic activation; in cultures, this
amplitude (quantal amplitude) was bidirection- means treatment with both tetrodotoxin and glu-
ally regulated by average activity: low activity tamate antagonists. Rather than transcription,
regimes pushed mEPSC amplitudes up over a the scaling is accomplished by local protein syn-
time scale of hours, whereas high activity regimes thesis, dependent on retinoic acid, which inserts
pushed them down. These changes in amplitudes homomeric GluA1 receptors at postsynaptic sites
were not accompanied by changes in mEPSC fre- (Chen et al., 2014). A couple of features of this
quency. Moreover, the changes in quantal ampli- type of scaling are surprising. One is how local
tude appeared to be multiplicative:the data were its actions are. Amajor idea about synaptic scal-
consistent with the synaptic plasticity process at ing has been that its global, multiplicative nature
work scaling all excitatory synapses up (or down) allows it to regulate overall synaptic strength
after activity deprivation (or enhancement) by a without affecting the relative strengths of differ-
single multiplicative factor. ent synapses (Nelson and Turrigiano, 2008). This
Experimental studies like this, in dissoci- is thought to be important because, if information
ated and slice cultures, gave rise to what might is stored in relative synaptic strengths, scaling
be called the canonical version of synaptic scal- would be able to perform a homeostatic function
ing (Figure 7.1): prolonged changes in average while preserving stored information. Put another
neuronal activity trigger opposing (compensa- way, homeostatic plasticity and Hebbian plastic-
tory) changes in excitatory synaptic strength. The ity might coexist without interfering with each
canonical story has several parts. Scaling is slow, other. Local synaptic scaling complicates the sit-
with a natural timescale of hours (Turrigiano uation considerably. One intriguing idea is that
et al., 1998; Thiagarajan et al., 2005; Stellwagen dendritic branches, rather than the neuron as a
and Malenka, 2006). Scaling is global and multi- whole, should be considered the basic computa-
plicative; it depends on somatic firing and affects tional unit (Branco and Hausser, 2010); local scal-
all of a neurons excitatory synapses (Burrone ing, as long as it is not too local, could then work
etal., 2002; Harms and Craig, 2005; Ibata etal., together Hebbian mechanisms (Rabinowitch and
2008; Goold and Nicoll, 2010). Scaling is expressed Segev, 2008). Asecond surprise is that local scal-
postsynaptically, as an increase or decrease in ing requires manipulations of both spiking and
the accumulation of glutamate receptorsboth receptor activation. Moreover, the receptor acti-
AMPA and N-methyl-D-aspartateat postsyn- vation in question is sensitive to spontaneous
aptic sites (Watt et al., 2000; Wierenga et al., rather than spike-evoked vesicle release, and the
2005; Cingolani etal., 2008; Gainey etal., 2009; spiking requirement may be on presynaptic neu-
Anggono et al., 2011). AMPA receptor scaling rons rather than postsynaptic ones (Sutton and
requires the GluA2 subunit of the AMPA recep- Schuman, 2009). These features raise the ques-
tor and results in a coordinated increase in the tion of under what conditions local scaling acts.
number of GluA1/GluA2-containing receptors Possibilities include very early in development,
(Wierenga et al., 2005; Cingolani et al., 2008; when synapses are just forming, or after some
Gainey etal., 2009; Anggono etal., 2011). Scaling kind of pathological insult.
is a cell-autonomous process; it can be triggered Although postsynaptic changes are widely
by manipulating the activity of a single neuron, seen following activity manipulation, many
regardless of the behavior of the network in which studies indicate that under some conditions
it is embedded (Burrone etal., 2002; Ibata etal., chronic activity manipulation results in coor-
2008; Goold and Nicoll, 2010). Scaling depends dinated presynaptic and postsynaptic modi-
on intracellular calcium and requires transcrip- fications (Burrone et al., 2002; De Gois et al.,
tion (Ibata et al., 2008; Goold and Nicoll, 2010; 2005; Thiagarajan et al., 2005; Erickson et al.,
Qiu etal.,2012). 2006; Wierenga et al., 2006; Gong et al., 2007;
Homeostatic Synaptic Scaling at Central Synapses 111

SC
AL
ING
P
LT
High Activity

LT NG
D ALI
Target Activity SC Target Activity

Low Activity

FIGURE7.1: Excitatory synaptic scaling. In its simplest form, scaling involves increasing or decreasing the num-
ber of excitatory receptors (red markers) at dendritic spines (blue protrusions) in order to keep spiking activity
near a target level. The number of receptors at all synapses are scaled up or down by the same multiplicative
factor, thus maintaining relative differences (ratios) between synapses. So, for example, synaptic scaling can
restore target activity while preserving changes in synapses induced by long-term potentiation (LTP) or long-term
depression(LTD).

Jakawich et al., 2010; Lindskog et al., 2010; a CaMKK/CaMKIV pathway are implicated
Lazarevic et al., 2011; Henry et al., 2012). The (Ibata et al., 2008; Gainey et al., 2009; but see
presynaptic modifications can include changes Goel etal.,2011).
in both release probability and glutamate trans- The question of whether or not scaling is
porter expression, which might in turn affect cell autonomous has received a lot of attention
how much neurotransmitter is packaged in (Turrigiano, 2012). Most studies suggest that it
each vesicle. Whether presynaptic modifications is. For example, manipulations of spiking firing
accompany postsynaptic ones may depend on and/or depolarization in individual neurons can
developmental age. Wierenga etal. (2006) found elicit scaling, even when the network as a whole is
that, in young cultures (<2 weeks in vitro), scal- unchanged (Burrone etal., 2002; Ibata etal., 2008;
ing was exclusively postsynaptic in origin but Goold and Nicoll, 2010). However, there is evi-
that, in older cultures, scaling included both dence that presynaptic activity and network-level
presynaptic and postsynaptic elements. Adiffer- activity are also important (Rutherford et al.,
ent set of studies, using hippocampal cultures, 1998; Stellwagen and Malenka, 2006; Jakawich
suggested that presynaptic/postsynaptic scaling et al., 2010). The network argument rests espe-
may involve different physical mechanisms than cially on the fact that two soluble, diffusible fac-
postsynaptic-only scaling (Thiagarajan et al. tors, brain-derived neurotrophic factor (BDNF)
2005; Lindskog et al., 2010; Groth et al., 2011). and the cytokine tumor necrosis factor alpha
These studies demonstrated that when presynap- (TNF), play important signaling roles for syn-
tic scaling is present, the new postsynaptic recep- aptic scaling. BDNF and TNF are released into
tors inserted in the membrane are homomeric the extracellular space by pyramidal neurons and
GluA1 receptors and that this process involves glial cells, respectively. How widely these factors
a pathway including the kinase CaMKII. spread is uncertain, but their places in the scaling
Both of these features are different than what process suggest that a neuron may respond not
has been reported for postsynaptic-only scal- only to its own average activity but to that of its
ing, in which GluA2-containing receptors and near neighbors.
112 Part II: Homeostatic Control

It is likely that part of the reason so many Scaling up and scaling down. As should be
different answers have been given to the ques- clear from the previous discussion, the molecular
tions surrounding synaptic scaling is that underpinnings of excitatory synaptic scaling are
the questions have mainly been examined in varied and a matter of considerable uncertainty.
culture systems, which are, perhaps, in some For example, consider what is known about
respects too artificial. Over the past dozen global, postsynaptic-only scaling. The triggering
years there have been a number of studies event for both scaling up and scaling down is an
examining the effects of chronic sensory dep- activity-dependent change in somatic calcium
rivation in vivo on synaptic properties in sen- influx that then affects signaling through a
sory cortex (Desai et al., 2002; Maffei et al., CaMKIV pathway (Ibata etal., 2008; Goold and
2004; Goel et al., 2006; Goel and Lee, 2007; Nicoll, 2010). CaMKIV is a protein kinase that
Maffei and Turrigiano, 2008; Goel etal., 2011; is part of the larger Ca2+/calmodulin-dependent
Hengen etal., 2013; Keck etal., 2013). The gen- family, which includes CaMKII and which
eral approach has been to subject animals to is closely associated with forms of synaptic
sensory deprivation and then to prepare brain plasticity that regulate receptor synthesis
slices in order to probe synaptic transmission. and trafficking (Wayman, 2008). It becomes
These studies have clearly demonstrated that active with phosphorylation by CaMKK and
a process very much like the synaptic scaling autophosphorylation following Ca2+/calmodulin
previously observed in culture operates in the binding. But downstream of CaMKIV, scaling
intact brain, but they have also shown that up and scaling down seem to diverge. Scaling
scaling is even more complicated than the cul- up depends on the actions of BDNF, TNF,
ture experiments suggest. In particular, scal- the immediate early gene Arc, and the AMPA
ing is sensitive to cell type, developmental age, receptor binding protein PICK1, whereas scaling
and the precise nature of the sensory depriva- down does not (Rutherford et al., 1998; Leslie
tion (e.g., lid suture versus intraocular tetrodo- et al. 2001; Shepherd et al., 2006; Stellwagen
toxin injection versus dark rearing). Moreover, and Malenka, 2006). Rather scaling down has
sensory deprivation in vivo engages not only roles for polo-like kinase 2, the immediate
synaptic scaling but Hebbian plasticity, mak- early gene Homer1a, and a ubiquitin pathway
ing interpretation of the experimental results regulated by Eph4A (Seeburg et al., 2008; Hu
somewhat challenging. etal., 2010; Fu etal., 2011). Furthermore, scaling
Interneurons. The data on how prolonged up is completely dependent on the scaffold
activity manipulation affects excitatory synapses postsynaptic density protein 95, whereas scaling
onto inhibitory interneurons are limited and down can be supported by either postsynaptic
somewhat contradictory. Using culture systems density protein 95 or postsynaptic density protein
and pharmacological activity manipulation, 93 (Sun and Turrigiano, 2011). It seems then that
three different groups have reported that the signaling pathways for scaling up and scaling
days-long activity blockade increases excitatory down are independent, so that activation of the
currents onto interneurons (Bartley etal., 2008), one might be accompanied by deactivation of the
decreases excitatory currents onto interneurons other. The implications of such a scenario have
(Chang etal., 2010), or has no effect (Rutherford yet to be elucidated.
etal., 1998). Two of these studies also examined
the effects of activity enhancement by incubation Inhibitory Synapses
in bicuculline; both reported an increase in If the main purpose of synaptic scaling was to
excitatory mEPSC amplitude (Rutherford et al., implement firing rate homeostasis, then one
1998; Chang et al., 2010). It is not clear how to would expect it should act on inhibitory synapses
reconcile these results. The differences may stem in the opposite way it acts on excitatory ones.
from differences between the neocortex and Considerable evidence suggests that, broadly
hippocampus, duration of activity manipulation, speaking, this is the case, though the picture
or type of interneuron examined. However, two is complicated by the considerable diversity of
of the groups found opposite results for a single interneuronal types (Rutherford et al., 1997;
class of interneuron, parvalbumin-expressing Kilman etal., 2002; Maffei etal., 2004; Hartman
fast-spiking interneurons (Bartley et al., etal., 2006; Bartley etal., 2008; Kim and Alger,
2008; Chang et al., 2010). Certainly this is a 2010; Peng etal., 2010; Rannals and Kapur, 2011;
subject would benefit from a more varied and Saliba et al., 2007; Sarti et al., 2013). Inhibitory
systematicstudy. scaling has been studied in more or less the same
Homeostatic Synaptic Scaling at Central Synapses 113

way as excitatory scaling:through pharmacologi- 2008). Efforts to understand the functional con-
cal manipulations in neocortical and hippocam- sequences of synaptic scaling and to incorporate
pal cultures or in vivo sensory deprivation in it into computational models of neural function
sensory cortex. Together these studies indicate have been informed by the fundamental ideas of
that inhibitory synaptic strengths onto pyrami- control theory and have, increasingly, begun to
dal neurons are bidirectionally regulated by pro- utilize its formalism (Davis, 2006; OLeary and
longed changes in activity levels:downregulated Wyllie, 2011; Queenan etal., 2012; Davis,2013).
after blockade, upregulated after enhancement. The basic problem control theory addresses
The regulation appears to involve both postsyn- is illustrated in Figure 7.2A. A controlled sys-
aptic and presynaptic modifications, including tem (sometimes called a process or a plant)
changes in receptor density, vesicle release, and receives inputs, which may include feedforward,
neurotransmitter packaging. However, there feedback, and noise/perturbation components.
have been some deviations from this simple It transforms these, through some unspecified
story, with increases in some classes of inhibi- means, into an output. The goal of control theory
tory synapse reported after manipulations that is to maintain the output as near to a target (or
should reduce or least decorrelate network activ- set-point) value as possible, responding quickly to
ity (Maffei etal., 2004; Kim and Alger,2010). external perturbations, noise, and changes in the
As with excitatory scaling, there are also open target value. It does this through both feedfor-
questions about whether inhibitory scaling is cell ward and feedback control. Feedforward inputs
autonomous and global. Hartman et al. (2006) may depend on the target and may even monitor
demonstrated that chronic activity blockade in noise/perturbations, but they are independent of
hippocampal cultures downregulates inhibition the actual output, whereas feedback inputs are
onto principal cells but that this only happens adjusted based on an error signal correspond-
when the activity of multiple neurons is blocked; ing to the difference between the measured out-
silencing individual presynaptic or postsynaptic put and the target value. To illustrate how these
neurons was not sufficient to induce inhibitory two types of controls work, consider a simple
scaling. This suggests that inhibitory scaling case: say that we want to use a heater to main-
is not cell autonomous but is instead a network tain the temperature of a room at some fixed level
property. On the other hand, Peng etal. (2010), throughout the course of a day. We know that the
also using hippocampal cultures, found that outside temperature will be colder in the morn-
enhancing activity in multiple ways (treatment ing than in the afternoon, and so one strategy we
with bicuculline or kainic acid, or overexpression might employ is simply to keep the heater on high
of a cation channel) upregulated inhibitory cur- early in the day and then turn it down low later
rents and that only the postsynaptic pyramidal on. This is an example of feedforward (or open
neurons activity mattered for this effectthat loop) control since we used only our prior knowl-
is, scaling here was cell autonomous. Moreover, edge of how the system behaves and did not actu-
as is true of excitatory scaling, alongside global ally measure the rooms temperature, much less
inhibitory scaling, there appears to be a rapid, adjust the heater setting based on this measure-
local, protein- and retinoic aciddependent ment. Feedforward control, while simple, is quite
form of inhibitory scaling (Sarti etal., 2013). The limited and fragile; it requires a good knowledge
relationship between the two forms of scaling of system properties and may not fully compen-
remains ambiguous. sate for perturbations (e.g., the unexpected open-
ing of a window). Amuch more robust strategy
F U N C T I O N A L I M P L I C AT I O N S would be to monitor room temperature (output),
compare it to our desired temperature (set point),
Synaptic Scaling asPart and continually adjust the heater setting based on
ofControlTheory the difference (error signal). This is an example of
Homeostatic regulation is, of course, not what is called feedback (or closed loop) control.
restricted to biological systems. Rather it is a Synaptic scaling, which is after all activity
recurring feature of complex systemsbiological, dependent, is generally thought to be an example
chemical, and physicalin which inputs are of a feedback control mechanism. To understand
transformed into outputs in the presence of noise it in this way, we ask three questions: (a) What
and external perturbations. The general theory is the process (system under control)? (b) How
of how such systems behave and are regulated is the error signal formed? (c) How is the error
is called control theory (strm and Murray, signal used to determine a control signal that
114 Part II: Homeostatic Control

(a) Noise/
(b)
Perturbations
Feedforward
model
Proportional
(Present)
Target Controlled System
(Set Point) (Process)
Target Integral + Controlled System
+ +
(Set Point) (Past) + (Process)
Feedback
Controller
Derivative
(Future)

FIGURE7.2: Synaptic scaling as part of control theory. (A)Control theory concerns how a system that receives
inputs and transforms these into an output maintains the output near a target or set point value in the presence of
noise and other perturbations. Control mechanisms can be divided into two generic types:feedforward and feed-
back. Feedforward control signals depend on the target value and in some cases the noise and perturbation inputs;
they do not explicitly depend on the systems output. Feedback control signals depend on an error signal formed
by the difference between the system output and the desired target output. To make the distinction concrete,
consider early development of the cortex. One expects the number of excitatory synapses each neuron receives
to grow dramatically over time. One strategy for maintaining stable output is to gradually reduce all synaptic
strengths over developmental time. Such a strategy requires prior knowledge (a model) of how synapse forma-
tion and maturation proceeds across development and constitutes an example of feedforward control. Adifferent
strategy is to monitor firing rates directly and to adjust synaptic strengths continually in order to keep firing rates
within appropriate limits; this is feedback control. Synaptic scaling, as a process that depends on neural activity,
is thought to be a feedback process. (B)Feedback control can take many forms, but the most generic description
is the proportional-integral-derivative controller. This description divides feedback controls dependence on the
error (difference between target output and actual output) into three parts, which are sensitive to the present error
(proportional), the accumulation of past errors (integral), and an estimate of future errors (derivative).

appropriately adjusts the feedback input in order Branco etal., 2008; Hou etal., 2008; Bque etal.,
to achieve control? 2011). This idea is attractive because it prevents
The process for synaptic scaling depends on any one branch from dominating over the others,
the spatial scale at which one imagines it oper- thus preserving the computational power inher-
ates. Most work, both experimental and compu- ent in complex dendritic arbors, and it allows
tational, has taken the process to be the single for the selection of spatial patterns of input that
neuron (Turrigiano, 2008). Its inputs are the syn- are particularly efficacious in controlling neu-
aptic currents, and its output is the firing rate or ronal output (Rabinowitch and Segev, 2008).
its pattern of action potentials. This is a natural A differentand equally attractivealternative
choice because of the experiments in culture that is to consider the neural circuit as a whole to
indicate individual neurons can respond to per- be the process (Maffei and Fontanini, 2009). In
turbations independent of the network in which this case the output is some measure of network
they are embedded and that identify somatic activity, such as the average firing rate of all the
firing as the important variable (Burrone et al., individual neurons that make up the network.
2002; Ibata etal. 2008; Goold and Nicoll, 2010). It This idea is plausible because of evidence that dif-
is also a choice that is straightforward to imple- fusible factors, such as BDNF and TNF, might
ment in a computational model (van Rossum serve as signaling molecules for synaptic scaling
et al., 2000; Renart et al., 2003). However, it is (Stellwagen and Malenka, 2006; Lindskog etal.,
important to be aware that this is not the only 2010); the spatial range over which they diffuse
choice possible. One attractive alternative is to would then set the scale for the circuit considered
assume that the process is an individual den- to make up the process.
dritic branch (or an even smaller structure); its To form an error signal one needs two
inputs are again synaptic currents but its output things:an observable signal related to the output
now is some measure of local (restricted to the and a target value against which to compare it. As
branch) synaptic activity (Sutton et al., 2006; is often true in biology, the obvious candidate for
Homeostatic Synaptic Scaling at Central Synapses 115

an observable signal is internal calcium, an ion evidence indicates that synaptic scaling is a
that closely tracks action potential and synaptic slow process, acting over hours to days, which
activity and that is known to play a fundamental proportional control does not readily accom-
role in many cellular processes. Other possibili- modate. Accordingly, computational modeling
ties exist, such as active monitoring of receptor of synaptic scaling sometimes includes an inte-
accumulation, but most of the focus in stud- gral term, which accumulates errors over time,
ies of this question has been on calcium and its giving scaling a long history dependence and
downstream processes (Thiagarajan etal., 2005; eliminating offset (van Rossum et al., 2000;
Ibata etal., 2008; Goold and Nicoll, 2010; Zhao Rowan and Neymotin, 2013; Yger and Harris,
et al., 2011). Much more uncertain is how the 2013). However, it is somewhat more difficult to
target value or set point is established. It would understand how a real neuron or neural circuit
seem likely that the set point must (somehow) be implements integral control. Although calcium is
encoded genetically, with variations between dif- itself a temporally filtered measure of spiking and
ferent cell types and developmental ages (Davis, synaptic activity, the timescale of the filtering is
2013). But, despite considerable progress in short (no more than seconds). Instead, attention
unraveling the molecular machinery of synaptic has been focused on calcium-dependent pro-
scaling and some intriguing ideas, the question cesses that may act on timescales much slower
of set point remains almost entirelyopen. than calcium itself (OLeary and Wyllie, 2011).
The central problem of control theory is how to The derivative control term has mostly not been
use an error signal to generate an appropriate con- used in modeling synaptic scaling because it
trol signal. The most generic algorithm for doing tends to make models very sensitive to noise and
this is called the proportional-integral-derivative it is difficult to justify biologically.
controller (strm and Murray, 2008). This algo-
rithm is used widely in engineering and can be ComputationalIdeas
captured by a simple equation (Figure7.2B): Synaptic scaling has been incorporated into
numerous computational models of corti-
cal function, both at the single-cell level and
f (t ) = K p * e(t ) + K i * e(t )dt+K d * de(t ) / dt
at the network level (van Rossum etal., 2000;
Renart et al., 2003; Sullivan and de Sa, 2006;
where f(t) is the control signal; KP, Ki, and Lazar et al., 2009; Liu and Buonomano, 2009;
Kd are constants; and e(t) is the error signal Rossi Pool and Mato, 2010; Tetzlaff et al.,
(= target output). The control signal might 2011; Keck etal., 2012; Rowan and Neymotin,
directly represent a feedback input (e.g., current) 2013; Yger and Harris, 2013). It has been
or an intermediate quantity (e.g., rate of change implemented in various ways, including the
of synaptic weights) that indirectly modulates proportional-integral-derivative formulation
feedback input. The three terms at the right of given previously, schemes in which the rate of
the equation represent the present error (propor- synaptic weight change depends on some func-
tional), the accumulation of past errors (integral), tion of current synaptic weights, and schemes
and an estimate of future errors (derivative). The in which synaptic weights are directly normal-
first term is the easiest to understand. It is posi- ized (Dayan and Abbott, 2005; Lazar et al.,
tive when output is below the target value and 2009; Tetzlaff etal., 2013). The modeling stud-
negative when it is above, with a magnitude that ies have identified several distinct functions
scales with the size of the discrepancy. It is also synaptic scaling might serve. In this subsection
fairly easy to understand how a proportional we highlight five ofthem.
error might be implemented biologically, espe- Firing rate homeostasis. The original
cially if the error signal was formed by internal motivation for studying synaptic scaling was
calcium; many biological processes relevant to to identify a biophysical mechanism that might
synaptic strength depend on calcium concentra- mediate firing rate homeostasis in cortical
tion. However, proportional control alone may networks (Turrigiano et al., 1998). Subsequent
not capture quite how synaptic scaling works. computational studies of small networks of
Proportional control will always leave the out- spiking neurons confirmed that this is indeed a
put short of the target by some finite amount, role scaling might play (Sullivan and de Sa, 2006;
and it is prone to instability if the proportional Tetzlaff et al., 2011, 2012; Rowan and Neymotin,
gain K P is set too high (strm and Murray, 2013). However, these same studies indicated that
2008). Moreover, most of the experimental the details of how scaling is implemented can
116 Part II: Homeostatic Control

matter a great deal as to whether stabilization of (2000) who found that combining synaptic
firing rates is actually achieved. In particular, to scaling with spike timing-dependent plasticity
ensure stability in the most general case, in the resulted in neural circuits that were stable and
presence of Hebbian plasticity and with generic had synaptic weight distributions that were
neuronal dynamics, synaptic scaling must not approximately log-normal (Tetzlaff et al., 2011),
only be weight-dependent but that dependence which compares well with quantal amplitude
must be strongly nonlinear (e.g., quadratic; Tetzlaff distributions measured experimentally (Song
etal., 2011, 2012). The linear multiplicative rule for etal.,2005).
synaptic scaling proposed originally (Turrigiano Working memory. Persistent activity in neural
etal., 1998)does not meet this requirement, but it circuits, particularly of the prefrontal cortex,
may be that experiments to date have simply not is thought to be the physical basis of working
been sensitive enough to determine the exact form memory (Fuster and Alexander, 1971; Miller etal.,
scaling takes or that scaling works in combination 1996; Wang, 2001; Baeg et al., 2003). A number
with some other form of homeostatic plasticity of mechanisms for generating and maintaining
(i.e., intrinsic plasticity) to stabilize neural circuits persistent activity have been proposed, including
(Lazar etal., 2009; Savin etal.,2010). slow, recurrent excitatory connections mediated
A different issue is that of timescale. Most by N-methyl-D-aspartate receptors (Wang, 2001;
studies of synaptic scalingbut not allindicate Egorov etal., 2002). Aweakness of some of these
that scaling happens slowly, over the course of proposals is that, in the absence of exquisite
hours (Turrigiano etal., 1998; Sutton etal., 2004, parameter fine-tuning, they require unrealistic
2006; Thiagarajan et al., 2005; Stellwagen and assumptions about the uniformity of neuronal
Malenka, 2006; Hou et al., 2008). This may be and synaptic properties (Wang, 2001). Real
very different than the timescale of Hebbian plas- neural networks lack such homogeneity. Using
ticity; experimental studies of long-term poten- a biologically realistic network model, including
tiation and depression indicate that they can be plausible heterogeneities, Renart et al. (2003)
induced in just minutes (Sjstrm et al., 2008). demonstrated that synaptic scaling, operating
The temporal mismatch might make it difficult on each neuron independently, can effectively
for the former to counter the latter in some cases. compensate for heterogeneities in cellular
In particular, a recent study by Zenke etal. (2013) excitability and synaptic properties, and it can
suggests that unless scaling can act within min- allow for robust persistent activity.
utes, it cannot effectively stabilize the activity of Short-versus long-term memory storage.
a generic, balanced network; the authors came Memory storage depends on biological
to this conclusion using both numerical simula- mechanisms, like long-term potentiation and
tions and a mean field model. Afew caveats are in depression, that act on a time scale of minutes; and
orderthe nature of the network (connectivity, yet memory consolidation is a process that can take
excitation-inhibition balance, external inputs) days. How to reconcile these different timescales is
and the form plasticity takes can matter a great a difficult problem because it requires that neural
deal (as is true for all models)but these results circuits have some means of separating synapses
do suggest that to counter the positive feedback relevant for long-term storage, which should be
instability introduced by Hebbian plasticity, consolidated, from synapses relevant for short-term
some homeostatic mechanism other than scal- storage, which should fade over time. Tetzlaff etal.
ing might be required. Examples of fast homeo- (2013) show that synaptic scaling, working on
static mechanisms are heterosynaptic plasticity an intermediate timescale (hours), might bridge
(Chistiakova et al., 2014) and weight-dependent the time gap between long-term potentiation/
Hebbian plasticity (Van Rossum etal.,2000). long-term depression and memory consolidation.
Synaptic competition. Global synaptic scaling In particular, they argued that neural circuits
makes plasticity at any one synapse depend on the that combine synaptic scaling with Hebbian
activity environment created by all of the other plasticity intrinsically exhibit a bifurcation in their
synapses. In particular it weakens quiet synapses dynamics that provides a natural way of separating
relative to active ones. As such, scaling might potentiated synapses bound for long-term storage
be a way of implementing competition between from those consigned to short-term storage.
synapses, which is thought to be important Sleep. The purpose of sleep is a matter of
for development of neural circuits (e.g., ocular debate, but an idea that has attracted much
dominance plasticity; Bienenstock et al., 1982). recent attention is that sleep promotes an overall
This idea was explored by van Rossum et al. synaptic weakening that offsets the synaptic
Homeostatic Synaptic Scaling at Central Synapses 117

strengthening that occurs during wakefulness but complementary. Using hippocampal dis-
(Tononi and Cirelli, 2014). Called the synaptic sociated cultures, Qui and colleagues found
homeostasis hypothesis, the mechanism of this that chronic (days) activity enhancement, by
weakening is not clear. Synaptic scaling has application of the GABA A antagonist bicucul-
been proposed as a candidate, though two lines line, increased expression of MeCP2 mRNA
of evidence argue against this view: changes and decreased average mEPSC amplitude onto
during sleep in the expression of some molecular pyramidal neurons. When MeCP2 expres-
factors tied to synaptic scaling, including BDNF, sion was acutely knocked down using an RNA
TNF, and Arc, are in the wrong direction interference method, the synaptic downscaling
to promote downscaling (Frank, 2012), and was blocked in those neurons underexpress-
firing patterns during sleep are complicated ing MeCP2. Conversely, using neocortical dis-
and may not be consistent with downscaling sociated cultures, Blackman et al. showed that
(Frank, 2012; Hengen et al., 2013; Tononi and chronic activity blockade, by application of the
Cirelli,2014). AMPA receptor antagonist DNQX, led to an
increase in mEPSC amplitude onto pyramidal
SY NAPTIC SCALING neurons that was absent when an RNA interfer-
AND DISORDERS ence method was used to acutely knock down
Although synaptic scaling has mainly been stud- MeCP2. Together these studies indicate that
ied in the context of normal brain function, normal MeCP2 function is necessary for bidi-
much recent work has explored ideas about how rectional synaptic scaling acting in a cell auton-
scaling might act under pathological conditions, omous fashion and suggest that loss of scaling
both in the young and in theold. is part of pathological postnatal development in
this disorder.
Neurodevelopment Fragile X syndrome. Fragile X syndrome is the
Neurodevelopmental disorders are complex and most common inherited form of mental
have a great variety of molecular components, retardation and the most common known
so it is remarkable that synaptic scaling has cause of autism spectrum disorder (Bagni
been directly linked with two of the best-studied and Greenough, 2005). Like Rett syndrome, it
experimental models:Rett syndrome and fragile is produced by the absence of single protein,
X syndrome (Henry, 2011; Na etal.,2013) fragile X mental retardation protein (FMRP).
Numerous synaptic and neuronal abnormalities
Rett syndrome. Rett syndrome is the leading have been reported to be associated with loss
cause of mental retardation in females, with some of FMRP, including those affecting long-term
girls also exhibiting autistic-like features such as and short-term synaptic plasticity, synaptic
a loss of social interaction and communication morphology, and the distribution of dendritic
(Chahrour and Zoghbi, 2007). It is produced ion channels (Bassell and Warren, 2008; Brager
by loss of function mutations in a single gene, et al., 2012; Routh et al., 2013). How FMRP,
which codes for the transcriptional regulator which is located near synaptic sites and acts as a
methyl-CpG binding protein 2 (MeCP2). Loss of translational regulator, affects synaptic scaling
MeCP2, an X-linked protein, is devastating:there is complicated. Using mutant mice in which
are few live male births in humans, and the Fmr1, the gene that codes for FMRP, had been
survivors die of encephalopathy soon after birth, knocked out, Soden and Chen (2010) found that
while females develop debilitating and eventually FMRP was necessary for the form of synaptic
fatal symptoms within the first year after birth. scaling that is mediated by retinoic acid and acts
The severity of the effects of MeCP2 loss and its locally through local protein translation (Aoto
delayed onset suggest that it plays a critical role et al., 2008). However, FMRP was apparently
in postnatal brain development. Even so, most of not necessary for the transcription-dependent,
the reported neural and synaptic abnormalities global form of synaptic scaling (Turrigiano
in MeCP2-knockout mice are surprisingly mild etal., 1998). The relationship between these two
(Na et al., 2013). One exception is synaptic types of scaling is not clear, as they may well
scaling. be mediated by different physical mechanisms,
Two groups have independently demon- with the local form sensitive to spontaneous
strated that MeCP2 plays a critical role in bidi- vesicle release and the global form to
rectional synaptic scaling (Blackman et al., spike-evoked release (Ibata et al., 2008; Sutton
2012; Qiu etal, 2012). Their results are distinct and Schuman,2009).
118 Part II: Homeostatic Control

Stroke et al., 2005). Both of these effectsdecreased


Stroke is caused by interruptions in the blood BDNF and increased TNFwould tend to
supply to the brain, which result in struc- upregulate AMPA receptor levels through synap-
tural and functional damage to oxygen- and tic scaling.
energy-deprived neurons. Recovery after stroke
is a complex process, including behavioral com- CONCLUDING REMARKS
pensation, rewiring, and synaptic plasticity Even before there was any experimental evi-
(Murphy and Corbett, 2009). The first steps in dence for synaptic scaling, it was anticipated on
recovery appear to involve restoration of neural general theoretical grounds by the need of neu-
activity in the peri-infarct region (i.e., the tis- ral networks for some mechanism of firing rate
sue surrounding the area of greatest damage), homeostasis and limiting synaptic strengths
which might create a permissive environment (Miller and MacKay, 1994). It was not the only
for axonal sprouting and dendritic spine for- candidate homeostatic mechanism (see, e.g.,
mation. The activity appears shortly (days) after Bienenstock et al., 1982), but it was among the
the initial stroke. This time course suggests that simplest and most direct. Experiments over the
synaptic scaling may be the underlying physi- past 15 or more years have demonstrated that
cal mechanism (Murphy and Corbett, 2009). synaptic scalinglike processes exist in a wide
Evidence from rodent models that this is true variety of neurobiological systems, including in
includes the poststroke hyperexcitability in the the neocortex, hippocampus, and spinal cord;
weeks immediately following injury and the tran- in mammal, Xenopus, and Drosophila; and in
sient appearance of low-frequency spontaneous young animals and mature animals. Ongoing
activity (0.11.0 Hz; Carmichael and Chesselet, experiments have elucidated the properties of
2002; Schiene etal., 1996; Winship and Murphy, synaptic scaling in considerable detail and dem-
2008). Also, local inflammation is common after onstrated that its phenomenology is rich and
stroke, which might lead to release of TNF. This varies depending on activity manipulation, neu-
cytokine is of course necessary for global synap- robiological system, and age. But perhaps what
tic scaling (Stellwagen and Malenka,2006). is most needed now is a better idea of its func-
tional consequences. Firing rate homeostasis was
Alzheimers Disease the starting pointand remains a focus of cur-
A key event in the neurodegeneration associ- rent workbut, as we have indicated, there are
ated with Alzheimers disease is the accumula- several other possible uses for scaling, including
tion of -amyloid protein in the brain (Walsh synaptic competition and working memory, and
and Selkoe, 2007). -amyloid accumulation is it may play important roles in several neurologi-
toxic to neurons because it affects a number of cal disorders. For technical reasons, experiments
signal-transduction pathways. In particular, it to date have not addressed these issues directly,
disrupts calcium homeostasis, with attendant and that is the most important challenge for
effects on synaptic function and cell death path- futurework.
ways. An intriguing, if speculative, idea is that
synaptic scaling creates a positive feedback loop References
that exacerbates the damage done by -amyloid Abbott, L.F., and Nelson, S.B. (2000). Synaptic plas-
(Small, 2008). The idea is simple: increased ticity:Taming the beast. Nat. Neurosci. 3(Suppl),
-amyloid increases cytoplasmic calcium by 11781183.
disrupting calcium homeostasis, leading to cell Abraham, W.C. (2008). Metaplasticity: Tuning
death; this neurotoxicity reduces synaptic and synapses and networks for plasticity. Nat. Rev.
spiking activity in spared but still vulnerable Neurosci. 9, 387399.
neurons; the reduced activity triggers homeo- Anggono, V. Clem, R.L., and Huganir, R.L. (2011).
static synaptic scaling; scaling boosts network PICK1 loss of function occludes homeostatic
activity and thus increases cytoplasmic calcium synaptic scaling. J. Neurosci. 31, 21882196.
even further, leading to more neuronal death. Aoto, J., Nam, C.I., Poon, M.M., Ting, P., and Chen, L.
Two molecular pieces of evidence support this (2008). Synaptic signaling by all-trans retinoic
idea:oligomeric -amyloid decreases BDNF lev- acid in homeostatic synaptic plasticity. Neuron 60,
els in cultured neurons (Tong etal., 2004; Garzon 308320.
and Fahnestock, 2007), whereas the cytokine strm, K.J., and Murray, R.M. (2008). Feedback
TNF has been found at elevated levels in post- systems: An introduction for scientists and engi-
mortem Alzheimers tissue (Jia et al., 2005; Ho neers. (Princeton, NJ:Princeton University Press).
Homeostatic Synaptic Scaling at Central Synapses 119

Baeg, E.H., Kim, Y.B., Huh, K., Mook-Jung, I., Kim, H.T., Chang, M.C., Park, J.M., Pelkey, K.A., Grabenstatter,
and Jung, M.W. (2003). Dynamics of population H.L., Xu, D., Linden, D.J., Sutula, T.P., McBain,
code for working memory in the prefrontal cor- C.J. and Worley, P.F. (2010). Narp regulates
tex. Neuron 40, 177188. homeostatic scaling of excitatory synapses on
Bagni, C., and Greenough, W.T. (2005). From mRNP parvalbumin-expressing interneurons. Nat.
trafficking to spine dysmorphogenesis:The roots Neurosci. 13, 1090-1097.
of Fragile X syndrome. Nat. Rev. Neurosci. 6, Chen, J.Y., Lonjers, P., Lee, C., Chistiakova, M.,
376386. Volgushev, M., and Bazhenov, M. (2013).
Bartley, A.F., Huang Z.J., Huber, K.M., and Gibson, J.R. Heterosynaptic plasticity prevents runaway syn-
(2008). Differential activity-dependent, homeo- aptic dynamics. J.Neurosci. 33, 1591515929.
static plasticity of two neocortical inhibitory cir- Chen, L., Lau, A.G., and Sarti, F. (2014). Synaptic
cuits. J. Neurophys. 100, 19831994. retinoic acid signaling and homeostatic synaptic
Bassell, G.J., and Warren, S.T. (2008). Fragile X syn- plasticity. Neuropharmacology 78, 312.
drome: Loss of local mRNA regulation alters Chistiakova, M., Bannon, N.M., Bazhenov, M., and
synaptic development and function. Neuron 60, Volgushev, M. (2014). Heterosynaptic plastic-
201214. ity:Multiple mechanisms and multiple roles. The
Bque, J.C., Na, Y., Kuhl, D., Worley, P.F., and Neuroscientist 20, 483498.
Huganir, R.L. (2011). Arc-dependent synapse- Cingolani, L.A., Thalhammer, A., Yu, L.M.,
specific homeostatic plasticity. Proc. Natl. Acad. Catalono, M., Ramos, T., Colicos, M.A., and
Sci. USA 108, 816821. Goda, Y. (2008). Activity-dependent regulation
Berardi, N., Pizzorusso, T., and Maffei, L. (2000). of AMPA receptor composition and abundance
Critical periods during sensory development. by 3 integrins. Neuron 58, 749762.
Curr. Opin. Neurobiol. 10, 138145. Collin, G., and van den Heuvel, M.P. (2013). The ontog-
Bienenstock, E., Cooper, L.N., and Munro, P.W. eny of the human connectome:Development and
(1982). Theory for the development of neuron dynamic changes of brain connectivity across
selectivity and binocular interaction in visual the life span. The Neuroscientist 19, 616628.
cortex. J. Neurosci. 2, 3248. Davis, G.W. and Bezprozvanny, I. (2001).
Blackman, M.P., Djukic, B., Nelson, S.B., and Maintaining the stability of neural function: a
Turrigiano, G.G. (2012). A critical and cell- homeostatic hypothesis. Annu. Rev. Neurosci.
autonomous role for MeCP2 in synaptic scaling 63, 847-869.
up. J. Neurosci. 32, 1352913536. Davis, G.W. (2006). Homeostatic control of neural
Brager, D.H., Akhavan, A.R., and Johnston, D. activity: From phenomenology to molecular
(2012). Impaired dendritic expression and plas- design. Annu. Rev. Neurosci. 29, 307323.
ticity of h-channels in the fmr1(-/y) mouse model Davis, G.W. (2013). Homeostatic signaling and the
of fragile X syndrome. Cell Rep. 1, 225233. stabilization of neural function. Neuron 80,
Branco, T., and Hausser, M. (2010). The single den- 718728.
dritic branch as a fundamental functional unit Dayan, P., and Abbott, L.F. (2005). Theoretical
in the nervous system. Curr. Opin. Neurobiol. neuroscience: Computational and mathemati-
20, 494502. cal modeling of neural systems. (Cambridge,
Branco, T., Staras, K., Darcy, K.J., and Goda Y. MA:MIT Press).
(2008). Local dendritic activity sets release De Gois, S., Schfer, M.K., Defamie, N., Chen, C.,
probability at hippocampal synapses. Neuron Ricci, A., Weihe, E., Varoqui, H., and Erickson,
59, 475485. J.D. (2005). Homeostatic scaling of vesicular glu-
Burrone, J., OByrne, M., and Murthy, V.N. (2002). tamate and GABA transporter expression in rat
Multiple forms of synaptic plasticity triggered neocortical circuits. J.Neurosci. 25, 71217133.
by selective suppression of activity in individual Desai, N.S. (2003). Homeostatic plasticity in the
neurons. Nature 420, 414418. CNS: Synaptic and intrinsic forms. J. Physiol.
Cannon, W.B. (1932). The wisdom of the body Paris 97, 391402.
(NewYork:W.W. Norton.). Desai, N.S., Cudmore, R.H., Nelson, S.B, and
Carmichael, S.T., and Chesselet, M.F. (2002). Turrigiano, G.G. (2002). Critical periods for
Synchronous neuronal activity is a signal for experience-dependent synaptic scaling in visual
axonal sprouting after cortical lesions in the cortex. Nat. Neurosci. 5, 783789.
adult. J. Neurosci. 22, 60626070. Egorov, A.V., Hamam, B.N., Fransn, E., Hasselmo,
Chahrour M., and Zoghbi, H.Y. (2007). The story M.E., and Alonso, A.A. (2002). Graded persistent
of Rett syndrome: From clinic to neurobiology. activity in entorhinal cortex neurons. Nature
Neuron 56, 422437. 420, 173178.
120 Part II: Homeostatic Control

Erickson, J.D., De Gois, S., Varoqui, H., Schafer, inactivity in hippocampal neurons. Proc. Natl.
M.K., and Weihe, E. (2006). Activity-dependent Acad. Sci. USA 108, 828833.
regulation of vesicular glutamate and GABA Harms, K.J., and Craig, A.M. (2005). Synapse com-
transporters: A means to scale quantal size. position and organization following chronic
Neurochem. Int. 48, 643649. activity blockade in cultured hippocampal neu-
Frank, C.A. (2014). Homeostatic plasticity at rons. J. Comp. Neurol. 490, 7284.
the Drosophila neuromuscular junction. Hartman, K.N., Pal, S.K., Burrone, J., and Murthy,
Neuropharmacology 78, 6374. V.N. (2006). Activity-dependent regulation of
Frank, M.G. (2012). Erasing synapses in sleep: Is it inhibitory synaptic transmission in hippocam-
time to be SHY? Neural Plasticity, Article ID pal neurons. Nat. Neurosci. 9, 642649.
264378. Hengen, K.B., Lambo, M.E., Van Hooser, S.D., Katz,
Fu, A.K., Hung, K.W., Fu, W.Y., Shen, C., Chen, Y., D.B., and Turrigiano, G.G. (2013). Firing rate
Xia, J., Lai, K.O., and Ip, N.Y. (2011). APC(Cdh1) homeostasis in visual cortex of freely behaving
mediates EphA4-dependent downregulation of rodents. Neuron 80, 335342.
AMPA receptors in homeostatic plasticity. Nat. Henry, F.E. (2011). A fragile balance at synapses:New
Neurosci. 14, 181189. evidence supporting a role for FMRP in homeo-
Fuster, J.M., and Alexander, G.E. (1971). Neuron static plasticity. J.Neurosci. 31, 66176619.
activity related to short-term memory. Science Henry, F.E., McCartney, A.J., Neely, R., Perez, A.S.,
173, 652654. Carruthers, C.J., Stuenkel, E.L., Inoki, K., and
Gainey, M.A., Hurvitz-Wolff, J.R., Lambo, M.E., Sutton, M.A. (2012). Retrograde changes in pre-
and Turrigiano, G.G. (2009). Synaptic scaling synaptic function driven by dendritic mTORC1.
requires the GluR2 subunit of the AMPA recep- J. Neurosci. 32, 1712817142.
tor. J. Neurosci.29, 64796489. Hensch, T.K. (2004). Critical period regulation.
Garzon, D.J., and Fahnestock, M. (2007). Oligomeric Annu. Rev. Neurosci. 27, 549579.
amyloid decreases basal levels of brain-derived Ho, G.J., Drego, R., Hakimian, E., and Masliah, E.
neurotrophic factor (BDNF) mRNA via specific (2005). Mechanisms of cell signaling and inflam-
downregulation of BDNF transcripts IV and V mation in Alzheimers disease. Curr. Drug
in differentiated human neuroblastoma cells. Targets Inflamm. Allergy 4, 247256.
J.Neurosci. 27, 26282635. Hou, Q., Zhang, D., Jarzylo, L., Huganir, R.L., and
Goel, A., Jiang, B., Xu, L.W., Song, L., Kirkwood, A., Man, H.Y. (2008). Homeostatic regulation of
and Lee, H.K. (2006). Cross-modal regulation AMPA receptor expression at single hippocam-
of synaptic AMPA receptors in primary sensory pal synapses. Proc. Natl. Acad. Sci. USA 105,
cortices by visual experience. Nat. Neurosci. 9, 775780.
10011003. Hu, J.H., Park, J.M., Park, S., Xiao, B., Dehoff, M.H.,
Goel, A., and Lee, H.K. (2007). Persistence of expe- Kim, S., Hayashi, T., Schwarz, M.K., Huganir,
rience-induced homeostatic synaptic plasticity R.L., Seeburg, P.H., Linden, D.J., and Worley,
through adulthood in superficial layers of mouse P.F. (2010). Homeostatic scaling requires group
visual cortex. J.Neurosci. 27, 66926700. I mGluR activation mediated by Homer1a.
Goel, A., Xu, L.W., Snyder, K.P., Song, L., Goenaga- Neuron 68, 11281142.
Vazquez, Y., Megill, A., Takamiya, K., Huganir, Ibata, K., Sun, Q., and Turrigiano, G.G. (2008). Rapid
R.L., and Lee, H.K. (2011). Phosphorylation of synaptic scaling induced by changes in postsyn-
AMPA receptors is required for sensory depri- aptic firing. Neuron 57, 819826.
vation-induced homeostatic synaptic plasticity. Jakawich, S.K., Nasser, H.B., Strong, M.J., McCartney,
PLoS One 6, e18264. A.J., Perez, A.S., Rakesh, N., Carruthers, C.J.,
Gong, B., Wang, H., Gu, S., Heximer, S.P., and Zhuo, and Sutton, M.A. (2010). Local presynaptic activ-
M. (2007). Genetic evidence for the require- ity gates homeostatic changes in pre-synaptic
ment of adenylyl cyclase 1 in synaptic scaling of function driven by dendritic BDNF synthesis.
forebrain cortical neurons. Eur. J.Neurosci. 26, Neuron 68, 11431158.
275288. Jia, J.P., Meng, R., Sun, Y.X., Sun, W.J., Ji, X.M., and
Goold, C.P., and Nicoll, R.A. (2010). Single-cell opto- Jia, L.F. (2005). Cerebrospinal fluid tau, Abeta1
genetic excitation drives homeostatic synaptic 42 and inflammatory cytokines in patients with
depression. Neuron 68, 512528. Alzheimers disease and vascular dementia.
Groth, R.D., Lindskog, M., Thiagarajan, T.C., Li, Neurosci. Lett. 383, 1216.
L., and Tsien, R.W. (2011). Beta Ca2+/CaM- Ju, W., Morishita, W., Tsui, J., Gaietta, G., Deerinck,
dependent kinase type II triggers upregulation T.J., Adams, S.R., Garner, C.C., Tsien, R.Y.,
of GluA1 to coordinate adaptation to synaptic Ellisman, M.H., and Malenka, R.C. (2004).
Homeostatic Synaptic Scaling at Central Synapses 121

Activity-dependent regulation of dendritic syn- neural networks in a self-organizing manner. J.


thesis and trafcking of AMPA receptors. Nat. Neurosci. 29, 1317213181.
Neurosci. 7, 244253. Maffei, A., Nelson, S.B., and Turrigiano, G.G. (2004).
Keck, C., Savin, C., and Lcke, J. (2012). Feedforward Selective reconfiguration of layer 4 visual cortical
inhibition and synaptic scalingtwo sides of the circuitry by visual deprivation. Nat. Neurosci. 7,
same coin? PLoS Comput. Biol. 8, e1002432. 13531359.
Keck, T., Keller, G.B., Jacobsen, R.I., Eysel, U.T., Maffei, A., and Fontanini, A. (2009). Network
Bonhoeffer, T., and Hbener, M. (2013). Synaptic homeostasis: A matter of coordination. Curr.
scaling and homeostatic plasticity in the mouse Opin. Neurobiol. 19, 168173.
visual cortex in vivo. Neuron 80, 327334. Maffei, A., and Turrigiano, G.G. (2008). Multiple
Kilman, V., van Rossum, M.C.W., and Turrigiano, modes of network homeostasis in visual cortical
G.G. (2002). Activity deprivation reduces minia- layer 2/3. J. Neurosci. 28, 43774384.
ture IPSC amplitude by decreasing the number Marder, E., and Goaillard, J.-M. (2006). Variability,
of postsynaptic GABA(A) receptors clustered at compensation and homeostasis in neuron and
neocortical synapses. J. Neurosci. 22, 13281337. network function. Nat. Rev. Neurosci. 7, 563574.
Kim, J., and Alger, B.E. (2010). Reduction in endo- Miller, K.D., and MacKay, D.J.C. (1994). The role
cannibinoid tone is a homeostatic mechanism of constraints in Hebbian learning. Neural
for specific inhibitory synapses. Nat. Neurosci. Comput. 6, 100126.
13, 592600. Miller, E.K., Erickson, C.A., and Desimone, R.
Knudsen, E.I. (2004). Sensitive periods in the devel- (1996). Neural mechanisms of visual working
opment of the brain and behavior. J. Cogn. memory in prefrontal cortex of the macaque. J.
Neurosci., 16, 14121425. Neurosci. 16, 51545167.
Lazar, A., Pipa, G., and Triesch, J. (2009). Murphy, T.H., and Corbett, D. (2009). Plasticity dur-
SORN: A self-organizing recurrent neural net- ing stroke recovery: From synapse to behavior.
work. Front. Comput. Neurosci.3, 23. Nat. Rev. Neurosci. 10, 861872.
Lazarevic, V., Pothula, S., Andres-Alonso, M., and Na, E.S., Nelson, E.D., Kavalali, E.G., and
Fejtova, A. (2013). Molecular mechanisms driv- Monteggia, L.M. (2013). The impact of MeCP2
ing homeostatic plasticity of neurotransmitter loss- or gain-of-function on synaptic plasticity.
release. Front. Cell. Neurosci. 7, 244. Neuropsychopharmacology 38, 212219.
Lazarevic, V., Schone, C., Heine, M., Gundelfinger, Nelson, S.B., and Turrigiano, G.G. (2008). Strength
E.D., and Fejtova, A. (2011). Extensive remodel- through diversity. Neuron 60, 477482.
ing of the presynaptic cytomatrix upon homeo- OBrien, R.J., Kamboj, S., Ehlers, M.D., Rosen,
static adaptation to network activity silencing. J. K.R., Fischbach, G.D., and Huganir, R.L. (1998).
Neurosci. 31, 1018910200. Activity-dependent modulation of synap-
Lee, K.F.H., Soares, C., and Bque, J.C (2014). Tuning tic AMPA receptor accumulation. Neuron 21,
into diversity of homeostatic synaptic plasticity. 10671078.
Neuropharmacology 78, 3137. OLeary, T., and Wyllie, D.J.A. (2011). Neuronal
Leslie, K.R., Nelson, S.B., and Turrigiano, G.G. homeostasis: Time for a change? J. Physiol. 20,
(2001). Postsynaptic depolarization scales quan- 48114826.
tal amplitude in cortical pyramidal neurons. J. Peng, Y.R., Zeng, S.Y., Song, H.L., Li, M.Y., Yamada,
Neurosci. 21, RC170. M.K., and Yu, X. (2010). Postsynaptic spiking
Lindskog, M., Li, L., Groth, R.D., Poburko, D., homeostatically induces cell-autonomous regu-
Thiagarajan, T.C., Han, X., and Tsien, R.W. lation of inhibitory inputs via retrograde signal-
(2010). Postsynaptic GluA1 enables acute ret- ing J. Neurosci. 30, 1622016231.
rograde enhancement of presynaptic function Pozo, K., and Goda, Y. (2010). Unraveling mecha-
to coordinate adaptation to synaptic inactivity. nisms of homeostatic synaptic plasticity. Neuron
Proc. Natl. Acad. Sci. USA 107, 806811. 66, 337351.
Lissin, D.V., Gomperts, S.N., Carroll, R.C., Christine, Pratt, K. and Aizenman, C.D. (2007). Homeostatic
C.W., Kalman, D., Kitamura, M., Hardy, S., regulation of intrinsic excitability and synaptic
Nicoll, R.A., Malenka, R.C., and von Zastrow, M. transmission in a developing visual circuit. J.
(1998). Activity differentially regulates the sur- Neurosci. 27;8268-8277.
face expression of synaptic AMPA and NMDA Pribiag, H., and Stellwagen, D. (2014). Neuroimmune
glutamate receptors. Proc. Natl. Acad. Sci. USA regulation of homeostatic synaptic plasticity.
95, 70977102. Neuropharmacology 78, 1322.
Liu, J., and Buonomano, D.V. (2009). Embedding Queenan, B.N., Lee, K.J., and Pak, D.T. (2012).
multiple trajectories in simulated recurrent Wherefore art thou, homeo(stasis)? Functional
122 Part II: Homeostatic Control

diversity in homeostatic synaptic plasticity. GABA A-receptor expression in the surround of


Neural Plast., Article ID 718203. cerebral photothrombosis. J. Cereb. Blood Flow.
Qiu, Z., Sylwestrak, E.L., Lieberman, D.N., Zhang, Metab. 16, 906914.
Y., Liu X.-Y., and Ghosh, A. (2012). The Rett syn- Seeburg, D.P., Feliu-Mojer, M., Gaiottino, J., Pak,
drome protein MeCP2 regulates synaptic scal- D.T., and Sheng, M. (2008). Critical role of CDK5
ing. J. Neurosci. 32, 989994. and polo-like kinase 2 in homeostatic synaptic
Rabinowitch, I., and Segev, I. (2008). Two opposing plasticity during elevated activity. Neuron 58,
plasticity mechanisms pulling a single synapse. 571583.
Trends Neurosci. 31, 377383. Shepherd, J.D., Rumbaugh, G., Wu, J., Chowdhury,
Rannals, M.D., and Kapur, J. (2011). Homeostatic S., Plath, N., Kuhl, D., Huganir, R.L., and Worley,
strengthening of inhibitory synapses is mediated P.F. (2006). Arc/Arg3.1 mediates homeostatic
by the accumulation of GABA(A) receptors. J. synaptic scaling of AMPA receptors. Neuron 52,
Neurosci. 31, 1770117712. 475484.
Renart A., Song, P., and Wang, X.J. (2003). Robust Siddoway, B., Hou, H., and Xia, H. (2014). Molecular
spatial working memory through homeostatic mechanisms of homeostatic synaptic downscal-
synaptic scaling in heterogeneous cortical net- ing. Neuropharmacology 78, 3844.
works. Neuron 38, 473485. Sjstrm, P.J., Rancz, E.A., Roth, A., and Husser, M.
Rich, M.M. and Wenner, P. (2007). Sensing and (2008). Dendritic excitability and synaptic plas-
expressing homeostatic synaptic plasticity. ticity. Physiol. Rev. 88, 769840.
Trends Neurosci. 30, 119-125. Small, D.H. (2008). Network dysfunction in Alzheimers
Rossi Pool, R., and Mato, G. (2010). Hebbian plas- disease: Does synaptic scaling drive disease pro-
ticity and homeostasis in a model of hypercol- gression? Trends Mol. Med. 14, 103108.
umn of the visual cortex. Neural Comput. 22, Soden, M.E., and Chen, L. (2010). Fragile X protein
18371859. FMRP is required for homeostatic plasticity and
Routh, B.N., Johnston, D., and Brager, D.H. (2013). regulation of synaptic strength by retinoic acid. J.
Loss of functional A-type potassium channels in Neurosci. 30, 1691016921.
the dendrites of CA1 pyramidal neurons from a Song, S., Sjstrm, P.J., Reigl, M., Nelson, S.B., and
mouse model of Fragile X syndrome. J. Neurosci. Chklovskii, D (2005). Highly nonrandom fea-
33, 1944219450. tures of synaptic connectivity in local cortical
Rowan, M., and Neymotin, S. (2013). Synaptic scal- circuits. PLoS Biol. 3, e68.
ing balances learning in a spiking model of neo- Stellwagen, D., and Malenka, R.C. (2006). Synaptic
cortex. Lect. Notes Comput. Sci. 7824, 2029. scaling mediated by glial TNF-alpha. Nature
Rutherford, L.C., DeWan, A., Lauer, H.M., 440, 10541059.
Turrigiano, G.G. (1997). Brain-derived neuro- Sullivan, T.J., and de Sa, V.R. (2006). Homeostatic
trophic factor mediates the activity-dependent synaptic scaling in self-organizing maps. Neural
regulation of inhibition in neocortical cultures. Netw. 19, 734743.
J. Neurosci. 17, 45274535. Sun, Q., and Turrigiano, G. (2011). GPSD-95 and
Rutherford, L.C., Nelson, S.B., and Turrigiano, G.G. PSD-93 play critical but distinct roles in synaptic
(1998). BDNF has opposite effects on the quantal scaling up and down. J. Neurosci. 31, 68006808.
amplitude of pyramidal neuron and interneuron Sutton, M.A., Ito, H.T., Cressy, P., Kempf, C., Woo,
excitatory synapses. Neuron 21, 521530. J.C., and Schuman, E.M. (2006). Miniature neu-
Saliba, R.S., Michels, G., Jacob, T.C., Pangalos, M.N., rotransmission stabilizes synaptic function via
and Moss, S.J. (2007). Activity-dependent ubiq- tonic suppression of local dendritic protein syn-
uitination of GABA(A) receptors regulates their thesis. Cell 125, 785799.
accumulation at synaptic sites. J. Neurosci. 27, Sutton, M.A., and Schuman, E.M. (2009).
1334113351. Partitioning the synaptic landscape: Distinct
Sarti, F., Zhang, Z., Schroeder, J., and Chen, L. microdomains for spontaneous and spike-trig-
(2013). Rapid suppression of inhibitory synaptic gered neurotransmission. Sci. Signal 2, pe19.
transmission by retinoic acid. J. Neurosci. 33, Tetzlaff, C., Kolodziejski, C., Timme, M., Tsodyks, M.,
11440-11450. and Wrgtter, F. (2013). Synaptic scaling enables
Savin, C., Joshi, P., and Triesch, J. (2010). Independent dynamically distinct short- and long-term mem-
component analysis in spiking neurons. PLoS ory formation. PLoS Comput. Biol. 9, e1003307.
Comput. Biol. 6, e1000757. Tetzlaff,C.,Kolodziejski,C.,Timme,M.,andWrgtter,F.
Schiene, K., Bruehl, C., Zilles, K. Q, M., Hagemann, G., (2011). Synaptic scaling in combination with many
Kraemer, M., and Witte, O.W. (1996). generic plasticity mechanisms stabilizes circuit con-
Neuronal hyperexcitability and reduction of nectivity. Front. Comput. Neurosci.5, 47.
Homeostatic Synaptic Scaling at Central Synapses 123

Tetzlaff, C., Kolodziejski, C., Timme, M., and Walsh, D.M., and Selkoe, D.J. (2007). A oligo-
Wrgtter, F. (2012). Analysis of synaptic scal- mersa decade of discovery. J. Neurochem. 101,
ing in combination with Hebbian plasticity 11721184.
in several simple networks. Front. Comput. Wang, G., Gilbert, J., and Man, H.-Y. (2012). AMPA
Neurosci.6, 36. receptor trafficking in homeostatic synaptic
Thalhammer, A., and Cingolani, L.A. (2014). Cell plasticity: Functional molecules and signaling
adhesion and homeostatic synaptic plasticity. cascades. Neural Plast., Article ID 825364.
Neuropharmacology 78, 2330. Wang, X.J. (2001). Synaptic reverberation underlying
Thiagarajan, T. C., Lindskog, M., and Tsien, R. W. mnemonic persistent activity. Trends Neurosci.
(2005). Adaptation to synaptic inactivity in hip- 24, 455463.
pocampal neurons. Neuron 47, 725737. Watt, A.J., and Desai, N.S. (2010). Homeostatic plas-
Tong, L., Balazs, R., Thornton, P.L., and Cotman, ticity and STDP: Keeping a neurons cool in a
C.W. (2004). Beta-amyloid peptide at sublethal fluctuating world. Front. Synaptic Neurosci.2, 5.
concentrations downregulates brain-derived Watt, A.J., van Rossum, M.C., MacLeod, K.M.,
neurotrophic factor functions in cultured corti- Nelson, S.B., and Turrigiano, G.G. (2000). Activity
cal neurons. J. Neurosci. 24, 67996809. coregulates quantal AMPA and NMDA currents
Tononi, G., and Cirelli, C. (2014). Sleep and the price at neocortical synapses. Neuron 26, 659670.
of plasticity:From synaptic and cellular homeo- Wayman, G.A., Lee, Y.S., Tokumitsu, H., Silva,
stasis to memory consolidation and integration. A.J., and Soderling, T.R. (2008). Calmodulin-
Neuron 81, 1234. kinases: Modulators of neuronal development
Turrigiano, G.G. (1999). Homeostatic plasticity in neu- and plasticity. Neuron 59, 914931.
ronal networks:The more things change, the more Wenner, P. (2011). Mechanisms of GABAergic
they stay the same. Trends Neurosci. 22, 221227. homeostatic plasticity. Neural Plast., Article ID
Turrigiano, G.G. (2008). The self-tuning neu- 489470.
ron:Synaptic scaling of excitatory synapses. Cell Wenner, P. (2014). Homeostatic synaptic plasticity
135, 422435. in developing spinal networks driven by excit-
Turrigiano, G.G. (2011). Too many cooks? Intrinsic atory GABAergic currents. Neuropharmacology
and synaptic homeostatic mechanisms in corti- 78, 5562.
cal circuit refinement. Annu. Rev. Neurosci. 34, Whitt, J.L., Petrus, E., and Lee, H.-Y. (2014).
89103. Experience-dependent homeostatic synaptic
Turrigiano, G.G. (2012) Homeostatic plasticity:local plasticity in neocortex. Neuropharmacology
and global mechanisms for stabilizing neuronal 78, 4554.
function. Cold Spring Harb. Perspect. Biol. 4, Wierenga, C., Ibata, K., and Turrigiano, G.G. (2005).
a005736. Postsynaptic expression of homeostatic plas-
Turrigiano, G.G., Leslie, K.R., Desai, N.S., ticity at neocortical synapses. J. Neurosci. 25,
Rutherford, L.C., and Nelson, S.B. (1998). 28952905.
Activity-dependent scaling of quantal amplitude Wierenga, C.J., Walsh, M.F., and Turrigiano, G.G.
in neocortical neurons. Nature 391, 892896. (2006). Temporal regulation of the expression
Turrigiano, G.G., and Nelson, S.B. (2000). Hebb and locus of homeostatic plasticity. J. Neurophysiol.
homeostasis in neuronal plasticity. Curr. Opin. 96, 21272133.
Neurobiol. 10, 358364. Winship, I.R., and Murphy, T.H. (2008). In vivo
Van Rossum, M.C.W., Bi, G.Q., and Turrigiano, calcium imaging reveals functional rewiring
G.G. (2000). Stable Hebbian learning from spike of single somatosensory neurons after stroke. J.
timing-dependent plasticity. J. Neurosci. 20, Neurosci. 28, 65926606.
88128821. Yger, P., and Harris, K.D. (2013). The Convallis rule
Vitureira, N., and Goda, Y. (2013). The interplay for unsupervised learning in cortical networks.
between Hebbian and homeostatic synaptic plas- PLoS Comput. Biol. 9, e1003272.
ticity. J. Cell. Biol. 203, 175186. Zenke, F., Hennequin, G., and Gerstner, W. (2013).
Vitureira, N., Letellier, M., and Goda, Y. (2012). Synaptic plasticity in neural networks needs
Homeostatic synaptic plasticity:From single syn- homeostasis with a fast rate detector. PLoS
apses to neural circuits. Curr. Opin. Neurobiol. Comput. Biol. 9, e1003330.
22, 516521. Zhao, C., Dreosti, E., and Lagnado, L. (2011).
Waites, C.L., Craig, A.M., and Garner C.C. (2005). Homeostatic synaptic plasticity through changes
Mechanisms of vertebrate synaptogenesis. Annu. in presynaptic calcium influx. J. Neurosci. 31,
Rev. Neurosci. 28, 251274. 74927496.
8
Homeostatic Role ofHeterosynaptic Plasticity
N I C H O L A S M . B A N N O N , M A R I N A C H I S T I A K O VA ,
AND MA XIM VOLGUSHEV

INTRODUCTION:LEV ELS the induction, is well suited for this role. We con-
O F H O M E O S TA S I S clude that heterosynaptic plasticity with experi-
Homeostatic regulation is one of the basic princi- mentally observed properties can serve as a
ples of the functioning of living organisms. In the mechanism of homeostatic regulation of synaptic
brain, a great variety of homeostatic mechanisms weight changes induced by associative learning.
is employed to keep a wealth of biophysical pro-
cesses within their operating range (Davis and A S S O C I AT I V E H E B B I A N
Bezprozvanny, 2001). These mechanisms oper- PLASTICITY
ate at multiple nested levels, such as regulation Plasticity is a universal property of synapses,
of metabolic activity, transmembrane gradients, vital for fundamental operations of the nervous
and intracellular concentration of ions; homeo- system. Synaptic plasticity mediates the forma-
stasis of synaptic weights; the balance of excita- tion and refinement of connectivity patterns in
tion and inhibition; the firing rate of neurons; the nervous system during development and also
and other, more global processes at systems and mediates effective adaptive behavior in changing
network levels. environments throughout life. Hebbs rule (Hebb,
We propose that the most effective homeo- 1949)formulates the basic principle according to
static mechanisms are intrinsic to a specific level, which synaptic weights change:synaptic connec-
such that the sensor that measures the state tions that repeatedly or persistently take part in
of the variable, the error detector that deter- firing of the postsynaptic neuron increase their
mines its deviation from a given set point, and strength. The original rule, suggested only for
the effector that provides the negative feedback potentiation of synaptic connections, was later
should all be built within the same level and extended to include depression: synapses that
utilize similar mechanisms, thus operating on a consistently do not take part in firing the post-
similar timescale as the processes they are regu- synaptic neuron decrease their strength. In 1973,
lating. Consider input-specific (or homosynap- Bliss and Lomo discovered the cellular basis of
tic) plasticity governed by Hebbian rules that associative learning, long-term potentiation
mediates developmental plasticity, learning, and (LTP) in the hippocampal formation (Bliss and
memory. Associative synaptic plasticity intro- Lomo, 1973). The authors found that after a brief
duces a perturbation to the strength of neurons period of strong activity (high-frequency stimu-
inputs. This perturbation is amplified by a posi- lation, or afferent tetanization) the amplitude of
tive feedback on synaptic weight changes, which test responses to electric stimuli was increased,
is intrinsic to Hebbian-type learning rules, thus and the increase persisted for hours. Further
pushing the system out of balance. We argue that research complemented the associativity rule
to achieve a high-fidelity homeostatic response with the rules of cooperativity and input speci-
to a change in synaptic weights, the underlying ficity (Bliss and Collingridge, 1993). The rule of
mechanism should have the same-level trigger cooperativity states that activation should be
and operate on the same timescale as Hebbian strong, exceeding a certain threshold, to induce
plasticity. Heterosynaptic plasticity, which plastic changes. The rule of input specificity
accompanies induction of homosynaptic changes maintains that changes take place only at syn-
but occurs at synapses that were not active during apses that were activated during the induction
Homeostatic Role of Heterosynaptic Plasticity 125
but not at other synapses. Altogether, these calcium signal and the mechanisms of plasticity
Hebbian-type learning rules capture the asso- expression (Abbott and Nelson, 2000; Malenka
ciative nature of synaptic changes and explain a and Bear, 2004; Feldman, 2009; Manninen etal.,
multitude of plastic phenomena in the nervous 2010). Expression of plasticity may involve both
system, ranging from refinement of connectivity presynaptic changes, such as an increase of the
during development (neurons that fire together probability of transmitter release in LTP, and
wire together) to extraction of casual relations postsynaptic changes, such as insertion or inter-
between events in the environment in Pavlovian nalization of -amino-3-hydroxy-5-methyl-
conditioning and other types of associative learn- 4-isoxazolepropionic acid (AMPA) receptors.
ing, motor learning, and acquisition of sequences Associative synaptic changes, at least at their
of behavioral actions. early phase, are induced within tens of seconds
Spike-timing dependent plasticity (STDP) to minutes.
explicitly implements relative timing between
firing of the neuron and activity at its inputs (pre- H E B B I A N P L A S T I C I T Y: W H Y
synaptic spikes) as a determinant of the direction IT CANNOT WORKALONE
and the magnitude of synaptic weight changes. In Synaptic changes induced according to
the typical STDP learning rule, synaptic inputs Hebbian-type rules provide a cellular mechanism
that were active shortly before a postsynaptic for encoding new memories. However, learning
action potential (pre before post) are potentiated, systems in which Hebbian-type plasticity is the
while synaptic inputs active shortly after the only rule that governs synaptic modifications has
postsynaptic spike (post before pre) are depressed two major problems:they are prone to runaway
(Markram etal., 1997; Magee and Johnston, 1997; dynamics of synaptic weights and activity, and
Abbott and Nelson, 2000; Caporale and Dan, they introduce only a weak degree of competition
2008). STDP, and Hebbian-type learning rules in between synapses.
general, extract casual relations between events Propensity for runaway dynamics is imposed
by using the temporal order of their occurrence. by the positive feedback on synaptic weight
This dictates the necessity of activity of both changes that is intrinsic to Hebbian-type learning
presynaptic and postsynaptic neurons for plas- rules. Potentiation, by making synapses stronger,
ticity induction. The necessity for concomitant increases their chances to take part in firing a
pre- and postsynaptic activation for plasticity neuron and thus increases the probability of these
induction and the rule of input specificity explic- synapses to be potentiated further. Similarly,
itly restrict plasticity to synaptic inputs that were synapses weakened by depression have a lower
active during the induction. Thus plasticity gov- chance to lead to spikes and thus an increased
erned by Hebbian-type learning is input specific probability for being further depressed. Because
and homosynaptic. of this positive feedback on synaptic weight
Synaptic changes underlying Hebbian plas- changes, synapses would tend to be either poten-
ticity are triggered by rises of intracellular [Ca 2+], tiated to the maximal value or depressed to zero.
which under natural conditions result from fir- The runaway of synaptic weights leads to runa-
ing of pre- and postsynaptic neurons at high way dynamics of activity. Runaway potentiation
frequency during afferent tetanization, or with of synaptic weights to maximal values results in
repeatedly occurring coincidence with pre- overexcitability of neurons and excessive activity,
cise temporal relation in STDP (Golding et al., while runaway depression would lead to silencing
2002; Nevian and Sakmann, 2006; Sjstrm of neurons. Both scenarios would lead to distur-
and Hausser, 2006). Importantly, such activ- bance of the inputoutput relations of neurons
ity patterns are not uncommon and may occur (Chen et al., 2013), thus compromising compu-
naturally in the living brain. The final outcome tational abilities of neuronal networks (Skorheim
of the induction (whether or not long-term plas- et al., 2014). Moreover, runaway potentiation of
ticity will be induced, the direction of change, synaptic weights and associated runaway activity
and the magnitude of potentiation or depression) are energetically unsustainable and may lead to
depends on the temporal dynamics and ampli- pathology. Thus homosynaptic changes governed
tude of intracellular [Ca2+] rise, the source of by associative Hebbian plasticity must be con-
Ca2+ influx, and specifics of the synapse (Lisman, strained by additional plasticity rules that keep
1989; Artola and Singer, 1993; Cummings etal., plastic synapses and learning networks within
1996; Yang et al., 1999). Accordingly diverse their operation range, maintaining a balance of
are the biochemical cascades triggered by the learning and network homeostasis.
126 Part II: Homeostatic Control

Hebbian-type learning rules introduce only a strong synaptic competition in learning systems
weak, if any, degree of competition between syn- with Hebbian-type rules has been appreciated
apses. The idea of competition, which maintains since early theoretical studies (von der Malsburg,
that synapses should strengthen at the expense 1973; Miller and MacKay, 1994; Miller, 1996). To
of other synapses, is well grounded from a bio- some extent, this can be achieved by carefully
logical perspective: while undergoing changes, balancing STDP rules, whereby an appropriate
synapses might compete for shared limited negative bias balances potentiation and depres-
resources, such as available energy, molecules, sion (Song etal., 2000; Van Rossum etal., 2000;
or plasticity factors. The need for competition Gtig etal., 2003; Babadi and Abbott, 2010). One
is best illustrated by activity-dependent shap- problem with this solution is that it imposes strict
ing of sensory representations during develop- requirements on the relative strength (amplitude
ment (Wiesel and Hubel, 1963; Aitkin et al., and duration) of potentiation and depression
1970; Merzenich et al., 1975; Thompson et al., windows. STDP can indeed lead to stabilization
1983; Feldman, 2009). The canonical example of the neurons mean firing rate if the integral
here is the development of projections of specific of the learning windows for potentiation and
thalamic afferents from the lateral geniculate depression is negative (Kempter et al., 2001).
nucleus to the visual cortex from a highly over- This requirement is, however, not consistent with
lapping projection of afferents representing two experimental evidence that has demonstrated
eyes in early life to segregated projections into an enormous heterogeneity in the parameters
ocular dominance columns in adults. Synaptic of STDP windows at different synapses, cells,
changes during everyday learning might be more developmental stages, and conditions of neuro-
subtle, but the ability of synaptic competition modulation (Nishiyama et al., 2000; Sjstrm
to accentuate the plasticity-induced contrast etal., 2001; Zhou etal., 2005; Haas etal., 2006;
between synaptic weights might enhance learn- Feldman, 2009). Arelated problem is that STDP
ing, especially in paradigms that involve dis- rules adjusted to maintain stability in a neuron
crimination (Skorheim et al., 2014). Although subject to a certain pattern of external drive may
specific homosynaptic induction protocols can still lead to runaway dynamics when activity
induce either potentiation or depression, and changes, such as increase of the input correlation.
thus change synaptic weights in both directions, Finally, an STDP rule provides only weak com-
these bidirectional changes can hardly support petition between synapses: only synapses from
synaptic competition because they require repe- presynaptic neurons that are repeatedly active
tition of specific and independent patterns of the shortly after the postsynaptic spikes, and thus
input activity. For two groups of inputs to change repeatedly fall into the depression window, will
in opposite directions, both groups should be become depressed. This weak competition does
repeatedly activated with specific but different not mimic biologically-plausible forms, such as
and independent patterns, such as low frequency competition for limited resources.
for induction of long-term depression (LTD) but One further mechanism that was suggested to
high frequency for induction of LTP, or be sys- reduce the positive feedback on synaptic weight
tematically active at different timings relative changes and counteract their runaway dynam-
to postsynaptic activity to change via an STDP ics is weight dependence of plastic changes (Oja,
mechanism. Although possible in theory, such a 1982). Weight dependence dictates that weaker
scenario imposes strict requirements on the rela- synapses can potentiate more, while stronger
tions between specifics of plasticity rules and pat- synapses potentiate less, and in the limit do not
terns of input activity. Thus, to support intrinsic change (Bi and Poo, 1998; van Rossum et al.,
competition between synapses, a mechanism for 2000; Hardingham et al., 2007). In this limit,
plastic changes outside of the Hebbian learning weight dependence imposes bounds on synaptic
rule is required. weights, though it does not prevent saturation of
synaptic weights at extreme values. Combination
MODELING of weight dependence of plastic changes with
P E R S P E C T I V E : W H AT I S depression-biased STDP rules improves the
NEEDED INADDITION stability of the activity level of model neurons
TOHEBBIANRULES? with plastic synapses (van Rossum et al., 2000;
The need for maintaining stability of the total Kempter etal., 2001; Gtig etal., 2003; Morrison
synaptic weights, the amount of synaptic drive etal., 2007; Babadi and Abbot, 2010; Gilson and
a neuron receives and neuronal activity, and for Fukai,2011).
Homeostatic Role of Heterosynaptic Plasticity 127

Achieving Stability Through between plastic synapses, for example for limited
Normalization available resources, can also be one of the natural
A simple and robust method of stabilization com- ways in which the magnitude of possible change
monly used in modeling to constrain runaway is restricted, thus preventing excessive plastic-
dynamics of neuronal activity is normalization ity. In a pioneering computational study, von der
(von der Malsburg, 1973; Oja, 1982). The concept Malsburg (1973), citing the limited evidence that
of normalization is that following an episode of was available 40 years ago, suggested that the
plasticity inducing weight changes at some syn- biological constraint could be limited dendritic
apses, all synaptic weights on the cell are read- space for which synapses are competing. In a later
justed so that their sum remains constant. This study, Oja (1982) speculated that the physiological
normalization has typically been implemented via basis would be competition for shared resources,
multiplicative and subtractive methods. In multi- such as receptor molecules, surface area, energy
plicative normalization (von der Malsburg, 1973; resources, or plasticity factors.
Oja, 1982), each weight is multiplied by an amount In the simplest sense, competition is pro-
necessary to maintain the overall net weight (Van vided by normalization, with different meth-
Ooyen, 2001). In the subtractive method, a fixed ods of normalization introducing different
amount of change (decay to compensate for the degrees of competition (Miller and MacKay,
effect of homosynaptic potentiation or increase 1994; Miller, 1996; Gerstner and Kistler, 2002).
to compensate for the effect of depression) is Multiplicative normalization introduces a weak
added to all synapses regardless of their weight competition, which permits a gradated separa-
(Miller and MacKay, 1994). While both meth- tion of synaptic weights but cannot explain the
ods of normalization, multiplicative and sub- competitive exclusion of thalamic afferents in
tractive, robustly prevent runaway dynamics of ocular dominance plasticity, when neurons in
total synaptic drive and activity, neither prevents the visual cortex become predominately inner-
runaway potentiation or depression of individual vated by afferents carrying information from a
synapses. Moreover, multiplicative and subtrac- single eye (Miller and MacKay, 1994). Amodel
tive normalization introduce a different degree with multiplicative normalization was able to
of competition between synapses and thus may segregate correlated inputs from noncorrelated
lead to different final distributions of synaptic inputs; however, it failed to segregate inputs
weights and functional connectivity (Miller and expressing stronger correlation out of a larger
MacKay, 1994; Miller, 1996; see later discussion). pool of weaker-correlated inputs. This latter set-
The use of normalization for achieving stability ting is more realistic:inputs from different eyes
of the activity level and synaptic competition was express a certain degree of correlation imposed
further elaborated in later studies (e.g., Kempter by the visual scene, although these correlations
et al., 2001; Elliott and Shadbolt, 2002; Wu and are weaker than correlations between activity
Yamaguchi, 2006; Finelli etal.,2008). of inputs from the same eye. Subtractive nor-
Note that because normalization affects all malization introduces strong competition suffi-
synapses of the neuron, irrespective of their cient to solve this problem and segregate inputs
recent activity, it postulates the existence of het- from the two eyes despite their correlation, but
erosynaptic plasticitychanges at synapses that it leads to emergence of synapses with extreme
were not activated during plasticity induction. weights (Miller and MacKay, 1994; Elliott and
Shadbolt, 2002). From a biological perspective,
The Need forCompetition subtractive normalization is less plausible than
Theoretical analyses have demonstrated that multiplicative, though neither of the two has a
strong competition between projecting fibers, such clear relation to biological processes that may
that the weight of synapses formed by one fiber underlie competition in real neurons. It has
should grow at the expense of synapses of other been suggested that the explanatory power of
fibers which must decrease their weights, is neces- models is limited if normalization is imposed
sary for processes such as the establishment of sen- as a mathematical convenience and does not
sory representations during development (Wiesel represent dynamics underlying a physiological
and Hubel, 1963; Aitkin et al., 1970; Merzenich mechanism (Van Ooyen, 2001). Ideally, com-
et al., 1975; Thompson et al., 1983; Feldman, petition should be a consequence of the learn-
2009). In the mature brain, synaptic competition ing rule and not require explicit additional
is instrumental in learning tasks that involve dis- rules (Gerstner and Kistler, 2002). By extension
crimination (Skorheim etal., 2014). Competition it could also be proposed that normalization
128 Part II: Homeostatic Control

should be a consequence (or part of) the learn- The authors found that, to achieve robust stabil-
ing rule. Indeed, in a model that implemented ity of the system, the homeostatic mechanism
activity-dependent competition over a resource must operate on a timescale comparable to the
necessary for plasticity, neurotrophic factors, timescale of plasticity itself (Zenke etal.,2013).
the normalization emerged naturally, with- The question of the trigger for the homeo-
out being explicitly implemented (Elliott and static mechanism that protects synapses from
Shadbolt, 2002). In a stable substate of this bio- runaway in the face of Hebbian plasticity is more
logically inspired model, the emerged Hebbian complicated because possibilities do not form a
learning dynamics were accompanied by a mul- one-dimensional, easily quantifiable continuum
tiplicative normalization that, unlike in mod- that can be conveniently studied in simulations
els with imposed normalization, permitted the (such as time constant) but rather consist of sev-
segregation of correlated inputs. Thus modeling eral discrete factors, such as total synaptic weight
competition for resources, which approximates limited by resource and energy constraints, or
the biological reality more closely, can manifest level of activity-dependent calcium influx, or fir-
the normalization necessary to prevent runaway ing rate of a cell or a population. These possibili-
and provide the competition to segregate inputs, ties are not mutually exclusive but may operate on
moving these features from an ad hoc rectifica- several different levels of homeostatic regulation.
tion to emergent properties of learningrules. Thus a successful physiological mechanism
that maintains stability of the dynamics of learn-
The Trigger and theTimescale ing systems with plastic synapses must, in addi-
ofthe Homeostatic Mechanism tion to the aforementioned requirements, operate
In most modeling studies, normalization is in tandem with homosynaptic plastic changes.
implemented directly into the learning rules, In sum, theoretical analysis clearly shows
which update synaptic weights in each itera- the necessity of homeostatic mechanism(s)
tion. As a result, first, the trigger for plasticity that prevent runaway dynamics imposed by
also inevitably triggers the normalization, and Hebbian-type rules on synaptic weights and neu-
second, the normalization operates on the same ronal activity. It also identifies certain required
timescale as the plasticity rule. While these two features of cellular-level candidate mechanisms.
features are an automatic consequence of the First, such mechanisms should be robust to both
model design, they are suggestive of additional the variability in the details of plasticity rules
characteristics required from a homeostatic observed in experiments and a wide range of
mechanism in systems with Hebbian-type activity patterns. Second, they should provide
learning rules. Indeed, a few studies in which normalization of synaptic weights and support
normalization was implemented separately synaptic competition. It is not clear if the nor-
converge at the conclusion that its time con- malization brings about competition or arises
stant should be short. In a model expressing from competition, or if normalization and com-
stable dynamics with STDP learning, the sepa- petition are mediated by diverse mechanisms.
rate timescale for homeostatic scaling regulated Third, they should operate on a timescale com-
by postsynaptic firing rate was relatively short, parable to the timescale on which Hebbian-type
with a time constant of 100s, which is compara- synaptic changes take place. This latter point
ble to the timescale of synaptic changes intro- would be automatically fulfilled if homeostatic
duced by learning (van Rossum et al., 2000). and Hebbian-type changes of synaptic weights
In a theoretical analysis of one-trial sequence share the trigger and are mediated by the same or
learning of place-fields in the hippocampus, overlapping intracellular processes.
Wu and Yamaguchi (2006) concluded that for
learning processes that occur within minutes, BIOLOGICAL
the physiological mechanism that constrains C A N D I D AT E : H O M E O S TAT I C
synaptic weights must also operate rapidly. The SY NAPTIC SCALING
question of timescale of homeostatic mecha-
nism that is necessary for maintaining stabil- Experimental Phenomena
ity of a system with plastic synapses has been A form of homeostatic plasticity that is often
directly addressed in a recent study by Zenke considered as a mechanism that keeps overall
et al. (2013). The time constant of the homeo- balance of synaptic weights and prevents runa-
static mechanism in this study was separate way dynamics of activity induced by Hebbian
from the time constant of the plasticity rule. learning rules is the phenomenon of synaptic
Homeostatic Role of Heterosynaptic Plasticity 129

scaling:compensatory scaling of synaptic weights 2002; Branco etal., 2008; Rabinowitch and Segev,
triggered by prolonged dramatic changes of 2008). Realistically, scaling is not a unitary phe-
global activity. Scaling modifies synaptic weights nomenon but a multitude of mechanisms aimed
to counteract changes of activity. Synaptic to counteract dramatic, long-lasting alterations
weights scale up after hours and days of activ- of overall neural activity and drive the firing rate
ity blockade by tetrodotoxin (Turrigiano et al., back to a set point (Rich and Wenner 2007; Watt
1998), or hyperpolarization of neurons caused and Desai 2010; Turrigiano, 2012). A common
by expression of inwardly rectifying potassium feature of these mechanisms is that their activa-
channels (Burrone etal., 2002), and scale down tion requires prolonged (hours/days) deviation of
after prolonged increases of activity by block- the triggering variable from its setpoint.
ade of inhibition (Turrigiano et al., 1998). This Homeostatic synaptic scaling, operating
scaling is multiplicative and thus maintains the alongside other plasticity mechanisms, might
relative balance of synaptic weights. Originally play a role in the dramatic reorganizations of
discovered in dissociated neuron cultures, scal- connectivity observed during visual cortex devel-
ing has been also demonstrated in the whole brain opment, such as ocular dominance plasticity and
after hours of deafferentation (Keck etal., 2013; experimental paradigms of monocular depri-
Vlachos etal., 2013; Becker etal., 2013). Scaling vation (Desai et al., 2002; Vitureira et al., 2012;
in the neocortex is developmentally regulated, Lambo and Turrigiano, 2013; Keck etal., 2013).
such that layer-4 neurons express a transient However, while scaling is well suited to adjust
ability to scale, and subsequently neurons from long-term and dramatic alterations of activ-
layers 2 and 3 demonstrate the phenomena that ity, and probably supplements and shapes other
persist through adulthood (Turrigiano,2012). homeostatic mechanisms during development,
Homeostatic synaptic scaling in cortical two features make it a poor candidate for serving
neurons is dependent on somatic calcium (Ca 2+) the acute constraining role necessary to combat
influx, gene transcription, and multiple intracel- runaway dynamics imposed by Hebbian-type
lular pathways that may operate in parallel or plasticity rules:the timescale and the trigger for
interact with each other (Rich and Wenner 2007; induction.
Ibata etal., 2008; Watt and Desai 2010; Turrigiano,
2012; Chen et al., 2014). Network-wide factors Timescale, Competition, and
such as activity-dependent brain-derived neu- Runaway Dynamics
rotrophic factor release or tumor necrosis fac- Homeostatic synaptic scaling operates on the
tor alpha from glia may also have a role in some timescale of hours and days. The rapid scal-
types of synaptic scaling (Kaneko et al., 2008; ing takes place after 4 hours of complete silenc-
Turrigiano, 2012). Evidence for both presynaptic ing of cultured neurons with tetrodotoxin (Ibata
and postsynaptic changes has been found in vari- et al., 2008). This timescale is compatible with
ous experimental paradigms (Turrigiano et al., that of developmental processes, such as for-
1998; Murthy et al., 2001; Burrone et al., 2002; mation of sensory representations in norm and
Ibata etal., 2008). However, typically, scaling is pathology (Wiesel and Hubel, 1963; Aitkin
attributed to postsynaptic mechanisms:changes et al., 1970; Merzenich et al., 1975; Thompson
in AMPA trafficking, resulting in changes of etal., 1983; Feldman, 2009), but it is at least two
the amplitude but not frequency of miniature orders of magnitude slower than the timescale
events, and changes of the amplitude but not of associative plasticity, which changes synap-
short-term plasticity of evoked responses (Ibata tic weights within minutes or even tens of sec-
et al., 2008; Turrigiano, 2012; Tatavarty et al., onds. Because of this dramatic difference of the
2013). Significantly, the biochemical cascades timescales, homeostatic synaptic scaling cannot
and mechanisms underlying AMPA traffick- mediate synaptic competition or normalize syn-
ing engaged by scaling are distinct from those aptic weights during ongoing associative synap-
engaged during Hebbian plasticity (either LTD tic plasticity, nor is it suitable for counteracting
or LTP; Turrigiano, 2012; Tatavarty etal.,2013). runaway dynamics that can be induced within
Although originally described as a global, seconds and minutes by Hebbian-type learning
cell-wide process, later studies raised the possibil- rules. This inconsistency between timescales
ity that scaling could be quasi-local (at the spatial of slow homeostatic scaling and fast associa-
resolution of dendritic branches), which could tive learning has been pointed out by Wu and
serve for localized normalization of weights in Yamaguchi (2006), who concluded that synaptic
a computationally useful way (Burrone et al., scaling does not seem to work for fast learning.
130 Part II: Homeostatic Control

Computational analysis of simple recurrent did see evidence for homeostatic scaling after
schemes built of neurons with Hebbian-type the lesions but noted that homeostatic scaling
synapses showed that the maximal amount of alone could not explain the observed recovery of
input that these simple circuits can tolerate with- activity in the deprived cortex (Keck etal., 2013).
out drifting into runaway dynamics decreases Note that even this more modest interven-
sharply with the increasing ratio of the time- tion represents extreme pathology. In contrast,
scales of the homeostatic and Hebbian plasticity. Hebbian-type synaptic plasticity can be induced
For the ratio of 100, which is just approaching by far more subtle events, and activity changes
the lower limit of experimentally measured val- that may result from associative synaptic plastic-
ues, the level of tolerated activity is close to zero ity might be also far less dramatic.
(Tetzlaff etal., 2012). The range of the timescales In sum, homeostatic synaptic scaling repre-
of a homeostatic mechanism compatible with sents a homeostatic mechanism engaged slowly
stable operation of a neuron with Hebbian-type in response to extreme and long-lasting chal-
plasticity has been systematically investigated lenges, and presumably these mechanisms would
in a recent theoretical study (Zenke etal., 2013). not be engaged until runaway dynamics had
The results demonstrated that, for achieving created a dramatically unstable and saturated
robust stability of a system with Hebbian-type network. Experimentally observed properties
plastic synapses, the mechanism that maintains of homeostatic synaptic scaling do not fit the
homeostasis and prevents runaway dynamics requirements delineated in theoretical studies
must operate on a timescale comparable to the for a hypothetical mechanism needed in addi-
plasticity itself (Zenke etal.,2013). tion to Hebbian-type learning rules. Both the
This conclusion is compatible with results trigger and the timescale of synaptic scaling
of other computational studies of which we are fundamentally different from those of the
are aware. To the best of our knowledge, in all Hebbian-type plasticity.
computer models in which synaptic scaling was
used to prevent runaway dynamics imposed by B I O L O G I C A L C A N D I D AT E :
Hebbian-type plasticity or to introduce synap- HETEROSY NAPTIC
tic competition, scaling was implemented on a PLASTICITY
short timescale, the same or comparable to the
timescale of the Hebbian plasticity. We are not Experimental Evidence
aware of a published computational study in Heterosynaptic plasticity refers to changes at syn-
which homeostatic synaptic scaling with experi- apses that were not presynaptically active dur-
mentally observed features (in particular the ing the induction of plasticity. Heterosynaptic
requirement of at least 4 hours of altered activity LTD accompanying induction of LTP was first
level to trigger synaptic changes) was shown to described in the hippocampus shortly after the
be effective in preventing runaway dynamics or phenomenon of LTP was discovered (Lynch etal.,
supporting synaptic competition during ongoing 1977). In CA1 pyramidal neurons, induction of
learning. LTP of Schaffer collateral-commissural synapses
at apical dendrites was accompanied by an LTD
Realism ofExperimental Paradigm at inputs to basal dendrites made by commissural
One further concern regarding possible involve- fibers that were not stimulated during the induc-
ment of homeostatic synaptic scaling in balanc- tion. Vice versa, induction of LTP at the basal
ing synaptic changes induced by Hebbian-type dendrites was accompanied by LTD at the apical
plasticity is the severity of changes that are dendrites. Heterosynaptic LTD accompanying
required to trigger the scaling. Typically, scaling the induction of homosynaptic LTP clearly has
up is induced by a complete silencing of activ- potential for both balancing plastic changes and
ity for many hours by tetrodotoxin application supporting synaptic competition.
(Turrigiano etal., 1998; Turrigiano, 2012). Such Heterosynaptic LTP was first discovered at
dramatic and global changes of activity are nei- synapses adjacent to potentiated inputs. After
ther likely nor compatible with normal operation pairing one input to a CA1 neuron, synapses
of the brain. A recent study demonstrated that formed by nearby fibers on that cell and even on
6 hours after complete bilateral retinal lesions, nearby neurons were potentiated too (Bonhoeffer
activity in the visual cortex was reduced to ~60% et al., 1989; Kossel et al., 1990; Schuman and
of prelesion level (Keck etal., 2013). The authors Madison, 1994a; Engert and Bonhoeffer, 1997).
Homeostatic Role of Heterosynaptic Plasticity 131

These results demonstrated that input specificity (Lee etal., 2012). Mechanisms of heterosynaptic
breaks down at short distances: LTP-protocols plasticity overlap with those mediating homo-
induce plasticity not only at the activated syn- synaptic plasticity, as indicated by a partial
apses but also at those not active during the occlusion between homo- and heterosynaptic
induction. plastic changes (Kuhnt etal., 1994; Cummings
Studies of the spatial distribution of plastic etal., 1996; Neveu and Zucker, 1996; Yang etal.,
changes in structures with regular organization 1999; Volgushev etal., 1999). This partial occlu-
of the inputs, such as the hippocampus or amyg- sion of expression mechanisms of homosynap-
dala, revealed a Mexican hattype profile of plas- tic and heterosynaptic plasticity may be due to
ticity (White etal., 1990; Royer and Par, 2003). the shared calcium dependence and activation
Induction of LTP at a set of synapses was accom- of common intracellular cascades by the Ca 2+
panied by a biphasic profile of heterosynaptic rises that lead to plastic changes. One further
changes: a weaker LTP at nearby inputs, LTD possibility involves shared pathways of retro-
at more distant inputs, and finally no changes grade signaling. Specifically the role of nitric
at yet more distantly located synapses. A sym- oxide and cannabinoid signaling pathways have
metrical profile was observed around the site of been demonstrated in both homosynaptic and
LTD induction: weaker LTD at close distances heterosynaptic plasticity. Nitric oxide has an
and LTP at more distant inputs (Royer and Par, established role as a retrograde signal during
2003). Importantly, the amount of potentiation plasticity induced by pairing or afferent teta-
and depression in these profiles was balanced, nization (Gally etal., 1990; Bhme etal., 1991;
so that neither LTP nor LTD induction at acti- ODell etal., 1991; Schuman and Madison 1991).
vated synapses resulted in net changes of the total Because nitric oxide spread is not limited to
synaptic input. Induction of balanced profiles the activated synapses, it also can affect non-
of plastic changes can provide a powerful local activated synapses. Indeed, retrograde signal-
mechanism of both normalization of synaptic ing via nitric oxidedependent pathways has
weights and synaptic competition. been explicitly demonstrated to be involved in
induction of heterosynaptic changes (Schuman
Trigger forHeterosynaptic and Madison 1994a,1994b; Volgushev et al.,
Plasticity:Bursts ofSpikes 2000; Nugent et al., 2007; Bright and Brickley,
and CalciumRise 2008; Phillips et al., 2008; Lange et al., 2012;
Heterosynaptic changes are triggered by rises Lee etal., 2012; reviewed in Hardingham etal.,
of intracellular Ca 2+ concentration, thus shar- 2013). LTD mediated by retrograde signaling
ing the trigger with Hebbian-type plastic- via endocannabinoids has been demonstrated
ity (Lisman, 1989; Artola and Singer, 1993; at both excitatory and inhibitory connections at
Cummings etal., 1996; Yang etal., 1999). Bursts both homo- and heterosynaptic sites (Sjstrm
of backpropagating action potentials induce et al., 2004; Chevaleyre et al., 2003, 2006; Pan
massive rises of [Ca 2+] throughout the dendritic etal., 2008; Chiu etal., 2010; Huang etal., 2008;
tree, which might be sufficient to reach plastic- Yasuda et al., 2008; reviewed in Heifets and
ity threshold at heterosynaptic sites (Spruston Castillo, 2009). Because activation of mGluRs
etal., 1995; Staubli and Ji, 1996; Larkum etal., can trigger the production and release of endo-
1999; Golding et al., 2002; Waters et al., 2003; cannabinoids in a calcium-independent manner
Lisman and Spruston, 2005; Sjstrm and (Maejima et al., 2001), this form of heterosyn-
Hausser, 2006; Remy and Spruston, 2007). aptic LTD can be Ca 2 independent but mGluR
Experimental protocols that deliver purely dependent (Chevaleyre etal., 2003; Chevaleyre
postsynaptic challenges to postsynaptic neu- etal., 2006; Pan etal., 2008; Huang etal., 2008;
rons can induce long-term plasticity. In the hip- Yasuda etal., 2008; Chiu etal.,2010).
pocampus and neocortex, either photolysis of Thus heterosynaptic plasticity can be induced
caged Ca 2+ (Neveu and Zucker, 1996; Yang etal., by the same protocols, occurs at the same time-
1999) or postsynaptic spiking is sufficient to scale, and shares mechanisms with Hebbian-type
induce plasticity (Kuhnt etal., 1994; Volgushev plasticity (for further discussion of induction of
et al., 1994; Volgushev et al., 2000; Cummings heterosynaptic changes by the protocols that
et al., 1996, Chistiakova and Volgushev, 2009; are typically used to induce homosynaptic plas-
Lee et al., 2012). Chelating intracellular Ca 2+ ticity, see Fig. 8 and related text in Chistiakova
impairs induction of heterosynaptic plasticity etal.,2014).
132 Part II: Homeostatic Control

Properties ofHeterosynaptic active during the induction, will experience post-


Plasticity synaptic activity that is not associated with acti-
Heterosynaptic, long-term plastic changes can be vation of their presynaptic fibers. Because spikes
induced in hippocampal and neocortical neurons can be evoked by activation of just a small frac-
by intracellular tetanizationbursts of spikes tion of all synapses, this situationpostsynaptic
evoked by short, depolarizing pulses applied activity without presynaptic activationwill
through the recording electrode (Figure 8.1A; be experienced by the majority of synapses at a
Kuhnt et al., 1994; Volgushev et al., 1994, 1997; neuron. Thus intracellular tetanization mimics
Volgushev et al., 1999; Volgushev et al., 2000; the conditions experienced by the majority of
Chistiakova and Volgushev, 2009; Lee et al., neurons synapses during the induction of asso-
2012). The rationale behind the intracellular teta- ciative plasticity. Moreover, because during the
nization protocol as a tool to study heterosyn- intracellular tetanization no synaptic inputs were
aptic plasticity is as follows:Each neuron in the stimulated, any synaptic changes can be consid-
neocortex receives thousands of synaptic inputs. ered heterosynaptic.
However, spiking of a neuron can be induced by How much postsynaptic activity is required
activation of only a fraction of these inputs, from to induce heterosynaptic plasticity? Our typi-
a few dozens to hundreds. Repetitive activation cal protocol of intracellular tetanization con-
of a specific group of synapses leading to repeti- sists of 10 bursts of three to seven spikes at 50
tive firing of a cell can, under certain conditions, to 100 Hz, presented as one train or repeated
induce synaptic plasticity. Other synapses, not three times with 1-minute intervals (Kuhnt etal.,

(a) (b) 4 (c)


Intracellular 3
Tetanization
EPSP amplitude, mV

2 1 mV
r = 0.52
? 1 300
***
? 0
EPSP amplitude change, %

?
2 200
?
1.5 1 mV
? 1 100
0.1 s
? 0.5
EPSP amplitude, mV

0
1.2

0.8 1 mV
100
40 mV 0 1 2 3
50 ms 0.4
Initial Paired-Pulse Ratio
0
x3 10 0 10 20 30 40 50
1s Time from Intracellular Tetanization, min

FIGURE 8.1: Long-term synaptic plasticity induced by intracellular tetanization. A:Scheme of an


intracellular tetanization experiment. Bursts of short depolarizing pulses (5 pulses at 100Hz; 10 bursts at 1Hz, 3
trains of 10 bursts) were applied through the recording electrode without presynaptic stimulation to induce bursts
of action potentials. Synaptic responses were recorded before and after the intracellular tetanization. Because
no inputs were stimulated during the induction, plasticity at all synapses can be considered heterosynaptic.
B:Examples of inputs that underwent potentiation (top), depression (middle), or did not change after intracellular
tetanization. Time courses of amplitudes of excitatory postsynaptic potentials (EPSPs) evoked by the first pulse in
a paired-pulse paradigm. The timing of intracellular tetanization is indicated by the arrows above each plot. Insets
show averaged responses to paired pulse stimuli before and after intracellular tetanization, from color-coded time
intervals. In this example, LTP and LTD were induced simultaneously at two inputs to the same neuron. Input
resistance of neurons measured by responses to small hyperpolarizing pulses applied before synaptic stimuli (not
shown) remained unchanged. C:Correlation between changes of EPSP amplitude after intracellular tetanization
and initial paired-pulse ratio. Data for N=136 inputs to pyramidal neurons in slices of visual cortex (N=60
inputs) and auditory cortex (N=76 inputs). Green symbols (star, square, and triangle) refer to the example inputs
fromB.
Modified, with permission, from Chen etal.,2013.
Homeostatic Role of Heterosynaptic Plasticity 133

1994; Volgushev et al., 1994, 1997; Volgushev initial paired-pulse ratio, and thus had low release
et al., 1999; Volgushev et al., 2000; Chistiakova probability, were typically potentiated. Inputs
and Volgushev, 2009; Lee etal., 2012; Chen etal., that expressed low initial paired-pulse ratio, and
2013). Altogether, 50 to 200 action potentials are thus had high release probability, were typically
generated during the intracellular tetanization. depressed or did not change. Thus heterosynaptic
This is well within the range of spiking in vivo, plasticity is weight dependent, and the direction
for example, during optimal stimulation of cat of weight change at each synapse is determined
visual cortex neurons (e.g., Volgushev etal., 2002, individually, depending on its initial properties.
2003) or even during slow oscillations in some Weight dependence is one further similar fea-
neurons (e.g., Volgushev etal., 2006). Moreover, ture of heterosynaptic and homosynaptic plas-
it is also within the range of postsynaptic activity ticity: it has also been reported for tetanization
during typical pairing protocols used for plastic- or pairing-induced LTP in the hippocampus and
ity induction. For example, the number of action neocortex (van Rossum et al., 2000; Sjstrm
potentials varied between 50 and 600 in pairing etal., 2001; Hardingham etal.,2007).
protocols used in seven representative publica- It has been suggested that the weight
tions (Markram etal., 1997; Hardingham etal., dependence of heterosynaptic plasticity reflects
2007; Ismailov etal., 2004; Froemke etal., 2005; history-dependent predispositions of synaptic
Zhou et al., 2005; Nevian and Sakmann 2006; inputs to undergo potentiation or depression
Sjstrm and Hausser 2006; Sjstrm et al., (Volgushev et al., 1997; Volgushev et al., 2000;
2008; see Chistiakova and Volgushev 2009, Fig.3 Chistiakova and Volgushev, 2009, Chistiakova
and Fig. 4 and accompanying text for details). et al., 2014). In this scenario, synapses that
This latter point raises an intriguing question of become weaker after a depression (and have lower
whether heterosynaptic plasticity can be induced release probability) have a lower predisposition
by regular pairing protocols used, for exam- for further depression but a stronger predispo-
ple, to induce STDP. Our recent analysis of the sition for potentiation. Synapses that became
published data (Feldman 2009; Sjstrm et al., stronger and have higher release probability
2001; Watt etal., 2004; Birtoli and Ulrich 2004; after a potentiation will have a higher predis-
Nevian and Sakmann 2006; Letzkus etal., 2006; position for depression. The idea of predisposi-
Hardingham et al., 2007) indicates that this is tions for plastic changes is closely related to the
indeed the case. The variance of response ampli- notion of a sliding threshold between depression
tude changes at unpaired inputs (i.e., synaptic and potentiation in the Bienenstock, Cooper,
inputs that were not stimulated during the pairing and Munro (BCM) rule (Bienenstock etal., 1982;
procedure and thus experienced only postsynap- Yeung etal., 2004), and to a more general con-
tic activity) is high, suggesting that although on cept of metaplasticity, according to which the
average the amplitude at unpaired inputs did not abilities of synapses to undergo potentiation or
change, both potentiation and depression must depression depend, among other factors, on the
have been observed in individual cases. However, recent history of their plastic changes (Abraham
because potentiation and depression at unpaired and Bear,1996).
inputs were roughly balanced, averaged changes In sum, heterosynaptic plasticity induced by
over the whole population were not significant intracellular tetanization expresses properties
(see Chistiakova et al., 2014, Fig. 8 and related that are well suited for serving as a robust mecha-
text for details). nism of normalization of synaptic weights:(a)it
Following intracellular tetanization in depresses strong and potentiates weak synapses,
pyramidal neurons from slices of rat visual thus preventing runaway dynamics of synaptic
cortex, amplitudes of synaptic responses could weights; (b) it is induced at nonactive synapses
increase, decrease, or remain unchanged by the same protocols that induce homosynaptic
(Figure 8.1B). In two independent inputs onto plasticity, providing for explicit competition; and
one cell, intracellular tetanization could simul- (c)it operates on the same timescale as homosyn-
taneously induce LTP in one input and LTD in aptic plasticity.
the other (Figure 8.1B). The direction of plastic
changes was correlated with the inputs initial Modeling:Heterosynaptic Plasticity
paired-pulse ratio, which is inversely related to Robustly Prevents Runaway
the release probability (Figure 8.1C; Volgushev Dynamics
etal., 1997; Volgushev etal., 2000; Lee etal., 2012; The hypothesis that heterosynaptic plasticity with
Chen et al., 2013). Inputs that expressed high experimentally observed properties can prevent
134 Part II: Homeostatic Control

runaway dynamics of synaptic weights and activ- how changing STDP parameters affects stability
ity has been tested in computer simulations of operation of the model neuron, the depression
(Chen et al., 2013). The model neuron received window of STDP was fixed, while the amplitude
synaptic inputs from 100 simulated presynaptic and the time constant of the potentiation window
neurons, each firing action potentials at an aver- was systematically varied. The range of tested
age frequency of 1 Hz, with Poisson distributed STDP rules included those strongly dominated
interspike intervals (Figure 8.2A). Activity of by depression, as well as those dominated by
presynaptic neurons was mildly correlated, with potentiation (see insets in Figure8.3).
averaged cross-correlation between pairs of spike The STDP-only model expressed nonsaturat-
trains 0.35 + 0.05. This input led to the firing ing behavior only with a very limited set of STDP
of the postsynaptic model neuron at ~1.8 Hz. In rules, specifically those dominated by depression
the example illustrated in Figure 8.2B, a STDP (Figure 8.3A, blue area of the matrix). As soon
rule with symmetrical windows for potentiation as the potentiation window of the STDP rule
and depression was implemented (Figure 8.2B, was ~75% of the depression window or stronger,
inset). In this model with an STDP-only learning synaptic weights and postsynaptic firing invari-
rule, synaptic weights showed runaway dynam- ably expressed runaway dynamics (Figure8.3A,
ics. By the end of the 100 s long simulation, all orange/red area of the matrix). Addition of het-
synaptic weights were saturated at the maximal erosynaptic plasticity to the model robustly pre-
value (Figure 8.2B, red). This led to a dramatic vented runaway dynamics and led to the stable
increase of the postsynaptic firing rate from ~1.8 distribution of synaptic weights within the oper-
Hz during the first 10 s of simulation to ~6.3 Hz ation range. The model equipped with heterosyn-
during the last 10 s of simulation. In the model aptic plasticity expressed stable behavior with all
with STDP windows strongly biased toward STDP windows testedfrom almost exclusively
depression (Figure8.2C, inset), synaptic weights depressing STDP rules to those strongly domi-
also expressed clear runaway dynamics, but nated by potentiation (Figure 8.3B). Notably,
toward the minimum value (Figure 8.2C, red). heterosynaptic plasticity, by preventing runaway
In this case, the postsynaptic neuron became dynamics of synaptic weight, also kept averaged
silent despite continuing activity of presynaptic firing rate around the operating point (Chen
neurons. etal.,2013).
Implementing in the model (in addition to Thus heterosynaptic plasticity makes a broad
STDP) heterosynaptic plasticity with experi- range of STDP parameters compatible with sta-
mentally observed properties effectively pre- ble operation of neurons and neuronal networks.
vented runaway dynamics of synaptic weights This is an important feature because experimen-
and activity. In the model with symmetrical tal evidence indeed shows wide variations of
STDP and heterosynaptic plasticity, synaptic STDP windows for potentiation and depression
weights slightly increased, but did not saturate, and of their relative strength in neurons and syn-
and formed a normal distribution around the aptic connections of different types (Abbott and
new mean value, within the operation range of Nelson, 2000; Nishiyama et al., 2000; Sjstrm
synapses (Figure 8.2D, red). Firing rate of the et al., 2001; Froemke et al., 2005; Zhou et al.,
postsynaptic neuron slightly increased from 2005; Haas etal., 2006; Caporale and Dan 2008;
~1.8 Hz to ~2.6 Hz. Heterosynaptic plasticity Feldman2009).
also prevented runaway dynamics of synaptic
weights and activity toward zero produced by Modeling:Heterosynaptic Plasticity
STDP windows with a negative bias (Figure8.2E, Permits Competition
red). These results demonstrate that heterosyn- Despite its strong stabilizing effect on synaptic
aptic plasticity can prevent runaway of synaptic weights, heterosynaptic plasticity does not pre-
weights to either extreme. vent synaptic competition and segregation of syn-
Further simulations demonstrated that the aptic weights. This has been tested in simulations
stabilizing effect of heterosynaptic plasticity on in which inputs to the model neuron were segre-
synaptic weights is long-lasting and robust to gated in two groups. The two groups of synapses
variations in the patterns of presynaptic activ- differed either by the correlation of presynaptic
ity, changes of calcium threshold for plasticity firing (high in one group, low in the other) or by
induction, and changes of parameters of STDP the average frequency of presynaptic firing. In
learning rules over a broad range of values. This the STDP-only model, inputs that were strongly
latter point is illustrated in Figure8.3. To explore correlated were rapidly potentiated and saturated
Homeostatic Role of Heterosynaptic Plasticity 135

(a) (b) (c)

(d) (e)

FIGURE 8.2: Heterosynaptic plasticity prevents STDP-induced runaway dynamics in a model.


A: Amodel neuron receiving synaptic inputs from 100 simulated presynaptic neurons. Presynaptic neurons fired
action potentials at an average frequency of 1Hz; their activity was mildly correlated (averaged cross-correlation
0.35 +0.05). In addition to synaptic activation, current could be injected into the postsynaptic neuron to evoke
spikes. BE: Distributions of synaptic weights at the beginning (initial synaptic weights, blue) and at the end of
simulation (final synaptic weights, red) in four neuron models. In models with only the STDP rule implemented
at each synapse, activity leads to runaway dynamics of synaptic weights (B, C). Implementing heterosynaptic
plasticity in addition to STDP rule prevents runaway dynamics of synaptic weights (D,E). B: In the model with
symmetrical windows for potentiation and depression (inset), synaptic weights express runaway potentiation,
and by the end of simulation all weights are saturated at the maximal value (0.03 mS/cm 2; note truncation of the
Y axis for the final weights). C: In the model with strong negative bias of STDP (inset), synaptic weights express
runaway depression leading to saturation of the weights of ~40percent of synapses at zero and silencing of the
postsynaptic neuron. D, E: The same models as in B and C are shown, but with the mechanism for heterosynaptic
plasticity implemented in addition to STDP. Note that synaptic weights do not express runaway dynamics and are
not saturated but remain normally distributed within the operationrange.
(Modified, with permission, from Chen etal.,2013).

at the maximum value, while distribution of the Thus heterosynaptic plasticity does not pre-
weights of weakly correlated inputs changed lit- vent segregation of groups of synaptic inputs
tle. In the model with both STDP and heterosyn- exhibiting activity patterns that differ by the
aptic plasticity, no inputs were saturated, but the degree of correlation or by firing frequency.
groups of weakly and strongly correlated inputs Moreover, it helps to preserve the ability of a
formed two clearly separate distributions, both neuron with plastic synapses for further learn-
within the operation range of synaptic weights ing: unsaturated synapses have higher potential
(Chen et al., 2013). Similarly, segregation was for further changes than those potentiated to the
observed when the two groups of synaptic inputs maximum or depressed to zero by STDP-only
differed by their average firing frequency rather learningrules.
than by the level of correlation. Note that com- In sum, heterosynaptic plasticity with experi-
petition imposed by heterosynaptic plasticity is mentally observed properties is a strong can-
able to support subtle differentiation of synaptic didate mechanism for counteracting runaway
weight changes. For example, if an initially large dynamics imposed on synaptic weights and
number of synapses are potentiated but later only activity by positive feedback of Hebbian-type
a portion of them remains consistently active, learning rules. Prevention of runaway dynam-
heterosynaptic plasticity will be able to suppress ics by heterosynaptic plasticity is robust over
the remaining synapses, thus allowing for selec- a broad range of activity patterns and details
tion of only the relevant groupa process that of Hebbian-plasticity rules, such as balance of
may mediate the differentiation stage of learning. STDP windows for potentiation and depression.
136 Part II: Homeostatic Control

(a) (b) Nonnormal


STDP only STDP + Heterosynaptic plasticity
Time constant + (ms) distribution
40 40 log(K2)
30 30 3
20 20 2
1
10 10
0
5 5
Normal
0.0002 0.001 0.0018 0.0025 0.0002 0.001 0.0018 0.0025
distribution
Potentiation window a+ amplitude, mS/cm2 Potentiation window a+ amplitude, mS/cm2

FIGURE 8.3: Heterosynaptic plasticity makes a broad range of STDP parameters compatible
with stable operation of neurons. A, B:Each box in the grids shows the DAgostino-Pearsons K 2 test for
normality of synaptic weight distribution after 100 seconds of simulations with different STDP potentiation win-
dows, with a+ and + as indicated on the X and Y axes. Insets show STDP windows for example boxes. The white
square indicates the symmetrical STDP learning rule. Synaptic weight distributions with high K 2 test values (>50),
indicating deviation from normality, typically contain most of the weights saturated at maximal or minimal val-
ues and thus expressed runaway dynamics. Note that in simulations with the STDP-only model (A), only a few
STDP rules, with strong bias toward depression, did not lead to runaway dynamics. Most STDP rules, including
examples shown in the bottom, led to runaway dynamics of synaptic weights. In contrast, the model with STDP
and heterosynaptic plasticity (B) did not express runaway dynamics over the whole range of tested STDP rules,
including those extremely biased toward potentiation or depression.
Modified, with permission, from Chen etal.,2013.

Heterosynaptic plasticity has a normalizing effect magnitude of potentiation is smaller at strong


on synaptic weight changes and can support syn- synapses, which probably were already poten-
aptic competition. Moreover, it shares the trigger, tiated, than at weak synapses, which did not
has partially overlapping mechanisms, and oper- experience prior potentiation or were previ-
ates on the same timescale as Hebbian-type plas- ously depressed (van Rossum et al., 2000;
ticity. Importantly, it operates at the same level Sjstrm etal., 2001; Hardingham etal., 2007).
(synapses and regulation of their weights) as the One further mechanism is a sliding calcium
Hebbian-type plasticity. These features make het- threshold for potentiation and depression,
erosynaptic plasticity an ideal candidate mecha- whereby, depending on the history of recent
nism for homeostatic control of synaptic weight activity and synaptic changes, the thresholds
changes. for potentiation and depression or intracellu-
lar calcium homeostasis change (Bienenstock
B I O L O G I C A L C A N D I D AT E S : et al., 1982; Yeung et al., 2004). These notions
OTHER MECHANISMS and mechanisms contribute to the concept of
C O U N T E R A C T I N G R U N AWAY metaplasticityhistory-dependent changes of
Extensive studies of Hebbian-type synap- the abilities of synapses to undergo potentiation
tic plasticity have suggested several further or depression (Abraham and Bear, 1996). These
mechanisms that may counteract or reduce the mechanisms are inherent to Hebbian-type plas-
tendency for runaway of synaptic weights and ticity rules and thus are ideally suited to shape
activity. One is saturation of plasticity, which the ability of synapses to change. By impos-
is the reduction of the magnitude of potentia- ing negative feedback on homosynaptic plas-
tion during a series of potentiation-inducing tic changes, they clearly can limit the runaway
episodes, with eventual complete loss of the tendency and thus decrease the instability of
ability for further potentiation (Colino et al., a system with plastic synapses. A drawback of
1992; Huang et al., 1992). Another one is these mechanisms, as of any mechanisms gov-
weight dependence of plasticity, whereby the erning homosynaptic plasticity, is that they
Homeostatic Role of Heterosynaptic Plasticity 137

(a) (b)
?
?

+
?
50 ms
?
?
Homosynaptic Homosynaptic Heterosynaptic
LTP LTD plasticity

(c) (d)
Homosynaptic LTP/LTD Heterosynaptic plasticity
Hom
Potentiation

Potentiation

Potentiation
He osy
ter nap
osy tic
n LTP
LT apt
Weight change

Weight change

Weight change
P ic

0 0 0 He
ter
os
LT ynap
Depression

Depression

Depression
Hom
osy D tic
nap
tic
LTD

Initial synaptic weight Initial synaptic weight Initial synaptic weight

FIGURE8.4: Scheme of induction of homosynaptic and heterosynaptic plasticity and the result-
ing changes of synaptic weights. A: In a typical plasticity experiment, a set of afferents to a neuron (red
inputs) is paired with bursts of postsynaptic spikes (red current step applied through the recording electrode).
All other inputs to this neuron (green ? marks) are not stimulated and thus represent heterosynaptic sites.
B: Scheme of STDP protocols for induction of homosynaptic potentiation (red, presynaptic stimulation before
postsynaptic firing), for homosynaptic depression (blue, presynaptic stimulation after postsynaptic spikes) and
purely postsynaptic challenge (green, postsynaptic spikes without presynaptic stimulation, as experienced dur-
ing plasticity induction by heterosynaptic sites, which are marked by the green ? symbolsinA). C: Scheme of
weight-dependent plastic changes in response to induction protocols from B.Left:expected outcome of homosyn-
aptic, associative Hebbian plasticity after induction of potentiation (red area) or depression (blue area). Changes at
few synapses, which were activated during the induction, are shown as circle symbols. Note the weight dependence
of both homosynaptic LTP and LTD, whereby very weak synapses exhibit less depression and very strong synapses
exhibit less potentiation. Right:expected outcome of heterosynaptic plasticity. Many more synapses may express
heterosynaptic changes (circle symbols), because the majority of synapses onto a neuron were not activated dur-
ing the induction but experienced strong postsynaptic spiking. Note that both the magnitude and the direction
of heterosynaptic changes depends on initial synaptic weight, so that initially strong synapses tend to depress,
while initially weak synapses tend to potentiate. D: Driving forces on weight changes imposed by homosynaptic
and heterosynaptic plasticity. Homosynaptic plasticity, LTP or LTD, is induced only at synapses active during the
induction and requires specific-activity patterns. Homosynaptic LTP leads to weight-dependent increase of syn-
aptic weights:potentiation is stronger at initially weak than at initially strong synapses. Homosynaptic LTD leads
to weight-dependent decrease of synaptic weights:depression of strong synapses is stronger than depression of the
weak synapses. Heterosynaptic plasticity can be induced at any synapse during episodes of strong postsynaptic
activity. Initially weak synapses are subject to weight-dependent heterosynaptic LTP. Initially strong synapses are
subject to weight-dependent heterosynapticLTD.

require presynaptic activation and cannot affect all respective synapses (Bliss and Lomo, 1973;
affect inactive synapses. This requirement lim- Daoudal et al., 2002; Zhang and Linden, 2003;
its the ability of homosynaptic mechanisms to Frick et al., 2004; Fink and ODell, 2009; Sehgal
serve as regulators of global, cell-wide synaptic et al., 2013). Excitability changes may counteract
homeostasis. synaptic changes, thus having a homeostatic effect
A family of nonsynaptic mechanisms regulat- (Zhang and Linden, 2003), or enhance and amplify
ing intrinsic excitability does not have this limita- synaptic changes, thus having an antihomeostatic
tion. These mechanisms can change excitability effect (Frick etal., 2004; Fink and ODell, 2009; see
of a dendritic branch or a whole neuron and thus Sehgal etal., 2013 for a recent review).
138 Part II: Homeostatic Control

CONCLUSIONS References
The interacting mechanisms of homosynaptic Abbott, L.F., Nelson, S.B. (2000). Synaptic plastic-
and heterosynaptic plasticity impose different ity:Taming the beast. Nat Neurosci 3, 11781183.
forces on synaptic weights, which drive synaptic Abraham, W.C., Bear, M.F. (1996). Metaplasticity:The
weights in opposite directions. Homosynaptic plasticity of synaptic plasticity. Trends Neurosci
plasticity induced by selective and specific activ- 19(4), 126130.
ity patterns drives synaptic weights to the limits Aitkin, L.M., Anderson, D.J., Brugge, J.F. (1970).
of their dynamic range. Heterosynaptic plasticity Tonotopic organization and discharge charac-
teristics of single neurons in nuclei of the lateral
simultaneously introduces a normalizing driving
lemniscus of the cat. J Neurophysiol 33, 421440.
force that pulls synaptic weight values away from
Artola, A., Singer, W. (1993). Long-term depres-
the extremes. As a consequence, every spike, or
sion of excitatory synaptic transmission and its
burst of spikes, becomes a homeostatic signal to
relationship to long-term potentiation. Trends
the cell. Because homosynaptic and heterosyn-
Neurosci 16(11), 480487.
aptic changes are induced by the same activity
Babadi, B., Abbot, L.F. (2010). Intrinsic stability of
patterns and take place on the same timescale, temporally shifted spike-timing dependent plas-
the weight of a synapse is determined by the bal- ticity. PLoS Comput Biol 6(11), e1000961.
ance of homosynaptic LTP, homosynaptic LTD, Becker, D., Zahn, N., Deller, T., Vlachos, A. (2013).
and the normalizing force of heterosynaptic Tumor necrosis factor alpha maintains denerva-
plasticity (Figure 8.4B). At strongly activated tion-induced homeostatic synaptic plasticity of
homosynaptic sites, the associative driving force mouse dentate granule cells. Front Cell Neurosci
may be dominant, leading to net potentiation or 7, 257.
depression of these subpopulations of synapses Bi, G.Q., Poo, M.M. (1998). Synaptic modifications
(Figure8.4C, left). Meanwhile the vast number of in cultured hippocampal neurons: Dependence
synapses that are at that moment inactive will be on spike timing, synaptic strength, and postsyn-
subject to the stabilizing effect of heterosynaptic aptic cell type. J Neurosci 18(24), 1046410472.
changes (Figure 8.4C, right). This allows neu- Bienenstock, E.L., Cooper, L.N., Munro, P.W. (1982).
rons to update relative strength of inputs while Theory for the development of neuron selectiv-
keeping synaptic weights within operation range ity: Orientation specificity and binocular inter-
and preserving abilities of synapses for further action in visual cortex. J Neurosci 2, 3248.
modifications. Importantly, heterosynaptic Birtoli, B., Ulrich, D. (2004). Firing mode-dependent
plasticity allows robust homeostasis of synaptic synaptic plasticity in rat neocortical pyramidal
weights and activity over a wide range of STDP neurons. J Neurosci 24, 49354940.
rules, Ca 2+ thresholds, and presynaptic activity Bliss, T.V., Collingridge, G.L. (1993). A synaptic
levels. Moreover, differential driving forces for model of memory:Long-term potentiation in the
the weight changes at active and inactive syn- hippocampus. Nature 361, 3139.
apses introduce competition between synapses Bliss, T.V., Lomo, T. (1973). Long-lasting potentia-
(Figure 8.4D). This competitive force imposed tion of synaptic transmission in the dentate area
by heterosynaptic plasticity emerges simultane- of the anaesthetized rabbit following stimulation
ously with Hebbian plasticity, fighting not just of the perforant path. J Physiol 232, 331356.
over shared mechanisms or resources but push- Bhme, G.A., Bon, C., Stutzmann, J.M., Doble, A.,
ing synapses with opposing driving forces, the Blanchard, J.C. (1991). Possible involvement
result of which is a balance between the forma- of nitric oxide in long-term potentiation. Eur J
tion of new patterns of synaptic weights and Pharmacol 199(3), 379381.
biological stability. Therefore heterosynaptic Bonhoeffer, T., Staiger, V., Aersten, A. (1989).
plasticity expresses all of the desired features of Synaptic plasticity in rat hippocampal slice cul-
tures: local Hebbian conjunction of pre- and
an intrinsic homeostatic mechanism for stabiliz-
postsynaptic stimulation leads to distributed
ing synaptic weight dynamics after learning.
synaptic enhancement. Proc Natl Acad Sci USA
86, 81138117.
ACK NOWLEDGMENTS Branco, T., Staras, K., Darcy, K.J., Goda, Y. (2008).
We are grateful for the support from the National Local dendritic activity sets release probability at
Institutes of Health (Grant R01MH087631) hippocampal synapses. Neuron 59, 475485.
and the Alexander von Humboldt-Foundation Bright, D.P., Brickley, S.G. (2008). Acting locally but
(Humboldt Research Award toMV). sensing globally:Impact of GABAergic synaptic
Homeostatic Role of Heterosynaptic Plasticity 139

plasticity on phasic and tonic inhibition in the Elliott, T., Shadbolt, N.R. (2002). Multiplicative syn-
thalamus. J Physiol 586(21), 50915099. aptic normalization and a nonlinear Hebb rule
Burrone, J., OByrne, M., Murthy, V.N. (2002). underlie a neurotrophic model of competitive
Multiple forms of synaptic plasticity triggered synaptic plasticity. Neural Comput 14, 13111322.
by selective suppression of activity in individual Engert, F., Bonhoeffer, T. (1997). Synapse specificity
neurons. Nature 420, 414418. of long-term potentiation breaks Down at short
Caporale, N., Dan, Y. (2008). Spike timing-depen- distances. Nature 388, 279284.
dent plasticity: A Hebbian learning rule. Annu Feldman, D.E. (2009). Synaptic mechanisms for
Rev Neurosci 31, 2546. plasticity in neocortex. Annu Rev Neurosci
Chen, J.Y., Lonjers, P., Lee, C., Chistiakova, M., 32, 33-55.
Volgushev, M., Bazhenov, M. (2013). Finelli, L.A., Haney, S., Bazhenov, M., Stopfer, M.,
Heterosynaptic plasticity prevents runaway syn- Sejnowski, T.J. (2008). Synaptic learning rules
aptic dynamics. J Neurosci 33(40), 1591515929. and sparse coding in a model sensory system.
Chen, L., Lau, A.G., Sarti, F. (2014). Synaptic retinoic PLoS Comput Biol 4(4), e1000062.
acid signaling and homeostatic synaptic plastic- Fink, A.E., ODell, T.J. (2009). Short trains of theta
ity. Neuropharmacology 78, 312. frequency stimulation enhance CA1 pyramidal
Chevaleyre, V., Castillo, P.E. (2003). Heterosynaptic neuron excitability in the absence of synaptic
LTD of hippocampal GABAergic synapses: A potentiation. J Neurosci 29(36), 1120311214.
novel role of endocannabinoids in regulating Frick, A., Magee, J., Johnston, D. (2004). LTP is
excitability. Neuron 38(3), 461472. accompanied by an enhanced local excitability
Chevaleyre, V., Takahashi, K.A., Castillo, P.E. (2006). of pyramidal neuron dendrites. Nat Neurosci
Endocannabinoid-mediated synaptic plasticity 7(2), 126135.
in the CNS. Annu Rev Neurosci 29, 3776. Froemke, R.C., Poo, M., Dan, Y. (2005). Spike-
Chistiakova, M., Bannon, N.M., Bazhenov, M., timing-dependent synaptic plasticity depends on
Volgushev, M. (2014). Heterosynaptic plastic- dendritic location. Nature 434, 221225.
ity:Multiple mechanisms and multiple roles. The Gally, J.A., Montague, P.R., Reeke, G.N., Edelman,
Neuroscientist 20(5), 483498. G.M. (1990). The NO hypothesis:possible effects
Chistiakova, M., Volgushev, M. (2009). of a short-lived, rapidly diffusible signal in the
Heterosynaptic plasticity in the neocortex. Exp development and function of the nervous sys-
Brain Res 199(34), 377390. tem. Proc Natl Acad Sci USA 87(9), 35473551.
Chiu, C.Q., Puente, N., Grandes, P., Castillo, P.E. Gerstner, W., Kistler, W.M. (2002). Mathematical
(2010). Dopaminergic modulation of endocan- formulations of Hebbian learning. Biol Cybern
nabinoid-mediated plasticity at GABAergic syn- 87, 404415.
apses in the prefrontal cortex. J Neurosci 30(21), Gilson, M., Fukai, T. (2011). Stability versus neuro-
72367248. nal specialization for STDP: Long-tail weight
Colino, A., Huang, Y.Y., Malenka, R.C. (1992). distributions solve the dilemma. PLoS One 6(10),
Characterization of the integration time for the e25339.
stabilization of long-term potentiation in area Golding, N.L., Staff, N.P., Spruston, N. (2002).
CA1 of the hippocampus. J Neurosci 12, 180187. Dendritic spikes as a mechanism for coopera-
Cummings, J.A., Mulkey, R.M., Nicoll, R.A., tive long-term potentiation. Nature 418(6895),
Malenka, R.C. (1996). Ca2+ signaling require- 326331.
ments for long-term depression in the hippo- Gtig, R., Aharonov, R., Rotter, S., Sompolinsky,
campus. Neuron 16(4), 825833. H. (2003). Learning input correlations through
Daoudal, G., Hanada, Y., Debanne D. (2002). nonlinear temporally asymmetric Hebbian plas-
Bidirectional plasticity of excitatory postsynap- ticity. J Neurosci 23, 36973714.
tic potentials (EPSP)-spike coupling in CA1 hip- Haas, J.S., Nowotny, T., Abarbanel, H.D.I. (2006).
pocampal pyramidal neurons. Proc Nat Sci USA Spike-timing-dependent plasticity of inhibitory
99, 1451514517. synapses in the entorhinal cortex. J Neurophysiol
Davis, G.W., Bezprozvanny, I. (2001). Maintaining 96, 33053313.
the stability of neural function: A homeostatic Hardingham, N., Dachtler, J., Fox, K. (2013). The
hypothesis. Annu Rev Neurosci 63, 847869. role of nitric oxide in pre-synaptic plasticity and
Desai, N.S., Cudmore, R.H., Nelson, S.B., Turrigiano, homeostasis. Front Cell Neurosci 7, 190.
G.G. (2002). Critical periods for experience- Hardingham, N.R., Hardingham, G.F., Fox, K.D.,
dependent synaptic scaling in visual cortex. Nat Jack, J.B. (2007). Presynaptic efficacy directs nor-
Neurosci 5(8), 783789. malization of synaptic strength in layer 2/3 rat
140 Part II: Homeostatic Control

neocortex after paired activity. J Neurophysiol of layer 5 pyramidal cells at a critical frequency
97, 29652975. of back-propagating action potentials. Proc Natl
Hebb, D.O. (1949). The organisation of behavior Acad Sci USA 96(25), 1460014604.
(NewYork:Wiley). Lee, C.M., Stoelzel, C., Chistiakova, M., Volgushev,
Heifets, B.D., Castillo, P.E. (2009). Endocannabinoid M. (2012). Heterosynaptic plasticity induced by
signaling and long-term synaptic plasticity. intracellular tetanization in layer 2/3 pyramidal
Annu Rev Physiol, 71, 283306. neurons in rat auditory cortex. J Physiol 590(10),
Huang, Y, Yasuda, H, Sarihi, A, Tsumoto, T (2008). 22532271.
Roles of endocannabinoids in heterosynap- Letzkus, J.J., Kampa, B.M., Stuart, G.J. (2006).
tic long-term depression of excitatory synaptic Learning rules for spike timing-dependent plas-
transmission in visual cortex of young mice. J ticity depend on dendritic synapse location. J
Neurosci 28(28), 70747083. Neurosci 26, 1042010429.
Huang, Y.Y., Colino, A., Selig, D.K., Malenka, R.C. Lisman, J. (1989). A mechanism for the Hebb and
(1992). The influence of prior synaptic activity on the anti-Hebb processes underlying learning
the induction of long-term potentiation. Science and memory. Proc Natl Acad Sci USA 86(23),
7, 730733. 95749578.
Ibata, K., Sun, Q., Turrigiano, G.G. (2008). Rapid Lisman, J., Spruston, N. (2005). Postsynaptic depo-
synaptic scaling induced by changes in postsyn- larization requirements for LTP and LTD:Acri-
aptic firing. Neuron 57, 819826. tique of spike timing-dependent plasticity. Nat
Ismailov, I., Kalikulov, D., Inoue, T., Friedlander, Neurosci 8(7), 839841.
M.J. (2004). The kinetic profile of intracellular Lynch, G.S., Dunwiddie, T., Gribkoff, V. (1977).
calcium predicts long-term potentiation and Heterosynaptic depression: A postsynaptic cor-
long-term depression. J Neurosci 24, 98479861. relate of long-term potentiation. Nature 266,
Kaneko, M., Stellwagen, D., Malenka, R.C., Stryker, 737739.
M.P. (2008). Tumor necrosis factor-alpha medi- Maejima, T., Hashimoto, K., Yoshida, T., Aiba, A.,
ates one component of competitive, experience- Kano, M. (2001). Presynaptic inhibition caused
dependent plasticity in developing visual cortex. by retrograde signal from metabotropic gluta-
Neuron 58, 673680. mate to cannabinoid receptors. Neuron 31(3),
Keck, T., Keller, G.B., Jacobsen, R.I., Eysel, U.T., 463475.
Bonhoeffer, T., Hbener, M. (2013). Synaptic Magee, J.C., Johnston, D. (1997). A synaptically con-
scaling and homeostatic plasticity in the mouse trolled, associative signal for Hebbian plastic-
visual cortex in vivo. Neuron 80(2), 327334. ity in hippocampal neurons. Science 275(5297),
Kempter, R., Gerstner, W., Van Hemmen, J.L. (2001). 209213.
Intrinsic stabilization of output rates by spike- Malenka, R.C., Bear, M.F. (2004). LTP and LTD:An
based Hebbian learning. Neural Comp 13, embarrassment of riches. Neuron 44, 521.
27092741. Manninen, T., Hituri, K., Kotaleski, J.H., Blackwell,
Kossel, A., Bonhoeffer, T., Bolz, J. (1990). Non- K.T., Linne, M.-L. (2010). Postsynaptic signal
Hebbian synapses in rat visual cortex. transduction models for long-term potentia-
Neuroreport 1, 115118. tion and depression. Front Comput Neurosci.
Kuhnt, U., Voronin, L. (1994). Interaction between 4, 152.
paired-pulse facilitation and long-term poten- Markram, H., Luebke, J., Frotscher, M., Sakmann, B.
tiation in area CA1 of guinea-pig hippocam- (1997). Regulation of synaptic efficacy by coin-
pal slices: Application of quantal analysis. cidence of postsynaptic APs and EPSPs. Science
Neuroscience 62, 391397. 275, 213215.
Lambo, M.E., Turrigiano, G.G. (2013). Synaptic and Merzenich, M.M., Knight, P.L., Roth, G.L. (1975).
intrinsic homeostatic mechanisms cooperate Representation of cochlea within primary audi-
to increase L2/3 pyramidal neuron excitability tory cortex in the cat. J Neurophysiol 38, 231249.
during a late phase of critical period plasticity. J Miller, K.D. (1996). Synaptic economics:Competition
Neurosci 33(20), 88108819. and cooperation in synaptic plasticity. Neuron
Lange, M.D., Doengi, M., Lesting, J., Pape, H.C., 17, 371374.
Jngling, K. (2012). Heterosynaptic long-term Miller, K.D., MacKay, D.J.C. (1994). The role of con-
potentiation at interneuronprincipal neuron straints in Hebbian learning. Neural Comp 6,
synapses in the amygdala requires nitric oxide 100126.
signalling. J Physiol 590(1), 131143. Morrison, A., Aertsen, A., Diesmann, M. (2007).
Larkum, M.E., Kaiser, K.M.M., Sakmann, B. (1999). Spike-timing-dependent plasticity in balanced
Calcium electrogenesis in distal apical dendrites random networks. Neural Comp 19, 14371467.
Homeostatic Role of Heterosynaptic Plasticity 141

Murthy, V.N., Schikorski, T., Stevens, C.F., Zhu, Y. Schuman, E.M., Madison, D.V. (1994b). Nitric oxide
(2001). Inactivity produces increases in neu- and synaptic function. Annu Rev Neurosci 17,
rotransmitter release and synapse size. Neuron 153183.
32(4), 673682. Sehgal, M., Song, C., Ehlers, V.L., Moyer, J.R. Jr
Neveu, D., Zucker, R.S. (1996). Long-lasting potenti- (2013). Learning to learnintrinsic plasticity as
ation and depression without presynaptic activ- a metaplasticity mechanism for memory forma-
ity J Neurophysiol 75, 21572160. tion. Neurobiol Learn Mem 105, 186199.
Nevian, T., Sakmann, B. (2006). Spine Ca2+ sig- Sjstrm, P.J., Husser, M. (2006). A cooperative
naling in spike-timing-dependent plasticity. J switch determines the sign of synaptic plasticity
Neurosci 26(43), 1100111013. in distal dendrites of neocortical pyramidal neu-
Nishiyama, M., Hong, K., Mikoshiba, K., Poo, M., rons. Neuron 51(2), 227238.
Kato, K. (2000). Calcium stores regulate the Sjstrm, P.J., Rancz, E.A., Roth, A., Husser, M.
polarity and input specificity of synaptic modi- (2008). Dendritic excitability and synaptic plas-
fication. Nature 408, 584588. ticity. Physiol Rev 88(2), 769840.
Nugent, F.S., Penick, E.C., Kauer, J.A. (2007). Opioids Sjstrm, P.J., Turrigiano, G.G., Nelson, S.B. (2001).
block long-term potentiation of inhibitory syn- Rate, timing, and cooperativity jointly deter-
apses. Nature 446(7139), 10861090. mine cortical synaptic plasticity. Neuron 32,
ODell, T.J., Hawkins, R.D., Kandel, E R., Arancio, 11491164.
O. (1991). Tests of the roles of two diffusible Sjstrm, P.J., Turrigiano, G.G., Nelson, S.B. (2004).
substances in long-term potentiation: evidence Endocannabinoid-dependent neocortical layer-5
for nitric oxide as a possible early retrograde LTD in the absence of postsynaptic spiking. J
messenger. Proc Natl Acad Sci USA 88(24), Neurophysiol 92(6), 33383343.
1128511289. Skorheim, S., Lonjers, P., Bazhenov, M. (2014). A spik-
Oja, E. (1982). A simplified neuron model as a ing network model of decision making employ-
principal component analyzer. J Math Biol. 15, ing rewarded STDP. PLoS One 9(3), e90821.
267273. Song, S., Miller, K.D., Abbott, L.F. (2000).
Pan, B., Hillard, C.J., Liu, Q.S. (2008). Competitive Hebbian learning through spike-
Endocannabinoid signaling mediates cocaine- timing-dependent synaptic plasticity. Nat
induced inhibitory synaptic plasticity in mid- Neurosci 3(9), 919926.
brain dopamine neurons. J Neurosci 28(6), Spruston, N., Schiller, Y., Stuart, G., Sakmann, B.
13851397. (1995). Activity-dependent action potential inva-
Phillips, K.G., Hardingham, N.R., Fox, K. (2008). sion and calcium influx into hippocampal CA1
Postsynaptic action potentials are required for dendrites. Science 268(5208), 297300.
nitric-oxide-dependent long-term potentiation Staubli, U.V., Ji, Z.X. (1996). The induction of homo-
in CA1 neurons of adult GluR1 knock-out and vs. heterosynaptic LTD in area CA1 of hippo-
wild-type mice. J Neurosci 28(52), 1403114041. campal slices from adult rats. Brain Res 714(1),
Rabinowitch, I., Segev, I. (2008). The inter- 169176.
play between homeostatic synaptic plastic- Tatavarty, V., Sun, Q., Turrigiano, G.G. (2013). How
ity and functional dendritic compartments. J to scale down postsynaptic strength. J Neurosci
Neurophysiol 96, 276283. 33(32), 1317913189.
Remy, S., Spruston, N. (2007). Dendritic spikes Tetzlaff, C., Kolodziejski, C., Timme, M., Wrgtter,
induce single-burst long-term potentiation. Proc F. (2012). Analysis of synaptic scaling in combi-
Natl Acad Sci USA 104(43), 1719217197. nation with Hebbian plasticity in several simple
Rich, M.M., Wenner, P. (2007). Sensing and express- networks. Front Comput Neurosci6, 36.
ing homeostatic synaptic plasticity. Trends in Thompson, I.D., Kossut, M., Blakemore, C. (1983).
Neurosci 30(3), 119125. Development of orientation columns in cat stri-
Royer, S., Par, D. (2003). Conservation of total syn- ate cortex revealed by 2-deoxyglucose autoradi-
aptic weight through balanced synaptic depres- ography. Nature 301, 712715.
sion and potentiation. Nature 422, 518522. Turrigiano, G.G. (2012). Homeostatic synaptic plas-
Schuman, E.M., Madison, D.V. (1991). A require- ticity: Local and global mechanisms for sta-
ment for the intercellular messenger nitric oxide bilizing neuronal function. Cold Spring Harb
in long-term potentiation. Science 254(5037), Perspect Biol 4(1), a005736.
15031506. Turrigiano, G.G., Leslie, K.R., Desai, N.S.,
Schuman, E.M., Madison, D.V. (1994a). Locally dis- Rutherford, L.C., Nelson, S.B. (1998). Activity-
tributed synaptic potentiation in the hippocam- dependent scaling of quantal amplitude in neo-
pus. Science 263, 532536. cortical neurons. Nature 391, 892896.
142 Part II: Homeostatic Control

Van Ooyen, A. (2001). Competition in the develop- tufts of layer 2/3 neocortical pyramidal neu-
ment of nerve connections: a review of models. rons in vitro and in vivo. J Neurosci 23(24),
Network:Comput Neural Syst 12, R1R47. 85588567.
Van Rossum, M.C., Bi, G.Q., Turrigiano, G.G. (2000). Watt, A.J., Desai. N.S. (2010). Homoeostatic plastic-
Stable Hebbian learning from spike timing- ity and STDP:Keeping a neurons cool in a fluc-
dependent plasticity. J Neurosci 20, 88128821. tuating world. Front Synaptic Neurosci2, 5.
Vitureira, N., Letellier, M., Goda, Y. (2012). Watt, A.J., Sjstrm, P.J., Husser, M., Nelson, S.B.,
Homeostatic synaptic plasticity: From single Turrigiano, G.G. (2004). A proportional but
synapses to neural circuits. Curr Op Neurobiol slower NMDA potentiation follows AMPA
22, 516521. potentiation in LTP. Nat Neurosci 7, 518524.
Vlachos, A., Ikenberg, B., Lenz, M., Becker, D., White, G., Levy, W.B., Steward, O. (1990). Spatial
Reifenberg, K., Bas-Orth, C., Deller, T. (2013). overlap between populations of synapses deter-
Synaptopodin regulates denervation-induced mines the extent of their associative inter-
homeostatic synaptic plasticity. Proc Natl Acad action during the induction of long-term
Sci USA 110(20), 82428247. potentiation and depression. J Neurophysiol,
Volgushev, M., Chauvette, S., Mukovski, M., 64(4), 11861198.
Timofeev, I. (2006). Precise long-range syn- Wiesel, T.N., Hubel, D.H. (1963). Effects of visual
chronization of activity and silence in neocorti- deprivation on morphology and physiology
cal neurons during slow-wave sleep. J Neurosci of cells in the cats lateral geniculate body. J
26(21), 56655672. Neurophysiol 26, 978993.
Volgushev, M., Chistiakova, M., Balaban, P., Eysel, Wu, Z., Yamaguchi, Y. (2006). Conserving total syn-
U.T. (2000). Retrograde signaling with nitric aptic weight ensures one-trial sequence learning
oxide at neocortical synapses. Eur J Neurosci 12, of place fields in the hippocampus. Neural Netw
42554267. 19, 547563.
Volgushev, M., Mittmann, T., Chistiakova, M., Yang, S.N., Tang, Y.G., Zucker, R.S. (1999). Selective
Balaban, P., Eysel, U.T. (1999). Interaction induction of LTP and LTD by postsynaptic
between intracellular tetanization and pairing- [Ca2+]i elevation. J Neurophysiol 81, 781787.
induced long-term synaptic plasticity in the rat Yasuda, H., Huang, Y., Tsumoto, T. (2008).
visual cortex. Neuroscience 93, 12271232. Regulation of excitability and plasticity by endo-
Volgushev, M., Pernberg, J., Eysel, U.T. (2002). A novel cannabinoids and PKA in developing hippocam-
mechanism of response selectivity of neurons in pus. Proc Natl Acad Sci USA 105(8), 31063111.
cat visual cortex. J Physiol 540(1), 307320. Yeung, L.C., Shouval, H.Z., Blais, B.S., Cooper,
Volgushev, M., Pernberg, J., Eysel, U.T. (2003). L.N. (2004). Synaptic homeostasis and input
Gamma-frequency fluctuations of the membrane selectivity follow from a calcium-dependent
potential and response selectivity in visual corti- plasticity model. Proc Natl Acad Sci USA 101,
cal neurons. Eur J Neurosci 17(9), 17681776. 1494314948.
Volgushev, M., Voronin, L.L., Chistiakova, M., Zenke, F., Hennequin, G., Gerstner, W. (2013).
Singer, W. (1994). Induction of LTP and LTD in Synaptic plasticity in neural networks needs
visual cortex neurons by intracellular tetaniza- homeostasis with a fast rate detector. PLoS
tion. Neuroreport 5, 20692072. Comput Biol 9, e1003330.
Volgushev, M., Voronin, L.L., Chistiakova, M., Singer, Zhang, W., Linden, D.J. (2003). The other side of the
W. (1997). Relations between long-term synaptic engram:Experience-driven changes in neuronal
modifications and paired-pulse interactions in intrinsic excitability. Nature Rev Neurosci 4(11),
the rat neocortex. Eur J Neurosci 9, 16561665. 885900.
Von der Malsburg, C. (1973). Self-organization of Zhou, Y.D., Acker, C.D., Netoff, T.I., Sen, K., White,
orientation sensitive cells in the striate cortex. J.A. (2005). Increasing Ca2+ transients by broad-
Kybernetik 14, 85100. ening postsynaptic action potentials enhances
Waters, J., Larkum, M., Sakmann, B., Helmchen, F. timing-dependent synaptic depression. Proc
(2003). Supralinear Ca2+ influx into dendritic Natl Acad Sci USA 102, 1912119125.
9
The Blood-Brain Barrier
Y O N ATA N S E R L I N , A L O N F R I E D M A N , A N D U W E H E I N E M A N N

THE BLOOD-BR AIN BARRIER endothelium is surrounded by cellular elements


I N T H E H E A LT H Y B R A I N such as pericytes and astroglial foot processes,
forming an additional continuous stratum that
Anatomy separates blood vessels from brain tissue. Around
To maintain normal brain function, the neural penetrating vessels and venules there is often some
extracellular environment must be maintained distance between EC and brain tissue, forming
within a narrow homeostatic range; this requires the Virchow-Robin space in which perivascular
a tight regulation of transportation of cells, mol- macrophages are found, which execute some of
ecules, and ions between the blood and the brain. the immune functions of the CNS. The intimate
Such tight regulation is maintained by a unique contact between neurons, astrocytes, microglia,
anatomical and physiological barrier formed col- pericytes, and blood vessels, and the functional
lectively in the central nervous system (CNS). interactions and signaling between these cells,
Awareness of existence of a physical interface form a dynamic functional unit known as the
between the CNS and the peripheral circulation neurovascular unit. Understanding the function
and the vascular capacity was first described of the neurovascular unit is key to our under-
by Paul Ehrlich (1885). Ehrlich described how standing of brain functions in health and disease,
dye injection into the blood circulation stained including neuronal firing, synaptic plasticity,
peripheral organs but not the spinal cord and the regulation of blood flow, and response to injury
brain. Later, Ehrlichs student Edwin Goldmann (thoroughly reviewed by Abbott,2014).
showed that direct injection of trypan blue into The neurovascular unit is illustrated in
the cerebrospinal fluid (CSF)-stained cells within Figure 9.1. The innermost luminal constituent
the CNS and not in the periphery (Goldmann, is comprised of a single specialized EC layer lin-
1913). An additional limiting element was later ing brain capillaries, exhibiting a greater num-
demonstrated by Reese and Karnovsky (1967) ber and volume of mitochondria, augmenting
who presented a solute exchange barrier between the selective molecular permeability of the BBB
the blood and the brain by means of endothelial (Oldendorf et al., 1977). The basement mem-
tight junction complexes. brane, a 30- to 40-nm thick lamina composed
Three barrier layers contribute to the separa- of collagen type IV, heparin sulfate proteogly-
tion of the blood and neural tissues:(a)a highly cans, laminin, fibronectin, and other extracel-
specialized endothelial cells (EC) layer compris- lular matrix proteins, encompasses pericytes
ing the blood-brain barrier (BBB) and partition- and EC and is closely adjacent to the plasma
ing the blood and brain interstitial fluid; (b)the membranes of astrocyte endfeet, enclosing the
blood-CSF barrier with the choroid plexus epi- cerebral capillaries (Hawkins and Davis, 2005).
thelium, which secretes CSF into the cerebral Transmembrane proteins (junctional adhesion
ventricles and separates the blood from the molecule-1, occludin, and claudins1, 3, 5, and
ventricular CSF; and (c) the arachnoid epithe- possibly 12)and cytoplasmic accessory proteins
lium between the blood and subarachnoid CSF (zonula occludens-1 and -2, cingulin, AF-6, and
(Abbott etal.,2006). 7H6) establish the tight junctions between adja-
The BBB components include the EC layer cent EC. Molecular and structural studies of
and its basement membrane, adjoined by tight tight junctions reveal a complex, dynamic, and
cell-to-cell junction proteins with specific trans- highly regulated molecular structure. Junctional
port mechanisms and pinocytic vesicles. The adhesion molecules maintain tight junction
144 Part II:Homeostatic Control

in homeostasis of extracellular concentration of


transmitters, metabolites, ions, and water but
also serve as stem cells early during development
and provide for templates for migratory neuronal
streams. Interaction between astrocytes and
neurons determine synaptic transmission, clear-
ance of neurotransmitters, plasticity, and control
blood flow (reviewed by Wong etal.,2013).

Development
A key stage in the development of the BBB lies
in the early communications of the evolving
endothelium with neural cells (Stewart and
Wiley, 1981). The BBB matures during fetal life
and is well formed by birth (Keep et al., 1995;
Moos and Mllgrd, 1993; Olsson et al., 1968;
Saunders, 1992; Saunders et al., 2000; Tauc
et al., 1984). Transport mechanisms may con-
tinue to develop in mammals born in a relatively
immature state (such as the rat and mouse) and
become fully functional only in the peri- or post-
natal period (Jones et al., 1992). The develop-
FIGURE9.1: The neurovascular unit. A schematic ment of the vascular endothelium is now known
drawing of the neurovascular unit showing the close to be provoked by neuroepithelial signaling
spatial relationships and interactions between endothe- through the Wnt/-catenin pathway to induce a
lial cells, pericytes, astrocytes, microglia, and neurons. CNS-specific vascular system and BBB speciali-
Adopted from Abbott and Friedman,2012. zation (Daneman etal., 2009; Liebner and Plate,
2010; Stenman etal.,2008).
An early feature of BBB development is the
properties, claudins facilitate tight barrier capa- formation of tight junctions. In humans, the
bilities, and occludins and zonula occludens-1 brain of a 14-week fetus expresses occludin and
regulate targeted signaling (Abbott et al., 2010; claudin-5 in the capillary endothelium with the
Bazzoni, 2006; Chen and Liu,2012). same distribution at cell margins as seen in an
Pericytes are enveloping brain microvessels adult (Virgintino etal., 2004). Human postmor-
and capillaries and are found in close proxim- tem studies of perinatal deaths and stillborn
ity to astrocytes and neurons. The ratio of peri- fetuses from approximately 12 weeks gestation
cytes to EC is assessed to be 1:3 (Shepro and have demonstrated that a barrier to trypan blue
Morel, 1993). Using multiple signaling path- exist from at least the beginning of the second
ways, pericytes play a critical role in the forma- trimester, equivalently to that of the adult human
tion and maturation of the BBB during tissue (Grontoft, 1954). Culture studies suggest that
development and regulation of tissue survival astrocytes have a key role in regulating the tight-
(Daneman et al., 2010). Pericytes control over ness of the BBB (Hartmann etal.,2007).
cerebral blood flow is potentially due to a regu-
latory effect on the capillary diameter through Physiology
actin fibers in the pericytic cell body (Hamilton Each of the three main CNS interface layersthe
etal., 2010). Pericyte dysfunction through aging BBB, the choroid plexus epithelium, and the epi-
was noted in animal models (Bell et al., 2010), thelium of the arachnoid materfunctions as a
and their absence results in loss of BBB integrity physical, transport, metabolic, and immunologic
and reduction in regional cerebral blood flow barrier. The barrier functions are dynamic and
(Armulik etal.,2010). respond to a wide range of regulatory signals
Astrocytes interact with pericytes and micro- from the blood and the brain. Tight junctions
vascular EC by endfeet protrusions ensheathing between adjacent cells restrict diffusion of polar
the capillaries. Interactions may also exist with solutes through the intercellular cleft (paracel-
smooth muscle cells at arterioles. Astrocytes play lular pathway). The barriers are permeable to O2
important roles in maintenance of the BBB and and CO2 and other gaseous molecules such as
The Blood-Brain Barrier 145

helium, xenon, N2, and many gaseous anesthet- CNS through cytokine-activated brain endothe-
ics. The permeability to xenon may provide a lium (Greenwood etal., 2003). Transport systems
high-resolution magnetic resonance imaging tool across the BBB are illustrated in Figure9.2.
by which small morphological alteration may be
detectable within the living tissue and also per- TR ANSPORT OFGLUCOSE
mit the analysis of binding sites using molecu- AND AMINOACIDS
lar probing techniques. Lipid soluble substances Carrier-mediated influx, which may be passive
can pass the BBB by diffusion. Principally, the or secondarily active, provide transport into
BBB is also permeable to water; however, sol- the CNS of essential polar molecules that can-
ute carriers on the apical and basal membranes not diffuse through the cell membrane, such as
together with ectoenzymes and endoenzymes glucose, amino acids, and nucleosides. EC of the
regulate small solute entry and efflux. Transfer brain microvasculature, astrocytes, and the cho-
of drugs and other agents is regulated by multi- roid plexus express the insulin-independent glu-
drug transporters that can limit their concentra- cose transporter GLUT1, a membrane-spanning
tions within the CNS. Multidrug transporters are glycoprotein containing 12 transmembrane
ubiquitous transport proteins that exploit adeno- domains with a single N-glycosylation site
sine triphosphate hydrolysis to funnel molecules (Carruthers etal., 2009). GLUT1 plays a vital role
across lipid membranes; they facilitate transport in brain glucose uptake and is highly expressed
of molecules into cells but may also prevent accu- in cells forming the blood-tissue barriers and in
mulation of molecules within the brain inter- astrocytes (Mann et al., 2003). Glucose concen-
stitial space. Multidrug resistance proteins and trations play a role in the regulation of GLUT1
P-glycoprotein-like proteins are expressed at the protein concentrations: hypoglycemia induces
BBB and limit access of drugs to the brain but upregulation of GLUT1, while hyperglycemia
also of other lipophilic molecules, including the does not seem to exhibit an effect (Devaskar etal.,
neurotoxic bilirubin, the degradation product of 1991; Simpson et al., 1999). Active mechanisms
hemoglobin, whichif entering the CNScan for controlling sugar transport across the BBB
cause major damage to the CNS (Abbott and and in astrocytes might be influenced by acute
Friedman, 2012). Recent studies indicate that regulation of cell surface GLUT1 levels (Simpson
transporter molecules are upregulated in some et al., 2001) and are potentially related to the
pathological conditions and thereby reduce drug energetic condition of brain tissue. Importantly,
levels within the brain and thus may explain GLUT1 is not the sole glucose transporter at the
treatment failures in some neurological and psy- BBB; the GLUT4 transporter for glucose seems
chiatric disorders (Potschka,2010). to be expressed as well. GLUT3 is expressed in
Large molecules (e.g., peptides and pro- neurons and is likely providing glucose uptake
teins) with particular growth and signal- into neurons, thus bypassing the glucose lactate
ing roles within the CNS enter the brain in a shuffle through astrocytes by which lactate is
restricted and regulated manner by adsorptive provided as an energy-rich substrate.
and receptor-mediated transcytosis. Smaller Another important aspect is brain protection
peptides may cross the BBB by either nonspecific against neuroactive substances such as aspartate
fluid-phase endocytosis or receptor-mediated and glutamate. The BBB is largely impermeable
transcytosis mechanisms. Similarly, 98% of to these amino acids. Glutamate metabolism by
all small molecules are not freely transported the liver provides for prompt transformation into
across the BBB (Pardridge, 2005). The barriers glutamine, and, consequently, consumption of
also regulate the recruitment and entry of leu- food containing high levels of glutamate such as
kocytes and innate immune elements and are tomatoes or food additives such as soy sauce does
involved in both the reactive and the surveil- not affect brain function. Aspartate consumed
lance functions of CNS immunity. Leukocyte by food is rapidly secreted through the kidney
migration involves a complex set of adhesion (Beyreuther etal.,2007).
molecules at the surface of leukocytes and vas- The limited transport of circulating mono-
cular EC. Tethering and rolling of leukocytes is amines through EC is attributed to the para-
achieved via integrins VLA-4 (41) and 47 cellular barrier capacity of tight junctions, the
(Laschinger and Engelhardt, 2000), and adhe- diffusional characteristics imposed by the lipid
sion molecules such as ICAM-1, VCAM-1, and bilayer, and specific transport proteins of the cell
PECAM-1 contribute to the adhesion and/or membrane. Intracellular levels of amino acids
migration of distinct subsets of leukocytes to the in the brain are correlated to the rates of influx
146 Part II:Homeostatic Control

Cell migration Passive Carrier- Carrier- Receptor- Adsorptive- Tight-


diffusion mediated mediated mediated mediated junction
efflux influx transocytosis transocytosis modulation

Lipid soluble drugs

Lipid soluble Amines Cytokines Cationised Polar solutes


Non-polar solutes Amino acids Insulin albumin
Glucose Transferrin Histones
Neucleosides
Small peptides

FIGURE 9.2: Potential routes for infiltration and transport across the endothelial cells forming
the blood-brain barrier (BBB). Cells may cross the BBB through or adjacent to the tight junctions. Solutes may
passively diffuse through the cell membrane. Active efflux carriers may pump some of these passively pen-
etrating solutes out of the endothelial cell. Carrier-mediated influx (passive or secondarily active) can trans-
port essential polar molecules, such as amino acids, glucose, and nucleosides, into the central nervous system.
Receptor-mediated transcytosis can transport macromolecules such as peptides and proteins across the endothe-
lium. Adsorptive-mediated transcytosis is induced nonspecifically by positively charged macromolecules and can
result in passage across the BBB. Tight junction modulation may occur, affecting the permeability of the paracel-
lular aqueous diffusional pathway.
Modified from Begley and Brightman,2003.

across the BBB, and synthesis of neurotrans- plasma (approximately 4.5 mM). In fact, potas-
mitters such as serotonin, dopamine, and hista- sium concentration can vary strongly during
mine are substrate limited (Smith and Takasato, body exercise, nutrition, or pathological con-
1986). For a sustainable supply of essential polar ditions (Bradbury et al., 1963; Hansen, 1985;
nutrients such as glucose and amino acids to the Medb and Sejersted, 1990)and may increase to
brain, there is a crucial role to specific solute car- levels as high as 10 mM in venous blood. If such
riers (SLC) across the BBB endothelium, and all an increase in potassium concentrations would
cells express a large number of SLCs in the cell occur in the brain, a significant change in neu-
membrane (Zhang et al., 2002). A wide variety ronal activity, often epileptic discharges, would
of SLCs mediate the movement of nutrients and be triggered. The BBB thereby protects the nerve
solutes in and out of the brain and are found on cells from such variations. The BBB is similarly
either the luminal or the abluminal membrane largely impermeable to most ions such as calcium
only or inserted into both membranes of the EC. (Ca2+) and magnesium (Mg2+).
Adetailed list of BBB SLCs are listed elsewhere Within the CNS, increases in [K+]o per action
(see Table2 in Abbott etal.,2010). potential have been estimated to be around 0.01
to 0.02 mM (Heinemann and Lux, 1977; Lux
TR ANSPORT OFIONS etal., 1986). During intense sensory stimulation
Preservation of an optimal environment for syn- (e.g., painful stimuli), [K+]o rises in the dorsal
aptic and neural function is achieved by specific horn by more than 2.5 mM (Heinemann et al.,
ion channels and transporters. Water molecules 1990; Lothman and Somjen, 1975; Sykov etal.,
can also cross the BBB through ion channels. 1980). A baseline elevation in [K+]o by about 1
Due to regulated ionic movement, potassium mM has also been observed during slow-wave
concentration in the CSF and brain interstitial sleep (Amzica etal., 2002)accompanied by a fall
fluid are maintained at 2.5 to 2.9 mM despite in [Ca2+]o by 0.2 mM (Massimini and Amzica,
the higher concentration of potassium in the 2001). As information is transmitted to the
The Blood-Brain Barrier 147

thalamus and cortex, increases in [K+]o during is distributed spatially via astrocytes and water
sensory stimulation become smaller, probably is excreted from the brain. Astrocytes have a key
reflecting sparsification of action potential firing. role in the homeostatic mechanisms maintaining
In the cortex, during sensory stimulation, typi- brain extracellular environment within a nar-
cally [K+]o rises to about 0.4 mM (Lux and Singer, row limit, despite continuous neuronal activity,
1973). Changes in [Ca2+]o are indeed small during and the perivascular endfeet at the BBB have a
sensory stimulation (in the cortex less than 0.2 particular role in these processes (Simard and
mM), while during seizures [Ca 2+]o can fall from Nedergaard, 2004). For example, extracellular
the baseline concentration of 1.2 mM to less than K+ ions accumulating during neuronal activ-
0.1 mM (Pumain et al., 1985). During seizures, ity are expected to enter astrocytes according to
potassium concentration can increase to 12 mM the electrochemical gradient and distributed to
in limbic structures and to 10 mM in the cortex neighboring astrocytes (via gap junctions) and
(Lux et al., 1986). Unless the BBB is damaged, astrocytic endfeet. The high density of inwardly
this increase in [K+]o will not affect serum potas- rectifying Kir4.1 on perivascular astrocytic end-
sium concentration. The rise in serum [K+] dur- feet makes them well suited for spatial buffering,
ing convulsions is in fact mostly due to striated depositing the K+ in the perivascular space. The
muscle activity. Based on observation during high density of AQP4 water channels in perivas-
epileptic activity, it was suggested that neurovas- cular astrocytic endfeet facilitates a similar
cular coupling depends on changes in potassium redistribution of water. Excess metabolic water
and calcium concentration (Kuschinsky et al., may join the interstitial fluid in perivascular
1972). Indeed, elevations in potassium concentra- spaces and cleared through the CSF. Similarly,
tion can hyperpolarize smooth muscle cells and the uptake of glutamate to astrocytes via specific
thereby cause vessel dilation (Nelson and Quayle, transporters (mainly EAAT1 and EAAT2) is Na+
1995), while reductions in Ca 2+ concentration dependent and accompanied by net uptake of
would reduce Ca2+ entry during electrogenesis ions and water, which will similarly clear at the
in these cells and therefore weaken muscle con- perivascular space and theBBB.
traction. Thus neurovascular coupling during
seizures and other pathological activities differs MACROMOLECULES:
from that observed during physiological acti- PEPTIDES AND PROTEINS
vation, where other signals are used to regulate Endocytic vesicles account for the main deliv-
blood flow (Attwell etal., 2010). Accordingly, the ery of large molecular weight substances such as
small changes in [K+]o and [Ca2+]o may have sign- proteins and peptides through the BBB. Protein
aling functions onto other nerve cells and glia synthesis in the brain is dependent on the sup-
but probably are not a major factor in physiologi- ply of essential amino acids; most are neutral
cal neurovascular coupling. However, alterations and large and thus incapable of passive diffu-
in the electrolyte composition may contribute to sion to the brain. The typical concentrations of
the steady potentials observed, for example, dur- plasma proteins are higher than the CSF protein
ing slow-wavesleep. content, apparently due to the ability of the BBB
pH also is actively regulated at the BBB to preclude the penetration of such macromol-
and the blood-CSF barrier (Jeong et al., 2006; ecules into the brain. Vesicular mechanisms
Michalke and Nischwitz, 2010). The neurovas- involve either receptor-mediated transcytosis or
cular unit and the BBB are also important in adsorptive-mediated transcytosis enabling the
the spatial buffering of electrolytes on neuronal transport of diverse large molecules and com-
activation. Astrocytes and their position between plexes. Asummary of a number of known tran-
capillaries and neurons are connected with gap scytotic mechanisms is presented elsewhere (see
junctions, allowing them to communicate with Table 4 in Abbott et al., 2010). Internalization
each other and with capillary EC in contact with into the endothelial cell cytoplasm and exocyto-
astrocytic processes. Neuronal firing and synap- sis to the opposite pole of the cell occurs follow-
tic transmission are associated with the influx of ing interaction between ligands and cell-surface
Na+ and Ca++ and the extracellular increase in the receptors, which leads to caveolae and vesicle
concentrations of K+ and neurotransmitters. In formation.
addition, glucose metabolism during neuronal A possible mechanism for a peptide-specific
activity generates water at the rate of ~28 nl/g transporter protein may facilitate its entrance
min-1 (Rapoport, 1976). While neurotransmitters through the membrane (Dogrukol-Ak et al.,
are recycled directly or via astrocytes, potassium 2009; Kastin and Pan, 2003). Agrowing body of
148 Part II:Homeostatic Control

evidence indicates that large molecular weight of local blood flow. This complex process also
serum proteins infiltration though a dysfunc- involves pericytes, microglia, and specialized cel-
tional BBB carries a potential risk for patho- lular compartments such as endothelial glycoca-
logical outcomes within the CNS. Thrombin, lyx (Stanimirovic and Friedman, 2012). While the
plasmin, and albumin were reported to induce role of the intact BBB in controlling the normal
local effects such as cellular activation, apopto- neurovascular coupling is not completely under-
sis, and epileptogenesis. The presence of some stood, recent studies in injured patients hint that
proteins in brain interstitial fluid can initiate under conditions in which the BBB is severely
signaling cascades resulting in seizures, acti- impaired, vessels show impaired response to
vation of glia, synaptic plasticity, and synap- neuronal activation (Winkler etal.,2012).
togenesis and cell damage (David et al., 2009; The BBB plays a crucial role in the main-
Lapilover etal., 2012; Maggio etal., 2008). The tenance of a strict extracellular environment
wide presence and expression of factor Xa (con- around synapses and axons. Neuron interfaces
verting prothrombin to thrombin), tissue plas- within the CNS depend on chemical and elec-
minogen activator (converting plasminogen trical signals, and thus a steady neural function
to plasmin), and the thrombin receptor PAR1 is dependent on the barrier capacity. Following
likely play a role in these pathological path- BBB dysfunction the extracellular microenviron-
ways (Gingrich and Traynelis, 2000; Gingrich ment is disturbed, thus resulting in abnormal
etal., 2000). Albumin extravasations from the neuronal activity and under some conditions lead
plasma into the brain milieu has been shown to to seizures (Marchi etal., 2007). Sensory-motor
be associated with astrocytic activation and the neurological dysfunction developing after patho-
development of network modifications leading logical vascular response and BBB opening may
to epilepsy (David etal., 2009; Ivens etal., 2007; be attributed to reduced metabolic efficacy, cel-
Nadal etal., 1995; also see later discussion). lular damage, and interference of homeostatic
mechanisms such as active transporters and elec-
trolyte buffering, required for neuronal activity
DRUG DELIVERY (Friedman, 2011). As mentioned, the importance
As mentioned, the fact that penetration of large of intact BBB in maintaining the orchestrated
molecules from the blood into the brain is pre- relationship between brain activity and changes
vented by the BBB evokes essential research and in blood flow is considered a key determinant
translational efforts aimed at development of in the pathophysiology of brain injuries. While
novel treatments for many CNS pathologies and neurovascular coupling may reflect a physiologi-
new radiopharmaceuticals for radio-labeled cal homeostatic response to increased metabolic
brain imaging techniques. A recent, thor- demand, recent animal and human data suggest
ough review (Pardridge, 2012) provides an that reduced energy supply and worsening of the
updated outline of classical modes of drug tissue metabolic state will promote cellular dam-
delivery to the brain, including transcranial age and slow energy-demanding homeostatic
delivery of drugs or small molecules, endog- mechanisms as active transporters required
enous carrier-mediated and receptor-mediated for neuronal repolarization. Under pathologi-
transport systems within the BBB and current cal conditions, the physiological neurovascular
approaches for reengineering of drugs to enable coupling may fail, and neuronal depolarization
BBB transport. during seizures or spreading depolarization may
be associated with no or inverse coupling (i.e.,
N E U R O N A L A N D VA S C U L A R vasoconstriction; Dreier,2011).
FUNCTIONS
It is well known that despite the fact that the BBB INDISEASE
human brain constitutes only ~2% of total body There is no dispute that increase in the perme-
mass, over 20% of total body oxygen and energy ability of the BBB is associated with brain influx
consumption is dedicated to proper brain func- of circulating proteins, water, and cells, which
tion (Shulman etal., 2003). The term neurovas- further lead to neurological manifestations.
cular coupling designates an integrated system Growing attention is currently being paid to
of neuronal and vascular cells and their milieu the potential role of more subtle and transient
working in concert to maintain brain homeosta- changes in BBB integrity as triggers for abnormal
sis by providing the energy demands of neuronal brain signalling, leading to a neurological disor-
activity via a tight, activity-dependent regulation der. Indeed, in recent years, remarkable human
The Blood-Brain Barrier 149

and animal data have accumulated, revealing of brain adenosine triphosphate-binding cas-
that BBB failure is a major determinant of neuro- sette transporter expression may also change,
logical disorders and in some cases may actually with upregulation of Pgp on astrocytes and
be the cause of disease. brain endothelium (Abbott etal., 2002; Marroni
etal.,2003).
BBB Dysfunction Is Common
in Neurological Diseases BBB Dysfunction Leads to
BBB dysfunction has been reported in patients Reorganization and Dysfunction
diagnosed with the most common neurologi- of the Neuronal Network
cal disorders. Leaky capillaries in primary brain Accumulating data suggest that increased syn-
tumors has become a major imaging hallmark, chronicity and excitability of the neuronal
seen as contrast enhancement in brain comput- network is promoted by an increase in BBB per-
erized tomography or magnetic resonance imag- meability. Acute and transient dysfunction of the
ing. Similarly, ring enhancement representing BBB may be associated with changes in the brain
BBB breakdown around traumatic, ischemic, and extracellular ionic environment (e.g., increase in
infectious brain injuries are common. A more [K+]o and decrease in [Ca 2+]o and [Mg2+]o; Ivens
subtle increase in BBB permeability has been et al., 2007; Marchi et al., 2007)reducing the
reported in patients with subarachnoid hemor- threshold for seizures. Clinical reports have
rhage and Alzheimers disease (AD). In animal implied a role for BBB breakdown in familial
models of AD, the accumulation of amyloid is hemiplegic migraineassociated spreading depo-
often first seen in close proximity to blood ves- larization (Dreier etal., 2005)and in the patho-
sels, with increased vessel permeability before genesis of seizures in the cerebral hyperperfusion
significant neuronal loss. The data from ani- syndrome (Ivens etal., 2010). It seems that while
mal models of AD showing subclinical seizures water influx and edema may contribute to patho-
(Noebels, 2011)is consistent with the hypothesis logical mechanism following BBB disruption
that a leaky BBB increases the network tendency and may reduce perfusion and disturb neuro-
to seize. Recent meta-analysis based on imaging vascular coupling, additional mechanisms are
and biochemical CSF studies for assessment of involved. Macromolecules, specifically serum
BBB leak in patients suffering from AD showed proteins, penetrate the brain after loss of BBB
a greater increase in BBB permeability and asso- integrity and were shown to accumulate within
ciation with significant reductions in the peri- the brain and trigger specific signaling pathways
cyte populations in the cortex and hippocampus affecting neurovascular functions. For exam-
among AD patients compared with neurologi- ple, serum albumin has been shown to induce
cally healthy human controls. These results were calcium signaling and DNA synthesis in astro-
also confirmed by postmortem brain tissue cytes in culture (Nadal etal., 1995)and in brain
studies (reviewed by Zlokovic, 2013). A human slices (Nadal etal., 1998). In vivo animal studies
apolipoprotein E isoform is particularly shown showed that brain exposure to serum albumin
to mediate neuronal injury in AD by leading to lead to activation of transforming growth fac-
damage of the cerebrovascular integrity and BBB tor beta (TGF-) signalling in astrocytes. The
breakdown via activation of a CypAnuclear transcriptional response in astrocytes is asso-
factor-Bmatrix-metalloproteinase-9 pathway ciated with increased expression of the glial
in pericytes (Bell etal.,2012). acidic fibrillary protein, a hallmark of activated
Some chronic neuropathologies such as mul- astrocytes in brain injury. Activated astrocytes
tiple sclerosis may involve an early phase of BBB downregulate Kir4.1 and connexins (thus lead-
dysfunction (involving the downregulation of ing to accumulation of extracellular K+ upon
claudin 1, 3 (Wolburg and Lippoldt, 2002) that neuronal activation), downregulate EAAT1 and
precedes neuronal damage, suggesting that vas- EAAI12 glutamate transporters (thus leading to
cular damage can lead to secondary neuronal accumulation of extracellular glutamate), and
disorder (see later discussion). In epilepsy, BBB upregulate inflammatory cytokines, including
breakdown was suggested in animal studies to TNF-, IL-6, IL-1, which were shown indepen-
be a direct cause of epileptogenesis. Pathological dently to increase BBB permeability (Heinemann
analysis in resected epileptic tissue indeed con- et al., 2012). Activated astrocytes are thus in a
firms the presence of IgG and serum albumin position to underlie neuronal hyperexcitabil-
within the brain, indicating BBB failure (Raabe ity via disturbance in the normal homeostasis
etal., 2012; van Vliet etal., 2007). Normal pattern of K+, glutamate, and cytokinesall known to
150 Part II:Homeostatic Control

promote neuronal depolarization and plastic- Novartis Found. Symp. 243, 3847; discussion
ity in excitatory synapsesat least partly due 4753, 180185.
to increased activation of N-methyl-d-aspartate Abbott, N.J., Patabendige, A.A.K., Dolman, D.E.M.,
receptors (Bezzi et al., 2001; David et al., 2009; Yusof, S.R., and Begley, D.J. (2010). Structure and
Lapilover et al., 2012). It is thus not surprising function of the blood-brain barrier. Neurobiol.
that when BBB breakdown persists, network Dis. 37,1325.
reorganization occurs, which might eventually Abbott, N.J., Rnnbck, L., and Hansson, E. (2006).
result in epilepsy with recurrent seizures. Apro- Astrocyte-endothelial interactions at the
longed increase in neuronal excitability has been blood-brain barrier. Nat. Rev. Neurosci. 7,4153.
strongly connected to neuronal toxicity and may Amzica, F., Massimini, M., and Manfridi, A. (2002).
point to an important mechanism linking BBB Spatial buffering during slow and paroxysmal
breakdown to neurodegeneration (Tomkins sleep oscillations in cortical networks of glial
et al., 2007). Other serum proteins (in addition cells in vivo. J. Neurosci. 22, 10421053.
to albumin), most notably the blood coagula- Armulik, A., Genov, G., Me, M., Nisancioglu,
M.H., Wallgard, E., Niaudet, C., He, L., Norlin, J.,
tion serine protease thrombin, were similarly
Lindblom, P., Strittmatter, K., et al. (2010).
shown to increase neuronal network excitability
Pericytes regulate the blood-brain barrier.
(Maggio etal., 2008)and to facilitate hippocam-
Nature 468, 557561.
pal long-term potentiation (Han et al., 2011;
Attwell, D., Buchan, A.M., Charpak, S., Lauritzen, M.,
Maggio etal.,2008).
Macvicar, B.A., and Newman, E.A. (2010). Glial
Accumulated evidence from clinical and
and neuronal control of brain blood flow. Nature
animal studies supports the notion that BBB 468, 232243.
dysfunction may serve as a primary signaling Bazzoni, G. (2006). Endothelial tight junctions:
event underlying neurovascular dysfunction. Permeable barriers of the vessel wall. Thromb.
While imaging approaches are being devel- Haemost. 95,3642.
oped to quantitatively measure BBB breakdown Begley, D.J., and Brightman, M.W. (2003). Structural
in patients (Chassidim et al., 2013), it is yet and functional aspects of the blood-brain bar-
necessary to confirm that vascular pathology rier. Prog. Drug Res. 61,3978.
predicts specific neurological syndromes. In Bell, R.D., Winkler, E.A., Sagare, A.P., Singh, I.,
stroke, BBB disturbance was shown to be a sole LaRue, B., Deane, R., and Zlokovic, B.V (2010).
parameter in magnetic resonance imaging risk Pericytes control key neurovascular functions
assessment for hemorrhagic transformation, and neuronal phenotype in the adult brain and
subsequent to thrombolysis in stroke patients during brain aging. Neuron 68, 409427.
(Neumann-Haefelin et al., 2002). Other poten- Bell, R.D., Winkler, E.A., Singh, I., Sagare, A.P.,
tial studies include those involving traumatic Deane, R., Wu, Z., Holtzman, D.M., Betsholtz, C.,
and ischemic injuries and their complications, Armulik, A., Sallstrom, J., et al. (2012).
including epilepsy and cognitive decline patients. Apolipoprotein E controls cerebrovascu-
If such clinical studies confirm a potential role for lar integrity via cyclophilin A. Nature 485,
BBB breakdown in neurological complications, 512516.
novel treatments directly targeting BBB stability Beyreuther, B.K., Freitag, J., Heers, C., Krebsfnger, N.,
and integrity should be developed and tested. Scharfenecker, U., and Sthr, T. (2007).
Lacosamide: A review of preclinical properties.
References CNS Drug Rev. 13,2142.
Abbott, N.J. (2014). Anatomy and physiology of the Bezzi, P., Domercq, M., Brambilla, L., Galli, R.,
bloodbrain barriers. In Drug delivery to the Schols, D., De Clercq, E., Vescovi, A., Bagetta, G.,
brain: Physiological concepts, methodologies Kollias, G., Meldolesi, J., et al. (2001).
and approaches, M. Hammarlund-Udenaes, CXCR4-activated astrocyte glutamate release via
E.C.M. de Lange, and R.G. Thorne, eds. (Springer TNFalpha: Amplification by microglia triggers
New York), pp. 321. neurotoxicity. Nat. Neurosci. 4, 702710.
Abbott, N.J., and Friedman, A. (2012). Overview and Bradbury, M.W., Stubbs, J., Hughes, I.E., and Parker, P.
introduction: The blood-brain barrier in health (1963). The distribution of potassium, sodium,
and disease. Epilepsia 53 (Suppl 6),16. chloride and urea between lumbar cerebrospinal
Abbott, N.J., Khan, E.U., Rollinson, C.M.S., Reichel, fluid and blood serum in human subjects. Clin.
A., Janigro, D., Dombrowski, S.M., Dobbie, Sci. 25, 97105.
M.S., and Begley, D.J. (2002). Drug resistance Carruthers, A., DeZutter, J., Ganguly, A., and
in epilepsy: The role of the blood-brain barrier. Devaskar, S.U. (2009). Will the original
The Blood-Brain Barrier 151

glucose transporter isoform please stand up! Am. Gingrich, M.B., Junge, C.E., Lyuboslavsky, P., and
J.Physiol. Endocrinol. Metab. 297, E836E848. Traynelis, S.F. (2000). Potentiation of NMDA
Chassidim, Y., Veksler, R., Lublinsky, S., Pell, G.S., receptor function by the serine protease throm-
Friedman, A., and Shelef, I. (2013). Quantitative bin. J. Neurosci. 20, 45824595.
imaging assessment of blood-brain barrier per- Goldmann, E. (1913). Vitalfrbung am zentral-
meability in humans. Fluids Barriers CNS10,9. nervensystem. (Berlin: Kniglich-Preuische
Chen, Y., and Liu, L. (2012). Modern methods for Akademie der Wissenschaften).
delivery of drugs across the blood-brain barrier. Greenwood, J., Amos, C.L., Walters, C.E., Couraud,
Adv. Drug Deliv. Rev. 64, 640665. P.-O., Lyck, R., Engelhardt, B., and Adamson, P.
Daneman, R., Agalliu, D., Zhou, L., Kuhnert, F., Kuo, (2003). Intracellular domain of brain endothelial
C.J., and Barres, B.A. (2009). Wnt/beta-catenin intercellular adhesion molecule-1 is essential for
signaling is required for CNS, but not non-CNS, T lymphocyte-mediated signaling and migra-
angiogenesis. Proc. Natl. Acad. Sci. USA 106, tion. J. Immunol. 171, 20992108.
641646. Grontoft, O. (1954). Intracranial haemorrhage and
Daneman, R., Zhou, L., Kebede, A.A., and blood-brain barrier problems in the new-born
Barres, B.A. (2010). Pericytes are required for : A pathologico-anatomical and experimental
blood-brain barrier integrity during embryogen- investigation. Acta Pathol. Microbiol. Scand.
esis. Nature 468, 562566. Suppl. 100,8109.
David, Y., Cacheaux, L.P., Ivens, S., Lapilover, E., Hamilton, N.B., Attwell, D., and Hall, C.N. (2010).
Heinemann, U., Kaufer, D., and Friedman, A. Pericyte-mediated regulation of capillary diam-
(2009). Astrocytic dysfunction in epileptogene- eter:Acomponent of neurovascular coupling in
sis:Consequence of altered potassium and gluta- health and disease. Front. Neuroenergetics2,5.
mate homeostasis? J. Neurosci. 29, 1058810599. Han, K.-S., Mannaioni, G., Hamill, C.E., Lee, J.,
Devaskar, S., Zahm, D.S., Holtzclaw, L., Chundu, K., Junge, C.E., Lee, C.J., and Traynelis, S.F. (2011).
and Wadzinski, B.E. (1991). Developmental Activation of protease activated receptor 1
regulation of the distribution of rat brain increases the excitability of the dentate granule
insulin-insensitive (Glut 1) glucose transporter. neurons of hippocampus. Mol. Brain4,32.
Endocrinology 129, 15301540. Hansen, A.J. (1985). Effect of anoxia on ion distribu-
Dogrukol-Ak, D., Kumar, V.B., Ryerse, J.S., Farr, S.A., tion in the brain. Physiol. Rev. 65, 101148.
Verma, S., Nonaka, N., Nakamachi, T., Ohtaki, H., Hartmann, C., Zozulya, A., Wegener, J., and Galla,
Niehoff, M.L., Edwards, J.C., et al. (2009). H.-J. (2007). The impact of glia-derived extracel-
Isolation of peptide transport system-6 from lular matrices on the barrier function of cerebral
brain endothelial cells: Therapeutic effects endothelial cells:An in vitro study. Exp. Cell Res.
with antisense inhibition in Alzheimer and 313, 13181325.
stroke models. J. Cereb. Blood Flow Metab. 29, Hawkins, B.T., and Davis, T.P. (2005). The
411422. blood-brain barrier/neurovascular unit in health
Dreier, J.P. (2011). The role of spreading depres- and disease. Pharmacol. Rev. 57, 173185.
sion, spreading depolarization and spreading Heinemann, U., and Lux, H.D. (1977). Ceiling of
ischemia in neurological disease. Nat. Med. 17, stimulus induced rises in extracellular potas-
439447. sium concentration in the cerebral cortex of cat.
Dreier, J.P., Jurkat-Rott, K., Petzold, G.C., Tomkins, O., Brain Res. 120, 231249.
Klingebiel, R., Kopp, U.A., Lehmann-Horn, F., Heinemann, U., Kaufer, D., and Friedman, A. (2012).
Friedman, A., and Dichgans, M. (2005). Opening Blood-brain barrier dysfunction, TGF signal-
of the blood-brain barrier preceding corti- ing, and astrocyte dysfunction in epilepsy. Glia
cal edema in a severe attack of FHM type II. 60, 12511257.
Neurology 64, 21452147. Heinemann, U., Schaible, H.G., and Schmidt, R.F.
Ehrlich, P. (1885). Das sauerstufbudurfnis des (1990). Changes in extracellular potassium
organismus: Eine Farbenanalytische Studie. concentration in cat spinal cord in response to
(Berlin:Hirschwald). innocuous and noxious stimulation of legs with
Friedman, A. (2011). Blood-brain barrier dys- healthy and inflamed knee joints. Exp. Brain Res.
function, status epilepticus, seizures, and epi- 79, 283292.
lepsy:Apuzzle of a chicken and egg? Epilepsia 52 Ivens, S., Gabriel, S., Greenberg, G., Friedman, A.,
(Suppl 8),1920. and Shelef, I. (2010). Blood-brain barrier break-
Gingrich, M.B., and Traynelis, S.F. (2000). Serine down as a novel mechanism underlying cere-
proteases and brain damageis there a link? bral hyperperfusion syndrome. J. Neurol. 257,
Trends Neurosci. 23, 399407. 615620.
152 Part II:Homeostatic Control

Ivens, S., Kaufer, D., Flores, L.P., Bechmann, I., of reactivity to afferent stimulation and facilitates
Zumsteg, D., Tomkins, O., Seiffert, E., epileptic seizures in rat hippocampal slices:Toward
Heinemann, U., and Friedman, A. (2007). TGF- understanding the functional consequences of
beta receptor-mediated albumin uptake into cerebrovascular insults. J. Neurosci. 28, 732736.
astrocytes is involved in neocortical epileptogen- Mann, G.E., Yudilevich, D.L., and Sobrevia, L.
esis. Brain 130 (Pt 2), 535547. (2003). Regulation of amino acid and glucose
Jeong, S.M., Hahm, K.D., Shin, J.W., Leem, J.G., Lee, C., transporters in endothelial and smooth muscle
and Han, S.M. (2006). Changes in magnesium cells. Physiol. Rev. 83, 183252.
concentration in the serum and cerebrospinal Marchi, N., Angelov, L., Masaryk, T., Fazio, V.,
fluid of neuropathic rats. Acta Anaesthesiol. Granata, T., Hernandez, N., Hallene, K.,
Scand. 50, 211216. Diglaw, T., Franic, L., Najm, I., et al. (2007).
Jones, H.C., Keep, R.F., and Butt, A.M. (1992). The Seizure-promoting effect of blood-brain barrier
development of ion regulation at the blood-brain disruption. Epilepsia 48, 732742.
barrier. Prog. Brain Res. 91, 123131. Marroni, M., Marchi, N., Cucullo, L., Abbott, N.J.,
Kastin, A.J., and Pan, W. (2003). Peptide transport Signorelli, K., and Janigro, D. (2003). Vascular
across the blood-brain barrier. Prog. Drug Res. and parenchymal mechanisms in multiple drug
61, 79100. resistance:Alesson from human epilepsy. Curr.
Keep, R.F., Ennis, S.R., Beer, M.E., and Betz, A.L. Drug Targets 4, 297304.
(1995). Developmental changes in blood-brain Massimini, M., and Amzica, F. (2001). Extracellular
barrier potassium permeability in the calcium fluctuations and intracellular potentials
rat:Relation to brain growth. J. Physiol. 488 (Pt 2), in the cortex during the slow sleep oscillation. J.
439448. Neurophysiol. 85, 13461350.
Kuschinsky, W., Wahl, M., Bosse, O., and Thurau, K. Medb, J.I., and Sejersted, O.M. (1990). Plasma
(1972). Perivascular potassium and pH as potassium changes with high intensity exercise.
determinants of local pial arterial diameter in J. Physiol. 421, 105122.
cats: A microapplication study. Circ. Res. 31, Michalke, B., and Nischwitz, V. (2010). Review
240247. on metal speciation analysis in cerebrospinal
Lapilover, E.G., Lippmann, K., Salar, S., Maslarova, fluid-current methods and results: A review.
A., Dreier, J.P., Heinemann, U., and Friedman, A. Anal. Chim. Acta 682,2336.
(2012). Peri-infarct blood-brain barrier dysfunc- Moos, T., and Mllgrd, K. (1993). Cerebrovascular
tion facilitates induction of spreading depolar- permeability to azo dyes and plasma proteins
ization associated with epileptiform discharges. in rodents of different ages. Neuropathol. Appl.
Neurobiol. Dis. 48, 495506. Neurobiol. 19, 120127.
Laschinger, M., and Engelhardt, B. (2000). Interaction Nadal, A., Fuentes, E., Pastor, J., and McNaughton,
of alpha4-integrin with VCAM-1 is involved P.A. (1995). Plasma albumin is a potent trig-
in adhesion of encephalitogenic T cell blasts ger of calcium signals and DNA synthesis
to brain endothelium but not in their transen- in astrocytes. Proc. Natl. Acad. Sci. USA 92,
dothelial migration in vitro. J. Neuroimmunol. 14261430.
102,3243. Nadal, A., Sul, J.Y., Valdeolmillos, M., and
Liebner, S., and Plate, K.H. (2010). Differentiation of McNaughton, P.A. (1998). Albumin elicits cal-
the brain vasculature:The answer came blowing cium signals from astrocytes in brain slices from
by the Wnt. J. Angiogenes. Res.2,1. neonatal rat cortex. J. Physiol. 509 (Pt 3), 711716.
Lothman, E.W., and Somjen, G.G. (1975). Nelson, M.T., and Quayle, J.M. (1995). Physiological
Extracellular potassium activity, intracellular roles and properties of potassium channels in
and extracellular potential responses in the spi- arterial smooth muscle. Am. J. Physiol. 268,
nal cord. J. Physiol. 252, 115136. C799C822.
Lux, H.D., Heinemann, U., and Dietzel, I. (1986). Neumann-Haefelin, C., Brinker, G., Uhlenkken, U.,
Ionic changes and alterations in the size of the Pillekamp, F., Hossmann, K.-A., and Hoehn, M.
extracellular space during epileptic activity. Adv. (2002). Prediction of hemorrhagic transforma-
Neurol. 44, 619639. tion after thrombolytic therapy of clot embo-
Lux, H.D., and Singer, W. (1973). Changes of extra- lism: An MRI investigation in rat brain. Stroke
cellular potassium concentration in cat visul 33, 13921398.
cotex after stimulation of specific and unspecific Noebels, J. (2011). A perfect storm:Converging paths
afferents. Pflug Arch. Eur. J.Phy. 343,R137. of epilepsy and Alzheimers dementia intersect in
Maggio, N., Shavit, E., Chapman, J., and Segal, M. the hippocampal formation. Epilepsia 52 (Suppl 1),
(2008). Thrombin induces long-term potentiation 3946.
The Blood-Brain Barrier 153

Oldendorf, W.H., Cornford, M.E., and Brown, W.J. of hypo- and hyperglycemia revisited. J.
(1977). The large apparent work capability of the Neurochem. 72, 238247.
blood-brain barrier: A study of the mitochon- Simpson, I.A., Vannucci, S.J., DeJoseph, M.R., and
drial content of capillary endothelial cells in Hawkins, R.A. (2001). Glucose transporter
brain and other tissues of the rat. Ann. Neurol. 1, asymmetries in the bovine blood-brain barrier.
409417. J. Biol. Chem. 276, 1272512729.
Olsson, Y., Klatzo, I., Sourander, P., and Steinwall, O. Smith, Q.R., and Takasato, Y. (1986). Kinetics of
(1968). Blood-brain barrier to albumin in embry- amino acid transport at the blood-brain barrier
onic new born and adult rats. Acta Neuropathol. studied using an in situ brain perfusion tech-
10, 117122. nique. Ann. NY Acad. Sci. 481, 186201.
Pardridge, W.M. (2005). The blood-brain bar- Stanimirovic, D.B., and Friedman, A. (2012).
rier: Bottleneck in brain drug development. Pathophysiology of the neurovascular
NeuroRx 2,314. unit: Disease cause or consequence? J. Cereb.
Pardridge, W.M. (2012). Drug transport across the Blood Flow Metab. 32, 12071221.
blood-brain barrier. J. Cereb. Blood Flow Metab. Stenman, J.M., Rajagopal, J., Carroll, T.J., Ishibashi, M.,
32, 19591972. McMahon, J., and McMahon, A.P. (2008).
Potschka, H. (2010). Transporter hypothesis of Canonical Wnt signaling regulates organ-specific
drug-resistant epilepsy:Challenges for pharma- assembly and differentiation of CNS vasculature.
cogenetic approaches. Pharmacogenomics 11, Science 322, 12471250.
14271438. Stewart, P.A., and Wiley, M.J. (1981). Developing
Pumain, R., Menini, C., Heinemann, U., Louvel, J., nervous tissue induces formation of blood-brain
and Silva-Barrat, C. (1985). Chemical synaptic barrier characteristics in invading endothelial
transmission is not necessary for epileptic sei- cells: A study using quail-chick transplantation
zures to persist in the baboon Papio papio. Exp. chimeras. Dev. Biol. 84, 183192.
Neurol. 89, 250258. Sykov, E., Czh, G., and Krz, N. (1980). Potassium
Raabe, A., Schmitz, A.K., Pernhorst, K., Grote, A., accumulation in the frog spinal cord induced by
von der Brelie, C., Urbach, H., Friedman, A., nociceptive stimulation of the skin. Neurosci.
Becker, A.J., Elger, C.E., and Niehusmann, P. Lett. 17, 253258.
(2012). Cliniconeuropathologic correlations Tauc, M., Vignon, X., and Bouchaud, C.
show astroglial albumin storage as a common (1984). Evidence for the effectiveness of the
factor in epileptogenic vascular lesions. Epilepsia bloodCSF barrier in the fetal rat choroid plexus.
53, 539548. A freeze-fracture and peroxidase diffusion study.
Rapoport, S.I. (1976). Blood-brain barrier in physiol- Tissue Cell 16,6574.
ogy and medicine (NewYork:Raven). Tomkins, O., Friedman, O., Ivens, S., Reiffurth, C.,
Reese, T.S., and Karnovsky, M.J. (1967). Fine struc- Major, S., Dreier, J.P., Heinemann, U., and
tural localization of a blood-brain barrier to Friedman, A. (2007). Blood-brain barrier disrup-
exogenous peroxidase. J. Cell Biol. 34, 207217. tion results in delayed functional and structural
Saunders, N.R. (1992). Development of the alterations in the rat neocortex. Neurobiol. Dis.
bloodbrain barrier to macromolecules. In 25, 367377.
Barriers and fluids of the eye and brain, M.B. Van Vliet, E.A., da Costa Arajo, S., Redeker, S., van
Segal, ed. (London:Macmillan). Schaik, R., Aronica, E., and Gorter, J.A. (2007).
Saunders, N.R., Knott, G.W., and Dziegielewska, Blood-brain barrier leakage may lead to pro-
K.M. (2000). Barriers in the immature brain. gression of temporal lobe epilepsy. Brain 130,
Cell. Mol. Neurobiol. 20,2940. 521534.
Shepro, D., and Morel, N.M. (1993). Pericyte physiol- Virgintino, D., Errede, M., Robertson, D.,
ogy. FASEB J. 7, 10311038. Capobianco, C., Girolamo, F., Vimercati, A.,
Shulman, R.G., Hyder, F., and Rothman, D.L. (2003). Bertossi, M., and Roncali, L. (2004).
Cerebral metabolism and consciousness. C. Immunolocalization of tight junction pro-
R.Biol. 326, 253273. teins in the adult and developing human brain.
Simard, M., and Nedergaard, M. (2004). The neu- Histochem. Cell Biol. 122,5159.
robiology of glia in the context of water and ion Winkler, M.K.L., Chassidim, Y., Lublinsky, S.,
homeostasis. Neuroscience 129, 877896. Revankar, G.S., Major, S., Kang, E.-J.,
Simpson, I.A., Appel, N.M., Hokari, M., Oki, J., Oliveira-Ferreira, A.I., Woitzik, J., Sandow, N.,
Holman, G.D., Maher, F., Koehler-Stec, Scheel, M., et al. (2012). Impaired neurovas-
E.M., Vannucci, S.J., and Smith, Q.R. (1999). cular coupling to ictal epileptic activity and
Blood-brain barrier glucose transporter:Effects spreading depolarization in a patient with
154 Part II:Homeostatic Control

subarachnoid hemorrhage: Possible link to blood-brain barrier:An engineering perspective.


blood-brain barrier dysfunction. Epilepsia Front. Neuroeng.6,7.
53(Suppl 6),2230. Zhang, E.Y., Knipp, G.T., Ekins, S., and Swaan, P.W.
Wolburg, H., and Lippoldt, A. (2002). Tight junctions (2002). Structural biology and function of sol-
of the blood-brain barrier: Development, com- ute transporters: Implications for identifying and
position and regulation. Vascul. Pharmacol. 38, designing substrates. Drug Metab. Rev. 34, 709750.
323337. Zlokovic, B. V (2013). Cerebrovascular effects of
Wong, A.D., Ye, M., Levy, A.F., Rothstein, J.D., apolipoprotein E: Implications for Alzheimer
Bergles, D.E., and Searson, P.C. (2013). The disease. JAMA Neurol. 70, 440444.
10
Inflammation and Immunomodulation
inEpilepsy and Its Comorbidities
A N D R E Y M A Z A R AT I A N D A N N A M A R I A V E Z Z A N I

P R E FA C E the identification of glia as main source of these


Inflammation is a prototypical response to injury molecules, highlights putative novel targets for
or infection. This homeostatic mechanism is acti- therapeutic interventions beyond classical anti-
vated in the challenged tissue in order to reestab- convulsive drugs that modulate neuronal chan-
lish homeostasis and initiate repair; however, nels and classical neurotransmitters.
inflammation can also promote tissue injury if
prompt resolution of the inflammatory cascade INTRODUCTION
is not attained in a timely and efficient manner. Inflammation is a prototypical response to infec-
Increasing evidence provides support to a patho- tion or tissue injury, and, as such, it represents
genic role of unabated brain inflammation in a key homeostatic endogenous mechanism
various central nervous system (CNS) diseases, activated for pathogen killing and removal,
including epilepsy. The induction of inflamma- thereafter promoting tissue healing and repair.
tory mediators after epileptogenic brain injuries It is now well established that the evolution
or seizures chiefly involves brain resident micro- of inflammation programs in tissue requires
glia and astrocytes but also extends to neurons strict control and fast resolution by endogenous
and microvessels and promotes later leukocytes anti-inflammatory molecules in order to avoid
recruitment. The involvement of glial cells in detrimental effects of inflammation on tissue
the generation of the inflammatory cascade physiology. Indeed, there are ample examples
in epilepsy highlights their role as key players in the literature showing that if endogenous
in innate immunity in the brain. Notably, glial anti-inflammatory control fails or is inefficient,
cells also contribute to normal brain develop- then inflammation results in the cell dysfunc-
ment and function via their physical and func- tion or death. Uncontrolled inflammation has
tional interactions with surrounding neurons, been suggested to play a pathogenic role in sev-
and the release of inflammatory mediators plays eral brain diseases, as well as in acute and chronic
a crucial role in many of their physiological neurodegeneration (Craft etal., 2005; Minghetti,
properties. These cells are, therefore, placed at 2005; Glass etal., 2010). In the frame of the sug-
the intersection between health and disease, thus gested role of inflammation in CNS pathology,
representing a notable example of how runaway increasing evidence in past 15 years has proven
homeostatic and physiological mechanisms may a reciprocal causal link between brain inflamma-
lead to pathologic and harmful sequelae. In this tion and epilepsy (Vezzani etal., 2011a). In this
chapter we describe clinical and experimental setting, inflammation represents a phenomenon
evidence supporting the role of brain inflam- associated with the innate immune response of
mation in pathogenic mechanisms of seizures, the brain exposed to epileptogenic injuries or sei-
cell loss, and comorbidities of epilepsy. We also zures. Inflammation chiefly involves microglia
describe the novel and increasingly recognized and astrocytes and is best defined as a process
neuromodulatory role of inflammatory mol- comprising of rapid injury-mediated release of
ecules and mechanisms underlying their effects cytokines and chemokines, which in turn acti-
on neuronal (hyper)excitability. The identifi- vate downstream inflammatory mediators and
cation of the pathogenic role of inflammatory related molecular processes in glia (i.e., autocrine
mediators in epilepsy and its comorbidities, and action), as well as in neurons and endothelial
156 Part II:Homeostatic Control

cells of the blood-brain barrier (BBB; i.e., parac- (infantile spasms, Lennox-Gastaut,
rine action). The induction of innate immunity Landau-Kleffner, Dravet, fever-induced
may subsequently activate adaptive immunity refractory epileptic encephalopathy in
mechanisms and the consequent leukocytes school age children).
extravasation in the brain parenchyma, a pro-
cess requiring an active role of the BBB (Vezzani Immunohistochemical evidence has shown
etal., 2011a; Vezzani and Friedman,2011). that activated microglia and astrocytes provide
Although the presence of inflammatory a common source for the synthesis and release of
molecules in the epileptic brain has been estab- inflammatory mediators across epilepsies with dif-
lished since the first characterization in 1958 ferent etiologies, whereas evidence for the activa-
of Rasmussens encephalitis (RE) as a chronic tion of adaptive immunity appears to depend on a
inflammatory disease (reviewed by Varadkar particular etiology. For example, limited or scarse
et al., 2014) the relevance of inflammation in infiltration of T cells has been reported in mTLE
epilepsy has been further expanded due to novel and FCD type 1 as compared to both FCD type 2
discoveries that (a)brain inflammation, as previ- (Iyer etal., 2010)and RE (Bien etal., 2002; Pardo
ously defined, is a common substrate of various et al., 2004), and circulating autoantibodies have
drug-resistant forms of epilepsy with differing been detected only in certain forms of systemic
etiologies not necessarily linked to autoimmune or neurological autoimmune disorders, and only
dysfunctions or active infections, and (b)inflam- rarely in idiopathic epilepsies (Bien and Scheffer,
matory mediators are indeed neuromodulators 2011; Vincent and Crino, 2011; Vincent etal.,2011).
endowed of CNS-specific roles (Viviani et al., Neurons, and baloon cells in FCD, also stain
2007; Vezzani etal., 2011b); in particular, they can for inflammatory molecules, as well as endothe-
directly affect neuronal activity independently of lial cells of the BBB (Ravizza et al., 2006a;
their classical immune role in response to infec- Aronica and Crino, 2011). Notably, BBB damage
tion or systemic inflammatory challenges. is often observed in perivascular areas in associa-
tion with the increased presence of inflammatory
EVIDENCE LINKING BR AIN mediators in perivascular astrocytes and their
I N F L A M M AT I O N T O E P I L E P S Y endfeet and in the perivascular activated micro-
INTHE CLINICAL SETTING glia. Finally, the extent of microglia activation
There are three main sets of evidence establish- and the level of expression of inflammatory mol-
ing a link between brain inflammation and such ecules in brain cells positively correlate with sei-
hallmarks of epilepsy as the onset and the recur- zure frequency and epilepsy duration at the time
rence of seizures, tissue neuropathology, and of surgery (Boer etal., 2006; Ravizza etal., 2006a;
comorbidities (Vezzani et al., 2011a; Aronica Pernhorst etal., 2013). These observations dem-
etal., 2012; Vezzani etal.,2013): onstrate the activation of inflammatory signaling
in human epileptic tissue; moreover, therapeu-
Various inflammatory mediators (e.g., tic effects of anti-inflammatory treatments in
cytokines, chemokines, prostaglandins, drug-resistant epilepsies highlight a possibility
complement factors) are overexpressed in of a causal relationship between brain inflam-
surgically resected epileptogenic foci in mation and the pathogenesis of epilepsy. Such
different forms of pharmacoresistant epi- a causal relationship has been established using
lepsy (e.g., tuberous sclerosis, glioneuronal experimental models of human disease.
tumors, focal cortical dysplasia [FCD],
mesial temporal lobe epilesy [mTLE],RE). EVIDENCE OFA CAUSAL
Activation of innate (i.e., microglia and R E L AT I O N S H I P B E T W E E N
astrocytes) and adaptive (i.e., extravasation of B R A I N I N F L A M M AT I O N
leukocytes, autoantibodies) immune mecha- A N D T H E PAT H O G E N E S I S
nisms in differing forms of human epilepsy. OFSEIZURES
Drugs and treatments with
anti-inflammatory properties (e.g., Innate Immunity and
adrenocorticotropic hormone, steroids, Inflammation:Focus on
intravenous immunoglobulins, plasma IL-1R1/TLR4 Signaling
exchange) show therapeutic effects in oth- Activation of innate immune mechanisms in
erwise drug-resistant pediatric syndromes microglia and astrocytes has a pivotal role in the
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 157

generation of the inflammatory cascade follow- in epilepsy brain tissue. Evidence of signaling
ing brain injuries or seizures via transcriptional activation has been reported both in astrocytes
activation of NFkB-dependent genes. Activation and in neurons in human epilepsy (e.g., malfor-
of the same type of signaling in neurons has dif- mations of cortical development, mTLE, and RE)
ferent physiopathologic outcomes that are medi- and in animal epilepsy models, for example those
ated by rapid posttraslational modifications of of mTLE, absence seizures in genetic absence epi-
voltage-gated or receptor-coupled ion channels lepsy rats from Strasbourg (GAERS) (serving as
and the consequent reduction of seizure thresh- a model of absence epilepsy in humans), as well
old (Table 10.1 and reviewed in Vezzani et al., as in kindling (i.e., occurence and progression
2011b). of motor seizures in response to repetitive sub-
Recent data have demonstrated that epilepsy threshold electical stimulation of limbic system;
is associated with the induction of the IL-1 recep- Vezzani etal., 2011b; Aronica etal.,2012).
tor type 1 (IL-1R1) and of Toll-like receptor 4 The role played by these signaling molecules
(TLR4) signaling in glia and neurons (Ravizza in seizure mechanisms (Table 10.1) has been
et al., 2006a; Boer et al., 2008; Ravizza et al., highlighted by pharmacological and genetic
2008a; Maroso etal., 2010; Zurolo etal., 2011). The studies. For example, blockade of the inflamma-
respective endogenous cytokines, IL-1 and high some (i.e., the macromolecular complex respon-
mobility group box 1 protein (HMGB1) that acti- sible for the IL-1 and HMGB1 release) using
vate their cognate receptors are also upregulated antagonists of P2X7 receptors that are activated

TABLE10.1 E X A M PL E S OFI N FL A M M ATORY SIGNA LI NG CASCA DE S AC T I VATED


I NTH E BR A I N DU R I NG SEI Z U R E S OR BYEPIL EP TOGEN IC I N J U R I E S
I NE X PER I M EN TA LMODEL S
Inflammatory Endogenous Intracellular Target Outcomes
pathway activators signaling (cell types)

IL-R1/TLR4 IL-1,HMGB1 Srckinase NR2B Seizures, cellloss


n.d. (neurons) Depression, Cognitive
NFkB HPA, 5-HT (neurons) deficits
Genes Inflammation,
(glia,BBB) BBB damage
RAGE HMGB1,S100 NFkB Glia Seizures, Inflammation
n.d. n.d. Cognitive deficits
P2X7 R ATP Inflammasome Glia Seizures,cell loss
COX-2/PGE2 Glutamate, cAMP/PKA/CREB EP2 Neuronal survival
Cytokines n.d. (neurons) Cognitive deficits
cAMP/Epac n.d. Inflammation, cell loss
EP2
(glia)

TNF-type1 TNF- PI3KAkt GluR2,AMPA GABA-A Seizures, cellloss


TNF- type 2 NFkB (neurons) Inflammation
n.d. n.d. Neuroprotection,
(glia) reduced seizures
n.d.a

Note: This table reports selected examples of inflammatory mechanisms activated in epilepsy that have been shown to play a role in
physiopathological outcomes, as demonstrated in experimental models. For additional information, see the reference list in the main
text and the latter sections of the article.
n.d.=not determined; NFkB, nuclear factor KB; NR2B=NMDA receptor subtype 2B; NMDA=N-methyl-D-aspartate;
BBB=blood-brain barrier; RAGE=receptor for advanced glycation end products; Epac=exchange protein directly activated by cAMP;
PKA, protein kinase A; CREB= cAMP response element-binding protein; PI3K=phosphoinositide 3-kinase; Akt=protein kinase B;
TNF=tumor necrosis factor; AMPA=-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; gamma-aminobutyric acid=GABA.
a
indirect data in receptor knockout mice suggest that NMDA, AMPA, and kainate receptor subtypes are downregulated by receptor
activation (Balosso etal.,2009).
158 Part II:Homeostatic Control

by ATP released by damaged cells (Engel et al., bacteria, which induces inflammation by acti-
2012), or caspase-1 antagonists that inhibit IL-1 vating TLR4. Acquired channelopathies have
biosynthesis, both drastically reduce the fre- been reported to be induced also by tumor
quency of seizures in rodent models of temporal necrosis-alpha (TNF-) factor affecting the
lobe epilepsy (TLE), decrease absence seizures in assembly and synaptic clustering of -amino-
GAERS (Ravizza etal., 2006b; Akin etal., 2011; 3-hydroxy-5-methyl-4-isoxazolepropionic acid
Maroso et al., 2011) and inhibit seizure propa- (AMPA) receptors and the membrane expression
gation during kindling (Ravizza et al., 2008b). of gamma-aminobutyric acid (GABA)-A recep-
Injection of IL-1R1 or TLR4 antagonists also tors. In particular, TNF- induces the membrane
alleviates experimental seizures in a variety of shift toward the extrasynaptic compartment of
models (Vezzani etal., 1999; Vezzani etal., 2000; GLUR2-lacking AMPA receptors, a mechanism
Vezzani et al., 2002; Marchi et al., 2009; Auvin involved in excitotoxicity and possibly in synaptic
etal., 2010; Maroso etal., 2010; Kwon etal., 2013). scaling (Beattie etal., 2002; Stellwagen etal., 2005;
The described anticonvulsive effects are congru- Stellwagen and Malenka, 2006). Furthermore,
ent with the proconvulsive activity of either cytokine-induced activation of intracellular
IL-1 or HMGB1 upon their intracerebral injec- kinases changes the phosphorylation and func-
tion before the convulsive challenge (Vezzani tion of voltage-gated Na+, K+, and Ca2+ channels in
et al., 1999; Vezzani et al., 2000; Balosso et al., neurons (Vezzani and Viviani,2015).
2008; Maroso etal., 2010; Iori etal., 2013; Balosso Additional mechanisms possibly underlying
et al., 2014). Accordingly, transgenic mice with the proictogenic properties of cytokines include
perturbed IL-1 or HMGB1 signaling show sig- their ability to inhibit glutamate reuptake by astro-
nificant alterations in their intrinsic seizure sus- cytes, to promote glutamate release from glia, and
ceptibility (Vezzani et al., 2008; Maroso et al., to enhance NMDA-mediated glutamate release
2010; Iori etal.,2013). from synaptic terminals, since all these effects pro-
Interestingly, recent data have shown that mote neuronal excitability (Devinsky etal.,2013).
proconvulsive activity of HMGB1 is specifically
linked to the redox state of its cysteine residues I N F L A M M AT O R Y M O L E C U L E S
after the molecule is released extracellularly. AND EPILEPTOGENESIS
Thus only the disulfide (i.e., oxidized) form of Mounting evidence suggests pathogenic role of
this molecule activates TLR4 and promotes sei- specific inflammatory signaling in the develop-
zures but not the all-thiol (i.e., reduced) form, ment of epilepsy after a primary brain injury.
which has instead chemoattractant properties. This information is emerging from studies
This finding highlights the importance of the using epilepsy models, where pharmacologi-
redox state of the extracellular milieu for medi- cal blockade of a specific target is attained at
ating the physiopathologic effects of HMGB1 on various times after the epileptogenic injury
neurons (Balosso etal.,2014). (usually after status epilepticus onset) but
One key question concerns mechanisms before epilepsy develops (Ravizza et al., 2011).
by which the activation of the IL-1R1/TLR4 For example, individual targeting of IL-1 and
signaling promotes seizures. Importantly, this cyclooxygenase 2 (COX-2) systems, or their
signaling induces rapid (i.e., within minutes) combination, affords neuroprotection (Kwon
Src kinasemediated phosphorylation of the etal., 2013; No etal., 2013)and in some com-
NR2B subunit of the N-methyl-D-aspartate binatorial experiments also reduces the fre-
(NMDA) receptor complex thus leading to the quency of spontaneous seizures (Kwon et al.,
increased neuronal Ca2+ influx (Viviani et al., 2013). Disease-modifying effects have been
2003; Balosso et al., 2008). This molecular event reported also by pharmacological interference
underlies the proconvulsive activity of both IL-1 with microglia or astrocyte activation (Ravizza
and HMGB1, as well as their effect in enhanc- et al., 2011; Wang et al., 2015). One study
ing glutamate-mediated neurotoxicity. Another reported antiepileptogenic effects achieved
mechanism of hyperexcitability mediated by by pharmacological blockade or genetic inac-
IL-1R1/TLR4 signaling is the downregulation tivation of adhesion molecules on brain ves-
of the HCN1 channel and the associated Ih cur- sels, thus impairing leukocyte extravasation
rent on dendrites of hippocampal pyramidal (Fabene et al., 2008). This study, however, has
neurons, as assessed in rats upon intraventricu- been challenged by subsequent evidence show-
lar lipopolysaccharide (LPS) injection (unpub- ing worsening effects on seizure onset and neu-
lished data), a component of Gram-negative ropathology when leukocyte extravasation was
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 159

prevented (Zattoni et al., 2011). Accordingly, One notable example of the dual role of inflam-
adoptive transfer of T lymphocytes in immu- matory molecules in the brain is represented by
nodeficient mice delayed seizures onset and the COX-2 induction (Table 10.1). This enzyme
mediated neuroprotection (Deprez etal.,2013). is activated in epilepsy models, first in neurons
The absence of either TLR4 or receptor for and subsequently in glial cells with a prominent
advance glycation end products (RAGE, a recep- induction in astrocytes (Vezzani et al., 2012).
tor for the HMGB1 and S100 protein family) in Genetic inactivation of COX-2 in pyramidal neu-
knockout mice did not interfere with the acute rons rescues cell loss when mice are exposed to
phase of status epilepticus but mediated a signifi- status epilepticus; accordingly neuroprotection is
cant reduction in spontaneous seizure frequency attained by administering antagonists of prosta-
in epileptic mice (Iori etal.,2013). glandin (PG) E2 EP2 receptor from 1 hour up to 3
This set of evidence highlights the need for hours from status epilepticus onset. Interstingly,
better understanding master regulators of the activation of EP2 receptors at the time of status
inflammatory cascade that dynamically evolves epilepticus induction is indeed neuroprotective,
during epileptogenesis and of discerning mech- thus indicating the dual role of PGE2/EP2 sig-
anisms with an homeostatic role from those nalig in determining seizure-induced cell loss
contributing to the disease progression. This or survival. Activation of cAMP/PKA/CREB in
information is instrumental for developing effec- neurons or cAMP/Epac proinflammatory signal-
tive anti-inflammatory treatments, which can be ing in glia appear to be the key determinant for
timely applied for arresting epileptogenesis after mediating cell survival or cell loss, respectively
a primary injury without interfering with the (Jiang et al., 2012). Different effects on seizures
beneficial role of inflammation. have also been reported depending on the type of
PG induced in a particular experimental model
T H E Y I N A N D YA N G O F T H E and the timing of anti-COX-2 intervention
I N F L A M M AT O R Y C A S C A D E (Kulkarni and Dhir, 2009; Vezzani etal.,2012).
The inflammatory cascade activated by IL-1R1/ TNF- is another inflammatory media-
TLR4 signaling leads to synthesis and release of tor that plays a dual beneficial or detrimental
various inflammatory mediators, which in turn role. Depending on the type of receptors pref-
may contribute either to pathology or to home- erentially activated in the specific pathologic
ostasis and repair. This yin and yang feature of contex, this cytokine mediates anticonvulsive
inflammation is well known, and many exam- effects by activating TNF- type 2 receptors or
ples are reported in the literature. In general, proconvulsive effects by activating TNF- type
the physiopathologic outcomes of inflamma- 1 receptor (Balosso et al., 2005; Balosso et al.,
tion are strictly context dependent and as such 2013; Weinberg etal., 2013). The extent of TNF-
are chiefly determined by the extent of changes increase in the brain also matters, whereby mod-
in the levels of specific inflammatory molecules erate increases appear to mediate inhibitory
and time of tissue exposure to them, as well as by effects on seizures while massive increases exac-
the type and cellular pattern of their respective erbate seizures (Akassoglou etal., 1997; Balosso
receptor expression. The microenvironment in etal.,2005).
which tissue inflammation develops, therefore,
is instrumental for defining the outcomes. The THE ROLE OF
time of onset and development of the inflamma- A DAPTIV E IM MU NIT Y
tory response after the primary injury or tissue
challenge is also crucial. A typical example is Human Evidence
represented by LPS-induced tolerance (Biswas The adaptive immune system can play a role in
and Lopez-Collazo, 2009), a phenomenon that epilepsy by activating autoantibody-mediated
protects brain tissue from subsequent ischemic mechanisms or by T cellmediated cytotoxicity.
damage or seizures (Dmowska etal., 2010), con- Invasion of immunoglobulins (Ig) and T lym-
trasted by the LPS-mediated increase in neu- phocytes in the brain can be found in a variety
ronal cell loss (Auvin etal., 2007)and reduction of encephalitis including RE, viral encephalitis
of seizure threshold (Sayyah et al., 2003; Riazi and a group of antibody-associated encepha-
et al., 2010). Such opposite outcomes strictly litis defined by the presence of specific neural
depend on precise timing before the induction autoantibodies in serum and/or CSF. In RE and
of tissue inflammation and the onset of brain other epileptic disorders, a wide spectrum of
insult. anti-neural antibodies has been detected which
160 Part II:Homeostatic Control

may result from a secondary immune response of the NMDAR (Dalmau etal., 2008). Clinically,
against antigens released during neuronal cell a prodromal stage with neurovegetative symp-
degeneration. A role of cytotoxic CD8+T lym- toms precedes the development of seizures,
phocytes has been described in RE; in particu- memory loss and psychiatric symptoms followed
lar, granzyme B positive cytotoxic T cells can be by a dramatic movement disorder (Dalmau etal.,
found in close apposition to neurons and astro- 2011). Prolonged courses of immunotherapy
cytes with polarization of the cytotoxic gran- may reverse these clinical signs. Brains of these
ules facing the targeted cell membrane (Bien patients show few inflammatory T cells and in
et al., 2002; Bauer et al., 2007). MRI studies most cases neuronal loss is mild (Dalmau etal.,
have found that infiltrating T cells in RE occur 2007; Bien etal., 2012). IgG binding to hippocam-
also outside of the epileptic network, suggesting pal neurons may lead to a state of NMDAR hypo-
that epileptic activity may not be directly associ- function (Hughes etal.,2010).
ated with the presence of infiltrating cells (Hauf Although autoantibodies are unlikely to be a
et al., 2009). Accordingly, an immunosuppres- major cause of seizures in idiopathic epilepsies,
sive drug tacrolimus had ameliorating effects on an increasing number of studies have detected
motor and cognitive dysfunctions and neuronal serum autoantibodies in 2% to 7% of these
cell degeneration in RE patients, but showed no patients (McKnight et al., 2005; Majoie et al.,
effect on seizure frequency (Bien etal., 2004; Bien 2006). Immunotherapies offered occasional ther-
etal.,2013). apeutic benefit (Giometto etal., 1998), although
The antibody-associated encephalitides are their overall efficacy is often disappointing.
characterized by a prominent seizure pheno-
type often involving limbic system (i.e., limbic E X P E R I M E N TA L S T U D I E S
encephalitis). Clinical evidence and emerging Pathological relevance of autoantibodies or
in vitro data give support to the pathogenicity of leuckocytes in epilepsy has been addressed in
some of these antibodies. They can be directed experimental in vitro and in vivo studies. Notable
against either intracellular (e.g. GAD65, amphi- examples are reportedhere.
physin, Hu, Yo and Ma2) or membrane (e.g.
the voltage-gated potassium channels (VGKC), Antibodies Against
NMDA, AMPA, or metabotropic glutamate Membrane Antigens
receptor subunits and the GABAB receptor) Infusion of NMDAR antibodies into the rodent
antigens. Negative passive transfer experiments brain caused a downregulation of NMDAR;
suggest that antibodies targeting intracellular accordingly, incubation of hippocampal neuron
antigens are not pathogenic since they cannot cultures with purified NR1-IgG from patients
access the intracellular antigens in intact neurons with NMDAR encephalitis caused a significant
(Vincent et al., 1999). Differently, antibody tar- reduction in NMDAR density and in both sur-
geting proteins associated to the VGKC-complex face and total NR2A/B subunits (Hughes et al.,
(i.e. Caspr2, LGI1, contactin-2) (Irani etal., 2010; 2010), resulting in a reduced synaptic NMDAR
Lai etal., 2010; Vincent and Irani, 2010)are sug- localization, which was reversed upon removal
gested to mediate memory loss, movement disor- of the antibodies. Applying patients serum anti-
ders and seizures (Thieben etal., 2004; Vincent bodies against AMPA receptors in hippocampal
etal., 2004; Chan etal., 2007). Importantly, ster- neurons produced similar results, along with the
oids, intravenous immunoglobulin G (IgG), and significant reduction in the density of membrane
plasmapheresis improve the described neuro- AMPA receptors (Lai etal., 2009). NMDA recep-
logical deficits, thus suggesting that these anti- tor antibodies also increased the concentration of
bodies are directly pathogenic (Vincent et al., glutamate in the extracellular space (Manto etal.,
2004). The presence of circulating and brain 2010), and whole-cell patch-clamp recordings of
tissue VGKC-complex antibodies is associated miniature excitatory post-synaptic currents in
with neuronal degeneration in the hippocam- cultured rat hippocampal neurons showed that
pus in the presence of C9neo, the end complex patients antibodies specifically decreased synap-
of complement activation (Bien etal., 2012). This tic NMDAR-mediated currents, without affect-
suggests that an antibody-mediated complement ing AMPA receptor-mediated currents (Hughes
activation may mediate neuronal celldeath. et al., 2010). Lalic et al. (2010) using whole-cell
Another form of encephalitis is associated patch-clamp recordings showed synaptic stimu-
with antibodies directed against the NR1 subunit lation of CA3 neurons in hippocampal slices
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 161

incubated with IgG from limbic encephalitis (e.g., formylated peptides released from damaged
patients, as well as epileptiform activity and mitochondria), then they may activate adaptive
increased mossy fiber-evoked synaptic responses. immunity. One crucial, still open question is
These results were mimicked by -dendrotoxin, whether this phenomenon has some relevance
an antagonist of the Shaker VGKC channel, sug- for tissue hyperexcitability or neuropathology.
gesting that the antibodies increase CA3 neurons Two divergent findings emerged from mouse
excitability by downregulating the activity of the models of TLE. In the pilocarpine model in mice,
VGKC complex. neurotrophils, macrophages, and T cells may
play a detrimental role in the postinjury phase
Antibodies Against Intracellular (i.e., epileptogenesis) since knockout mice lack-
Antigens ing key adhesion molecules or wild-type mice
Although the cytoplasmic location of GAD treated with anti-integrins antibodies (i.e., two
(i.e. the enzyme responsible for the synthesis of procedures that impair leukocytes extravasa-
GABA) suggests that the related antibodies are tion) develop less frequent spontaneous seizures
unlikely to be pathogenic, the application of and show less cell loss as compared to wild-type
GAD-positive sera from epilepsy patients onto epileptic mice (Fabene et al., 2008). Conversely,
cultured hippocampal neurons increases the fre- when epilepsy is triggered by intracerebral kainic
quency of the postsynaptic inhibitory potentials. acid in mice, macrophages and T cells play a pro-
This evidence supports the hypothesis of patho- tective role by delaying the onset and reducing
logically active components in the patients sera the recurrence of spontaneous seizures (Zattoni
(Vianello etal.,2008). et al., 2011); prevention of neurotrophils entry
into the brain tissue appears to be one mecha-
Role ofLeukocytes inSeizureModels nism of neuroprotection. For a correct interpre-
There is evidence for the involvement of the tation of these results, it is important to consider
peripheral immune cells in some forms of epi- that pilocarpine uses a peculiar mechanism of
lepsy. It appears that their contribution to mech- action to induce epilepsy: it directly activates
anisms of the disease differs depending on the muscarinic receptors on circulating leukocytes,
nature of the epileptogenic trigger. In particular, which in turn promote BBB leakage, thus allow-
perivascular extravasation of CD4+ and CD8+ ing enough pilocarpine to enter the brain to
T lymphocytes has been reported in the mouse induce seizures (Vezzani and Janigro,2009).
hippocampus after prolonged seizures induced in
mice by either systemic injection of convulsant I N F L A M M AT I O N A N D
doses of the muscaric receptor agonist pilocar- COMORBIDITIES
pine (Fabene etal., 2008)or by intrahippocam- Many epilepsy patients present with inter-
pal administration of kainic acid (Zattoni etal., ictal neurobehavioral disorders at a signifi-
2011). This phenomenon may reflect cell extrava- cantly higher rate than the general population.
sation due to injury or seizure-induced leakage of Neurobehavioral comorbidities of epilepsy have
BBB. Alternatively, lymphocytes may be primed been receiving growing attention. Indeed, with
to pass into the perivascular space by vessel significant progress made in the management
inflammation following cell injury or seizures. of seizures proper, it is becoming increasingly
Upregulation of adhesion molecules on endothe- obvious that effective seizure control is not nec-
lial cells of brain microvessels and chemoattrac- essarily accompanied by the alleviation of con-
tion is induced by cytokines and chemokines current neuropsychiatric disorders. As a result,
released from perivascular astrocytes and comorbidities may have higher negative impact
microglia (Librizzi et al., 2007; Librizzi et al., on patients quality of life than even frequency
2012), thus favoring luminal interactions and of seizures (Gilliam etal., 1997; Garcia-Morales
extravasation of circulating leukocytes (Fabene et al., 2008). The importance of addressing
et al., 2008). Although the presence of circulat- comorbidities of epilepsy has been recognized by
ing antigens activating T cells has not been dem- the National Institute of Neurological Disorders
onstrated, one might envisage that brain-born and Stroke, which included them in its Epilepsy
molecules released by damaged or activated Research Benchmarks in 2007 and again in
neurons and/or glial cells may drop out of the 2013 (National Institutes of Health, National
CSF into the systemic circulation. If these mol- Institute of Neurological Disorders and Stroke,
ecules express molecular mimicry of pathogens 2007,2013).
162 Part II:Homeostatic Control

A dominating school of thought regarding Brain IL-1 has been regarded as a key fac-
causes of a high rate of neurobehavioral per- tor in the inflammation-associated depression
turbations among epilepsy patients is that epi- (Dunn et al., 2005; Goshen and Yirmiya, 2009;
lepsy and concurrent nonconvulsive conditions Lang and Borgwardt, 2013). For example, cen-
share certain pathophysiological mechanisms. trally administered IL-1 induced behavioral
Considering that brain inflammation has been (Dunn and Swiergiel, 2005)and neuroendocrine
implicated in mechanisms of several neuropsy- (Parsadaniantz etal., 1997)symptoms of depres-
chiatric disorders, it is conceivable that inflam- sion in rodents. Therefore, the established chronic
matory processes that are triggered in the brain overexpression of IL-1 and its receptor in the
by an epileptogenic insult may, concurrently epileptic hippocampus (Ravizza et al., 2008a),
with seizures, lead to the development of neu- aside from contributing to mechanisms of epi-
robehavioral abnormalities. We discuss the role lepsy proper, may lead to depression. Outside the
of inflammation in the pathophysiology of three hippocampus, IL-1 is suppresses the firing of
common comorbidities of epilepsy: depression, raphe serotonin neurons (Brambilla etal., 2007),
autism, and cognitive impairments. thus conceivably leading to depression via sub-
sequent reduced activity in serotonergic afferents
Depression (Figure10.1).
Between 10% and 60% of epilepsy patients pre- Animals with chronic epilepsy, which is pre-
sent with symptoms of depression (Mendez cipitated by either status epilepticus or kindling,
et al., 1986; Kanner et al., 2012). The comor- exhibit behavioral deficits indicative of depres-
bidity has been examined along the lines sion, particularly anhedonia and failure to cope
of pathophysiological mechanisms shared with stress (Koh et al., 2007; Mazarati et al.,
by epilepsy and major depressive disorder 2007; Mazarati et al., 2008). These impairments
(MDD). For example, the dysfunction of cen- develop as a result of consecutive hyperactiv-
tral monoamine neurotransmitters serotonin ity of the HPA axis, upregulation of 5-HT1A
and norepinephrine, and the hyperactivity of autoreceptors, and subsequently diminished
hypothalamo-pituitary-adrenocortical (HPA) raphe-hippocampal serotonergic tone (Mazarati
axis have been regarded as key mechanisms of et al., 2008; Mazarati et al., 2009; Pineda et al.,
MDD (Baumann etal., 1999; Ordway etal., 2003; 2011). These perturbations are not amenable to
Carroll etal., 2007; Lowry etal., 2008; Michelsen fluoxetine therapy (Pineda etal., 2012), thus rep-
etal., 2008)and at the same time have been docu- resenting a case of selective serotonin reuptake
mented in patients with various forms of epilepsy inhibitor (SSRI)-resistant depression (discussed
(Kondziella etal., 2007; Kanner etal.,2012). further next). At the same time, direct intra-
The connection between chronic inflammation hippocampal administration of interleukin-1
and MDD is well established. Thus biomarkers of receptor antagonist (IL-1ra), an endogenous com-
inflammation have been consistently found in the petitive inhibitor peptide of IL-1 receptor type 1,
blood of depression patients (Bremmer etal., 2008; alleviated (albeit not fully reversed) depression
Muller et al., 2011; Krishnadas and Cavanagh, symptoms, including behavioral, neuroendo-
2012). Furthermore, patients with primarily crine, biochemical, and receptor impairments
chronic inflammatory diseases (e.g., rheumatoid (Mazarati etal., 2010). This suggests that depres-
arthritis) develop depression at a significantly sion in epileptic subjects may, at least in part,
higher rate than general population (40% vs. 5% stem from the overexpression of hippocampal
to 15%; Bruce, 2008). Chemokine immunother- IL-1 and/or its receptor, with the subsequent
apy is known to induce symptoms of depression involvement of the HPA axis and brain seroton-
(Dantzer and Kelley, 1989). In an experimental ergic transmission (Figure10.1).
setting, Gram-negative infection mimicked in In addition to precipitating symptoms of
rodents by LPS produces sickness behavior, which depression, the enhanced IL-1 signaling in the
includes such symptoms of depression as anhedo- epileptic hippocampus may also play role in the
nia and failure to cope with stress (Dantzer, 2006). SSRI resistance of epilepsy-associated depres-
Moreover, even depressive impairments not pri- sion. SSRI resistance represents significant chal-
marily associated with inflammatory stimuli (e.g., lenge in MDD management, as between 30%
depression induced by stress) are amenable to the and 50% of depression patients do not respond
treatment with anti-inflammatory drugs, such as or poorly respond to this class of antidepressants
minocycline and celecoxib (Molina-Hernandez (Barbui etal., 2002; Rush etal., 2006). It has been
etal., 2008; Guo etal.,2009). suggested that SSRI resistance is present in MDD
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 163

Normal
Proinflammatory event
Depression cases of SSRI-resistant MDD that do not stem
IL-1 (a) IL-1 from the 5-HT1A receptor gene polymorphism
Epilepsy (b)
(Levin etal.,2007).

HPA HPA Autism


axis 5-HT axis 5-HT Epidemiologically, reciprocal connection
between epilepsy and autism has been well
established:approximately 30% of patients with
5-HT1A autism develop epilepsy at some point of their
5-HT1A
lives, and at the same time around 30% of patients
FIGURE 10.1: Perturbations of inflammatory with epilepsy as a primary diagnosis fit the cri-
signaling: Role in seizures and in epilepsy- teria of being diagnosed with autism (Tuchman
associated depression. Under normal conditions, and Rapin, 2002; Clarke etal., 2005; Seidenberg
the expression of IL-1 in the hippocampus is relatively etal.,2009).
low; therefore, its modulation of the hypothalamo- Causes of the comorbidity remain unclear.
pituitary-adrenocortical (HPA) axis is negligible. Several scenarios have been suggested, ranging
Within the physiological parameters, the HPA axis from an incidental coappearance to compris-
exerts little or no effects on somatodendritic 5-HT1A ing two manifestations of the same pathology
receptors in raphe nuclei. Raphe 5-HT1A receptors (e.g., fragile X chromosome; Levisohn, 2007).
regulate 5-HT release from the raphe into the hip- However, since both epilepsy and autism are
pocampus via equilibrated autoinhibitoryloop. multifactorial and multicausative diseases, the
nature of the association between the two con-
We can envisage two main, nonmutually exclusive sce-
ditions is very likely to vary among different
narios:(a)a primary inflammatory event may concur
patients.
to trigger the onset and recurrence of epileptic seizures
Causes of autism proper are being exam-
via the activation of IL-1 signaling, and in parallel the
ined along three lines. Genetic mechanisms
same mechanism may produce symptoms of depres-
have been extensively studied; a growing
sion by altering the HPA axis; (b)epilepsy may lead to
number of genes has been identified and their
or perpetuate the overexpression of IL-1 in the hip-
role has been supported by experimental evi-
pocampus, which in turn hyperactivates the HPAaxis.
dence (Moy and Nadler, 2008; Abrahams and
A pathological hyperactive HPA axis upregulates Geschwind, 2010; Eapen, 2011). At the same
raphe 5-HT1A receptors. This shifts regulation of time, autism concordance among monozygotic
5-HT release in favor of autoinhibition and results in twins is between 50% and 70%, and, despite a
the diminished serotonergic tone, the latter producing growing number of candidate genes, cases of
depression. autism traceable to a genetic trait remain at 15%
(Hallmayer etal., 2011). Further, spontaneous
resolution of autism symptoms in a cohort of
patients with polymorphism of 5-HT1A recep- patients (or optimal-outcome individuals) sug-
tor gene, which translates into the upregulation gests the transient nature of etiological fac-
of 5-HT1A autoreceptors (Lemonde etal., 2003; tors in some cases (Fein et al., 2013). These
Richardson-Jones etal., 2010). At the same time, observations strongly implicate environmental
5-HT1A upregulation may be acquired and not risks. Indeed, exposure of pregnant women to
necessarily inherited. For example, an excessively infections, valproic acid, ethanol, and stress
hyperactive HPA axis leads to the upregulation have been suggested as risk factors for the
of 5-HT1A autoreceptors (Bellido et al., 2004; development of autism in offspring (Patterson,
Judge et al., 2004). In epilepsy, the HPA hyper- 2009; Atladottir et al., 2010; Landrigan, 2010;
activity is triggered and sustained by chronically Dufour-Rainfray etal., 2011). The third school
enhanced IL-1 signaling (Mazarati etal., 2010). of thought combines genetic and environmen-
Hence, the disruption of the latter may not only tal causes under the concept of a double-hit
alleviate depression but restore the effectiveness umbrella.
of SSRIs. Indeed, the combined administration Among environmental factors, infec-
of IL-1ra and fluoxetine fully reversed depres- tions during pregnancy have been receiving
sion in chronic epileptic rats (Pineda et al., growing clinical and experimental support.
2012). This finding may be applicable not only to Epidemiological studies provide a compelling
epilepsy-associated depression but also to other association between maternal infection and
164 Part II:Homeostatic Control

the increased chance of development of autism Maternal immune


Offspring
in children (Ciaranello and Ciaranello, 1995; activation
Jonakait, 2007; Moy and Nadler, 2008; Atladottir
etal., 2010; Atladottir etal., 2012). Atladottir etal. IL-6 Autism
found that viral infection in the first trimester
Maternal
and bacterial infection in the second trimester infection
of pregnancy were associated with autism in the
IL-1 Epilepsy
offspring, with hazard ratios of 2.98 and 1.42,
respectively (Atladottir et al., 2010); another
study by this group found that maternal influenza
was associated with a twofoldand prolonged Other
episodes of fever with a threefoldincreased
risk of autism in the offspring (Atladottir et al., FIGURE10.2: Maternal immune activation (MIA)
2012). Experimental studies have established as a risk factor for the development of autism
that injection of pregnant rodents with either and epilepsy in the offspring. Among various
LPS or polyinosinicpolycytidylic acid (Poly I:C), components of the MIA triggered by infection, IL-6
which mimic Gram-negative and viral infections, is necessary and sufficient for producing autism in
respectively, lead to the development in offspring the offspring in the adulthood. Neither IL-6 nor IL-1
of autism-like impairments (Patterson, 2002; alone produces epileptic phenotype. However, com-
Shi etal., 2003; Fatemi etal., 2005; Boksa, 2010; bined induction of IL-6 and IL-1 is both sufficient and
Patterson, 2011a, 2011b; Schwartzer et al., 2013). necessary for predisposing the offspring to epilepsy.
Poly I:C-induced maternal immune activation Hence, the autismepilepsy comorbidity following an
(MIA) in rhesus monkeys produced offspring infection during pregnancy depends on the extent of
with impaired sociability and restricted behavior the exposure of embryonic brain to IL-6 andIL-1.
(Bauman etal., 2013). Electrophysiological studies
in the MIA offspring revealed impaired commu-
nication between the hippocampus and prefron- Among components of MIA, interleukin-6
tal cortex, which represents a hallmark of autism (IL-6) has been established as a factor solely
spectrum disorder (Baharnoori etal.,2009). responsible for the development of autism in off-
Detrimental effects of maternal infection spring. Thus anti-IL-6 antibody coadministered
occur through MIA (Figure 10.2). The latter, with Poly I:C in pregnant mice prevented the
frequently described as the cytokine storm, occurrence of autism phenotype in the offspring,
involves massive activation of inflammatory while mice treated with recombinant IL-6 pro-
pathways that, when acting on the embryonic duced the offspring with autism-like behavioral
brain, induce perturbations in neuronal develop- impairments similar to those induced by Poly I:C
ment, connectivity, metabolism, and function- (Smith etal., 2007). In a separate study (Pineda
ing, ultimately resulting in autism (Jonakait, et al., 2013), prenatal exposure to IL-6, but not
2007; Ponzio et al., 2007; Buehler, 2011; Deng to IL-1, led to autism-like behavior in the off-
etal., 2011; Patterson, 2011b; Oskvig etal.,2012). spring of mice, while none of the two cytokines
Concurrently with autism, MIA primes off- administered alone produced epileptic pheno-
spring to epilepsy (Figure 10.2). Thus the offspring type. However, when IL-6 and IL-1 were admin-
of rats treated with LPS during pregnancy exhib- istered together, the offspring presented with the
ited increased severity of pilocarpine-induced accelerated hippocampal kindling rate and pro-
status epilepticus, as well as exacerbated neurode- longed kindling retention (i.e., the effects were
generation in the hippocampus (Yin etal., 2013). similar to those induced by PolyI:C).
MIA induced by Poly I:C in genetically normal Conversely, immune blockade of IL-1 pre-
mice increased the severity of status epilepticus vented facilitatory effects of prenatal Poly I:C on
induced by intrahippocampal administration of kindling epilepsy but not on autism behavior,
kainic acid and subsequent neurodegeneration while the blockade of IL-6 prevented both autism
in the hippocampus of the offspring (Kirschman and epileptic phenotypes induced by MIA. These
etal., 2011). Prenatal exposure to Poly I:C accel- observations led to a hypothesis that the develop-
erated the rate of hippocampal kindling epilep- ment of autism without concurrent epilepsy and
togenesis during adolescence and prolonged the autism accompanied by epilepsy in the offspring
retention of a kindling-induced epileptic state following MIA depends on the extent of the
(Pineda etal.,2013). exposure of embryonic brain to IL-6 and IL-1
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 165

(Pineda etal., 2013; Figure 10.2). If corroborated celecoxib alleviated learning deficits in rats in
in patients, these findings may translate both into which chronic epilepsy had been induced by sys-
early diagnostic (through measuring plasma lev- temically administered kainic acid (Gobbo and
els of the cytokines) and preventive (e.g., through OMara,2004).
using cytokine antibodies) measures that would Memory impairments represent one of hall-
respectively predict and significantly alleviate marks of the LPS sickness (Tarr etal., 2011), thus
the risk of autism and epilepsy in patients atrisk. implicating TLR4. In addition, recent studies
Postnatal inflammation has been associ- have been pointing toward the RAGE-mediated
ated with autism as well. For example, increased inflammatory pathway as a negative regulator
blood levels of RAGE ligands such as HMGB1 of cognitive functions. In particular, increased
and S100A9 (Boso et al., 2006; Emanuele et al., RAGE signaling has been implicated in mecha-
2010), as well as the enhanced reactivity of TLR2 nisms of memory impairments in Alzheimers
and TLR4 expressing monocytes (Enstrom etal., disease (Arancio etal.,2004).
2010), have been documented in people with As it has been discussed earlier, HMGB1 is
autism. Increased levels of monocyte chemoat- overexpressed in the epileptic tissue and may
tractant protein 1 and TNF1 were documented precipitate ictogenesis via activating both TLR4
in the cortex, white matter, and cerebellum of (Maroso etal., 2010)and RAGE (Iori etal., 2013).
patients with autism (Vargas etal., 2005). Acute It is therefore conceivable that stimulation of
exposure of neonatal rat pups to a combination both TLR4 and RAGE by HMGB1 may, along
of LPS to mimic Gram-negative bacterial infec- with exacerbating seizures, lead to memory dys-
tion, doxorubicin to produce diffuse brain dam- function (Figure 10.3). Indeed, central adminis-
age involving the forebrain and brainstem, and tration of HMGB1 in wild type as well as TLR4
p-chlorophenylalanine to deplete central seroto- and RAGE knockout mice impaired animals
nin resulted in complex subacute perturbations performance in the novel object recognition
comprised of infantile spasms, social disen- test, while pharmacological blockade of TLR4 in
gagement, and restricted behavior (Scantlebury RAGE knockout mice abolished amnestic effects
etal.,2010). of HMGB1 (Mazarati etal., 2011). Thus excessive
HMGB1 signaling, which has been established
Cognitive Impairments in the epileptic brain, may lead to concurrent
Memory and cognitive impairments are com-
mon among epilepsy patients (Giovagnoli
and Avanzini, 1999) and have been consist- Seizures
ently reported in animal epilepsy models (b)
(Lenck-Santini and Holmes, 2008; Muller (a)
Proinflammatory
et al., 2009). Causes of these impairments have event
been attributed to neuronal cell loss (Holmes (c)
et al., 2002) or dysfunction (Lenck-Santini and HMGB1
Holmes, 2008; Karnam etal., 2009), as well as to
adverse effects of antiepileptic drugs (Sankar and TLR4 RAGE
Holmes,2004).
Several mediators of inflammation exert
detrimental effects on cognition. For example, Cognitive impairments
COX-2, an enzyme that is ubiquitously involved
in inflammatory responses and is constitu- FIGURE10.3: High mobility group box 1 protein
tively expressed in the hippocampus (Yamagata (HMGB1)-dependent pathways, seizures and
et al., 1993), negatively regulates learning and cognitive impairments in epilepsy. (a)Recurring
memory (Teather et al., 2002). The increase of seizures amplify HMGB1 expression and its extracel-
COX-2 expression has been reported in the hip- lular release in the hippocampus. HMGB1 then may
pocampus of patients with temporal TLE (Das lead to cognitive impairments via the activation of both
et al., 2012; Desjardins et al., 2003; Rojas et al., a Toll-like 4 receptor (TLR4) and receptor for advance
2014), as well as in an animal model of TLE glycation end products (RAGE). Alternatively a primary
(Kawaguchi etal., 2005). Deletion of COX-2 from inflammatory event may lead (b) to epilepsy through
forebrain neurons improved memory deficits discussed mechanisms (e.g., activation of IL-1R1/TLR4
in mice with pilocarpine-induced TLE (Levin signaling) and (c)to concurrent cognitive impairments
et al., 2012). Accordingly, a COX-2 inhibitor via the activation of HMGB1 signaling.
166 Part II:Homeostatic Control

selective memory deficits via activating both the accumulated evidence makes a strong case
TLR4 and RAGE (Figure 10.3). In addition, in for anti-inflammatory interventions in epilepsy
light of a possible role of RAGE in Alzheimers patients.
dementia, it is tempting to speculate that the Many drugs are clinically available for the
HMGB1-induced overstimulation of RAGE may management of various inflammatory diseases;
represent a mechanistic link explaining the high therefore their application in epilepsy may be
incidence of comorbidity between epilepsy and streamlined, provided that they would show effi-
Alzheimers disease (Noebels,2011). cacy in clinical trials. Examples include COX-2
In conclusion, brain inflammation may inhibitors, IL-1ra (Kineret; used for the treat-
constitute if not a primary cause then at least a ment of rheumatoid arthritis), monoclonal anti-
confounding factor in mechanisms of neurobe- bodies against TNF- (Adalimumab, Infliximab,
havioral comorbidities of epilepsy. Inflammatory used in rheumatoid arthritis and Crohns dis-
processes triggered in the epileptic brain may ease) or IL-1 (Canakinumab, approved for the
translate into interictal behavioral abnormalities. treatment of cryopyrin-associated periodic syn-
Hence, inflammatory factors may represent tar- drome, clinical trials for chronic obstructive pul-
gets for evidence-based therapeutic interventions, monary disease), and IL-6 (Siltuximab, clinical
as well as biomarkers that may effectively assess trials for non-Hodgkins lymphoma and multiple
the risk of the development of comorbidities of myeloma) or treatments targeting hyperactivated
epilepsy. glial cells (e.g., fingolimod, used in multiple scle-
rosis, minocycline). The fact that some of these
CONCLUDING REMARKS AND drugs do not readily permeate BBB in the normal
T H E R A P E U T I C I M P L I C AT I O N S brain makes their use challenging, although this
There has been an ongoing interest in nonneu- may potentially limit their CNS effects to condi-
ronal mechanisms of epilepsy, epileptogenesis, tions when BBB is damaged (as discussed earlier).
and ictogenesis. This interest is explained by a The clinical development of VX-765, a compound
widely acknowledged mismatch between the that blocks IL-1 biosynthesis by inhibiting
advancements in understanding the role of clas- interleukin converting enzyme, represents an
sical neurotransmitters (particularly GABA and example of a more advanced approach, whereby
glutamate) in epilepsy on the one hand and the an anti-inflammatory drug has been evaluated
lack of progress in pharmacotherapy based on in a phase 2 clinical study for the treatment of
targeting these neurotransmitter systems on pharmacoresistant epilepsy (http://clinicaltrials.
the other hand. Indeed, a significant propor- gov/ct2/show/NCT01048255;www.epilepsy.com/
tion of epilepsy patients remains nonrespond- files/Pipeline2012/67).
ent or poorly respondent to antiepileptic drugs
(even the ones of newer generations), which are ACK NOWLEDGMENTS
designed to effectively normalize the function AV is grateful to Fondazione Monzino (Grant
of classical transmitter systems. This mismatch 20032005) and EPITARGET (FP7/20072013,
has prompted a search for alternative mecha- grant agreement n602102) for their support given
nisms and novel therapeutic targets. Discovery to the laboratory of Experimental Neurology at
of active anticonvulsant roles of regulatory pep- the Istituto di Ricerche Farmacologiche Mario
tides (such as somatostatin, neuropeptide Y, and Negri, which allowed to obtain some of the find-
galanin), adenosine, and endocannabinoids rep- ings reported in this review article.
resent examples of such efforts. The ubiquitous
nature of inflammation makes it reasonable to References
assume that inflammatory responses are trig- Abrahams BS, Geschwind DH (2010) Connecting
gered by seizures when epilepsy is a primary genes to brain in the autism spectrum disorders.
condition, or reciprocally inflammation stem- Arch Neurol 67:395399.
ming from other causes may prime brain to epi- Akassoglou K, Probert L, Kontogeorgos G, Kollias G
lepsy and concurrent neurobehavioral disorders (1997) Astrocyte-specific but not neuron-specific
(Figures 10.1 and 10.3). The discussed data cor- transmembrane TNF triggers inflammation and
roborate these assumptions in both clinical and degeneration in the central nervous system of
experimental settings. Certainly, it remains to transgenic mice. J Immunol 158:438445.
be seen whether the promise of targeting inflam- Akin D, Ravizza T, Maroso M, Carcak N, Eryigit
mation for the effective treatment of epilepsy T, Vanzulli I, Aker RG, Vezzani A, Onat FY
and its comorbidities will be fulfilled; however, (2011) IL-1 is induced in reactive astrocytes in
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 167

the somatosensory cortex of rats with genetic Vezzani A (2009) Molecular and functional
absence epilepsy at the onset of spike-and-wave interactions between TNF-alpha receptors and
discharges, and contributes to their occurrence. the glutamatergic system in the mouse hippo-
Neurobiol Dis 44:259269. campus: implications for seizure susceptibility.
Arancio O etal. (2004) RAGE potentiates A-induced Neuroscience 161:293300.
perturbation of neuronal function in transgenic Barbui C, Hotopf M, Garattini S (2002) Fluoxetine
mice. Embo J 23:40964105. dose and outcome in antidepressant drug trials.
Aronica E, Crino PB (2011) Inflammation in epi- Eur J Clin Pharmacol 58:379386.
lepsy:clinical observations. Epilepsia 52 Suppl 3: Bauer J, Elger CE, Hans VH, Schramm J, Urbach H,
2632. Lassmann H, Bien CG (2007) Astrocytes are a
Aronica E, Ravizza T, Zurolo E, Vezzani A (2012) specific immunological target in Rasmussens
Astrocyte immune response in epilepsy. Glia encephalitis. Ann Neurol 62:6780.
60:12581268. Bauman MD, Iosif AM, Smith SE, Bregere C,
Atladottir HO, Henriksen TB, Schendel DE, Parner ET Amaral DG, Patterson PH (2013) Activation of
(2012) Autism after infection, febrile episodes, the maternal immune system during pregnancy
and antibiotic use during pregnancy:An explor- alters behavioral development of rhesus monkey
atory study. Pediatrics 130:e1447e1454. offspring. Biol Psychiatry 75(4):332341.
Atladottir HO, Thorsen P, Ostergaard L, Schendel DE, Baumann B, Danos P, Diekmann S, Krell D, Bielau
Lemcke S, Abdallah M, Parner ET (2010) H, Geretsegger C, Wurthmann C, Bernstein HG,
Maternal infection requiring hospitalization Bogerts B (1999) Tyrosine hydroxylase immu-
during pregnancy and autism spectrum disor- noreactivity in the locus coeruleus is reduced in
ders. J Autism Dev Disord 40:14231430. depressed non-suicidal patients but normal in
Auvin S, Shin D, Mazarati A, Nakagawa J, Miyamoto J, depressed suicide patients. Eur Arch Psychiatry
Sankar R (2007) Inflammation exacerbates Clin Neurosci 249:212219.
seizure-induced injury in the immature brain. Beattie EC, Stellwagen D, Morishita W, Bresnahan
Epilepsia 48 Suppl 5:2734. JC, Ha BK, Von Zastrow M, Beattie MS, Malenka
Auvin S, Shin D, Mazarati A, Sankar R (2010) RC (2002) Control of synaptic strength by glial
Inflammation induced by LPS enhances epilep- TNFalpha. Science 295:22822285.
togenesis in immature rat and may be partially Bellido I, Hansson AC, Gomez-Luque AJ, Andbjer B,
reversed by IL1RA. Epilepsia 51 Suppl 3:3438. Agnati LF, Fuxe K (2004) Corticosterone strongly
Baharnoori M, Brake WG, Srivastava LK (2009) increases the affinity of dorsal raphe 5-HT1A
Prenatal immune challenge induces develop- receptors. Neuroreport 15:14571459.
mental changes in the morphology of pyramidal Bien CG, Bauer J, Deckwerth TL, Wiendl H,
neurons of the prefrontal cortex and hippocam- Deckert M, Wiestler OD, Schramm J, Elger CE,
pus in rats. Schizophr Res 107:99109. Lassmann H (2002) Destruction of neurons
Balosso S, Liu J, Bianchi ME, Vezzani A (2014) by cytotoxic T cells: A new pathogenic mecha-
Disulfide-containing high mobility group nism in Rasmussens encephalitis. Ann Neurol
box-1 promotes N-methyl-D-aspartate recep- 51:311318.
tor function and excitotoxicity by activating Bien CG, Gleissner U, Sassen R, Widman G, Urbach
Toll-like receptor 4-dependent signaling in H, Elger CE (2004) An open study of tacrolimus
hippocampal neurons. Antioxid Redox Signal therapy in Rasmussen encephalitis. Neurology
21(12):17261740. 62:21062109.
Balosso S, Maroso M, Sanchez-Alavez M, Ravizza T, Bien CG, Scheffer IE (2011) Autoantibodies and epi-
Frasca A, Bartfai T, Vezzani A (2008) A novel lepsy. Epilepsia 52 Suppl 3:1822.
non-transcriptional pathway mediates the Bien CG, Tiemeier H, Sassen R, Kuczaty S, Urbach H,
proconvulsive effects of interleukin-1. Brain von Lehe M, Becker AJ, Bast T, Herkenrath P,
131:32563265. Karenfort M, Kruse B, Kurlemann G, Rona S,
Balosso S, Ravizza T, Aronica E, Vezzani A (2013) Schubert-Bast S, Vieker S, Vlaho S, Wilken B,
The dual role of TNF- and its receptors in sei- Elger CE (2013) Rasmussen encephalitis:
zures. Exp Neurol 247:267271. Incidence and course under randomized therapy
Balosso S, Ravizza T, Perego C, Peschon J, Campbell IL, with tacrolimus or intravenous immunoglobu-
De Simoni MG, Vezzani A (2005) Tumor necro- lins. Epilepsia 54:543550
sis factor-alpha inhibits seizures in mice via p75 Bien CG, Vincent A, Barnett MH, Becker AJ, Blumcke I,
receptors. Ann Neurol 57:804812. Graus F, Jellinger KA, Reuss DE, Ribalta T,
Balosso S, Ravizza T, Pierucci M, Calcagno E, Schlegel J, Sutton I, Lassmann H, Bauer J (2012)
Invernizzi RW, Di Giovanni G, Esposito E, Immunopathology of autoantibody-associated
168 Part II:Homeostatic Control

encephalitides: Clues for pathogenesis. Brain (2005) The prevalence of autistic spectrum dis-
135:16221638. order in children surveyed in a tertiary care epi-
Biswas SK, Lopez-Collazo E (2009) Endotoxin toler- lepsy clinic. Epilepsia 46:19701977.
ance: New mechanisms, molecules and clinical Craft JM, Watterson DM, Van Eldik LJ (2005)
significance. Trends Immunol 30:475487. Neuroinflammation:Apotential therapeutic tar-
Boer K, Jansen F, Nellist M, Redeker S, van den get. Expert Opin Ther Targets 9:887900.
Ouweland AM, Spliet WG, van Nieuwenhuizen O, Dalmau J, Gleichman AJ, Hughes EG, Rossi
Troost D, Crino PB, Aronica E (2008) JE, Peng X, Lai M, Dessain SK, Rosenfeld
Inflammatory processes in cortical tubers and MR, Balice-Gordon R, Lynch DR (2008)
subependymal giant cell tumors of tuberous scle- Anti-NMDA-receptor encephalitis: Case series
rosis complex. Epilepsy Res 78:721. and analysis of the effects of antibodies. Lancet
Boer K, Spliet WG, van Rijen PC, Redeker S, Troost D, Neurol 7:10911098.
Aronica E (2006) Evidence of activated microg- Dalmau J, Lancaster E, Martinez-Hernandez E,
lia in focal cortical dysplasia. J Neuroimmunol Rosenfeld MR, Balice-Gordon R (2011) Clinical
173:188195. experience and laboratory investigations in
Boksa P (2010) Effects of prenatal infection on brain patients with anti-NMDAR encephalitis. Lancet
development and behavior: A review of find- Neurol 10:6374.
ings from animal models. Brain Behav Immun Dalmau J, Tuzun E, Wu HY, Masjuan J, Rossi JE,
24:881897. Voloschin A, Baehring JM, Shimazaki H, Koide
Boso M, Emanuele E, Minoretti P, Arra M, Politi P, R, King D, Mason W, Sansing LH, Dichter MA,
Ucelli di Nemi S, Barale F (2006) Alterations Rosenfeld MR, Lynch DR (2007) Paraneoplastic
of circulating endogenous secretory RAGE anti-N-methyl-D-aspartate receptor encephalitis
and S100A9 levels indicating dysfunction of associated with ovarian teratoma. Ann Neurol
the AGE-RAGE axis in autism. Neurosci Lett 61:2536.
410:169173. Dantzer R (2006) Cytokine, sickness behavior, and
Brambilla D, Franciosi S, Opp MR, Imeri L (2007) depression. Neurol Clin 24:441460.
Interleukin-1 inhibits firing of serotonergic neu- Dantzer R, Kelley KW (1989) Stress and immu-
rons in the dorsal raphe nucleus and enhances nity:An integrated view of relationships between
GABAergic inhibitory post-synaptic potentials. the brain and the immune system. Life Sci
Eur J Neurosci 26:18621869. 44:19952008.
Bremmer MA, Beekman AT, Deeg DJ, Penninx Das A, Wallace GC, Holmes C, McDowell ML, Smith
BW, Dik MG, Hack CE, Hoogendijk WJ (2008) JA, Marshall JD, Bonilha L, Edwards JC, Glazier
Inflammatory markers in late-life depres- SS, Ray SK, Banik NL (2012) Hippocampal tis-
sion: Results from a population-based study. J sue of patients with refractory temporal lobe
Affect Disord 106:249255. epilepsy is associated with astrocyte activation,
Bruce TO (2008) Comorbid depression in rheu- inflammation, and altered expression of chan-
matoid arthritis: Pathophysiology and clinical nels and receptors. Neuroscience 220:237246.
implications. Curr Psychiatry Rep 10:258264. Deng MY, Lam S, Meyer U, Feldon J, Li Q, Wei R,
Buehler MR (2011) A proposed mechanism for Luk L, Chua SE, Sham P, Wang Y, McAlonan GM
autism: an aberrant neuroimmune response (2011) Frontal-subcortical protein expression
manifested as a psychiatric disorder. Med following prenatal exposure to maternal inflam-
Hypotheses 76:863870. mation. PLoS One 6:e16638.
Carroll BJ, Cassidy F, Naftolowitz D, Tatham NE, Deprez F, Zattoni M, Mura ML, Frei K, Fritschy JM
Wilson WH, Iranmanesh A, Liu PY, Veldhuis JD (2013) Adoptive transfer of T lymphocytes in
(2007) Pathophysiology of hypercortisolism in immunodeficient mice influences epileptogen-
depression. Acta Psychiatr Scand Suppl:90103. esis and neurodegeneration in a model of tempo-
Chan D, Henley SM, Rossor MN, Warrington EK ral lobe epilepsy. Neurobiol Dis 44:174184.
(2007) Extensive and temporally ungraded ret- Desjardins P, Sauvageau A, Bouthillier A, Navarro
rograde amnesia in encephalitis associated with D, Hazell AS, Rose C, Butterworth RF (2003)
antibodies to voltage-gated potassium channels. Induction of astrocytic cyclooxygenase-2 in
Arch Neurol 64:404410. epileptic patients with hippocampal sclerosis.
Ciaranello AL, Ciaranello RD (1995) The neurobi- Neurochem Int 42:299303
ology of infantile autism. Annu Rev Neurosci Devinsky O, Vezzani A, Najjar S, De Lanerolle
18:101128. NC, Rogawski MA (2013) Glia and epi-
Clarke DF, Roberts W, Daraksan M, Dupuis A, lepsy: Excitability and inflammation. Trends
McCabe J, Wood H, Snead OC, 3rd, Weiss SK Neurosci 36:174184.
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 169

Dmowska M, Cybulska R, Schoenborn R, Piersiak T, associated with glutamic-acid-decarboxylase


Jaworska-Adamu J, Gawron A (2010) Behavioural autoantibodies. Lancet 352:457.
and histological effects of preconditioning with Giovagnoli AR, Avanzini G (1999) Learning and
lipopolysaccharide in epileptic rats. Neurochem memory impairment in patients with temporal
Res 35:262272. lobe epilepsy:Relation to the presence, type, and
Dufour-Rainfray D, Vourch P, Tourlet S, Guilloteau D, location of brain lesion. Epilepsia 40:904911.
Chalon S, Andres CR (2011) Fetal exposure Glass CK, Saijo K, Winner B, Marchetto MC, Gage
to teratogens: Evidence of genes involved in FH (2010) Mechanisms underlying inflamma-
autism. Neurosci Biobehav Rev 35:12541265. tion in neurodegeneration. Cell 140:918934.
Dunn AJ, Swiergiel AH (2005) Effects of interleukin-1 Gobbo OL, OMara SM (2004) Post-treatment,
and endotoxin in the forced swim and tail sus- but not pre-treatment, with the selective
pension tests in mice. Pharmacol Biochem Behav cyclooxygenase-2 inhibitor celecoxib mark-
81:688693. edly enhances functional recovery from kainic
Dunn AJ, Swiergiel AH, de Beaurepaire R (2005) acid-induced neurodegeneration. Neuroscience
Cytokines as mediators of depression: What 125:317327.
can we learn from animal studies? Neurosci Goshen I, Yirmiya R (2009) Interleukin-1 (IL-1):
Biobehav Rev 29:891909. A central regulator of stress responses. Front
Eapen V (2011) Genetic basis of autism:is there a way Neuroendocrinol 30:3045.
forward? Curr Opin Psychiatry 24:226236. Guo JY, Li CY, Ruan YP, Sun M, Qi XL, Zhao BS,
Emanuele E, Boso M, Brondino N, Pietra S, Barale F, Luo F (2009) Chronic treatment with celecoxib
Ucelli di Nemi S, Politi P (2010) Increased serum lev- reverses chronic unpredictable stress-induced
els of high mobility group box 1 protein in patients depressive-like behavior via reducing
with autistic disorder. Prog Neuropsychopharmacol cyclooxygenase-2 expression in rat brain. Eur J
Biol Psychiatry 34:681683. Pharmacol 612:5460.
Engel T, Jimenez-Pacheco A, Miras-Portugal MT, Hallmayer J, Cleveland S, Torres A, Phillips J, Cohen B,
Diaz-Hernandez M, Henshall DC (2012) P2X7 Torigoe T, Miller J, Fedele A, Collins J, Smith K,
receptor in epilepsy: Role in pathophysiology Lotspeich L, Croen LA, Ozonoff S, Lajonchere C,
and potential targeting for seizure control. Int J Grether JK, Risch N (2011) Genetic heritabil-
Physiol Pathophysiol Pharmacol 4:174187. ity and shared environmental factors among
Enstrom AM, Onore CE, Van de Water JA, Ashwood P twin pairs with autism. Arch Gen Psychiatry
(2010) Differential monocyte responses to TLR 68:10951102.
ligands in children with autism spectrum disor- Hauf M, Wiest R, Nirkko A, Strozzi S, Federspiel A
ders. Brain Behav Immun 24:6471. (2009) Dissociation of epileptic and inflamma-
Fabene PF eta l. (2008) Arole for leukocyte-endothelial tory activity in Rasmussen encephalitis. Epilepsy
adhesion mechanisms in epilepsy. Nat Med Res 83:265268.
14:13771383. Holmes GL, Khazipov R, Ben-Ari Y (2002)
Fatemi SH, Pearce DA, Brooks AI, Sidwell RW (2005) Seizure-induced damage in the developing
Prenatal viral infection in mouse causes differen- human:Relevance of experimental models. Prog
tial expression of genes in brains of mouse prog- Brain Res 135:321334.
eny:Apotential animal model for schizophrenia Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L,
and autism. Synapse 57:9199. Tsou R, Parsons TD, Lynch DR, Dalmau J,
Fein D, Marton M, Eigesti I, Kelley E, Naigles LR, Balice-Gordon RJ (2010) Cellular and synaptic
Schultz RT, Stevens M, Helt M, Orinstein A, mechanisms of anti-NMDA receptor encephali-
Rosenthal M, Troyb E, Tyson K (2013) Optimal tis. J Neurosci 30:58665875.
outcome in individuals with a history of autism. Iori V, Maroso M, Rizzi M, Iyer AM, Vertemara R,
J Child Psychol Psychiatry 54:195205. Carli M, Agresti A, Antonelli A, Bianchi ME,
Garcia-Morales I, de la Pena Mayor P, Kanner AM Aronica E, Ravizza T, Vezzani A (2013) Receptor
(2008) Psychiatric comorbidities in epilepsy: for advanced glycation endproducts is upregu-
Identification and treatment. Neurologist lated in temporal lobe epilepsy and contrib-
14:S15S25. utes to experimental seizures. Neurobiol Dis
Gilliam F, Kuzniecky R, Faught E, Black L, Carpenter 58:102114.
G, Schrodt R (1997) Patient-validated content of Irani SR, Bera K, Waters P, Zuliani L, Maxwell S,
epilepsy-specific quality-of-life measurement. Zandi MS, Friese MA, Galea I, Kullmann DM,
Epilepsia 38:233236. Beeson D, Lang B, Bien CG, Vincent A (2010)
Giometto B, Nicolao P, Macucci M, Tavolato B, Foxon R, N-methyl-D-aspartate antibody encepha-
Bottazzo GF (1998) Temporal-lobe epilepsy litis: Temporal progression of clinical and
170 Part II:Homeostatic Control

paraclinical observations in a predominantly Kwon YS, Pineda E, Auvin S, Shin D, Mazarati A,


non-paraneoplastic disorder of both sexes. Brain Sankar R (2013) Neuroprotective and anti-
133:16551667. epileptogenic effects of combination of
Iyer A, Zurolo E, Spliet WG, van Rijen PC, Baayen JC, anti-inflammatory drugs in the immature brain.
Gorter JA, Aronica E (2010) Evaluation of J Neuroinflammation10:30.
the innate and adaptive immunity in type I Lai M, Hughes EG, Peng X, Zhou L, Gleichman AJ,
and type II focal cortical dysplasias. Epilepsia Shu H, Mata S, Kremens D, Vitaliani R,
51:17361773. Geschwind MD, Bataller L, Kalb RG, Davis R,
Jiang J, Ganesh T, Du Y, Quan Y, Serrano G, Qui Graus F, Lynch DR, Balice-Gordon R, Dalmau J
M, Speigel I, Rojas A, Lelutiu N, Dingledine R (2009) AMPA receptor antibodies in limbic
(2012) Small molecule antagonist reveals encephalitis alter synaptic receptor location.
seizure-induced mediation of neuronal injury Ann Neurol 65:424434.
by prostaglandin E2 receptor subtype EP2. Proc Lai M, Huijbers MG, Lancaster E, Graus F, Bataller L,
Natl Acad Sci USA 109:31493154. Balice-Gordon R, Cowell JK, Dalmau J (2010)
Jonakait GM (2007) The effects of maternal inflam- Investigation of LGI1 as the antigen in limbic
mation on neuronal development: possible encephalitis previously attributed to potassium
mechanisms. Int J Dev Neurosci 25:415425. channels:Acase series. Lancet Neurol 9:776785.
Judge SJ, Ingram CD, Gartside SE (2004) Lalic T, Pettingill P, Vincent A, Capogna M (2010)
Moderate differences in circulating corticos- Human limbic encephalitis serum enhances hip-
terone alter receptor-mediated regulation of pocampal mossy fiber-CA3 pyramidal cell syn-
5-hydroxytryptamine neuronal activity. J aptic transmission. Epilepsia 52:121131.
Psychopharmacol 18:475483. Landrigan PJ (2010) What causes autism? Exploring
Kanner AM, Schachter SC, Barry JJ, Hersdorffer DC, the environmental contribution. Curr Opin
Mula M, Trimble M, Hermann B, Ettinger AE, Pediatr 22:219225.
Dunn D, Caplan R, Ryvlin P, Gilliam F (2012) Lang UE, Borgwardt S (2013) Molecular mechanisms
Depression and epilepsy:Epidemiologic and neu- of depression: Perspectives on new treatment
robiologic perspectives that may explain their high strategies. Cell Physiol Biochem 31:761777.
comorbid occurrence. Epilepsy Behav 24:156168. Lemonde S, Turecki G, Bakish D, Du L, Hrdina PD,
Karnam HB, Zhou JL, Huang LT, Zhao Q, Shatskikh T, Bown CD, Sequeira A, Kushwaha N, Morris SJ,
Holmes GL (2009) Early life seizures cause Basak A, Ou XM, Albert PR (2003) Impaired
long-standing impairment of the hippocampal repression at a 5-hydroxytryptamine 1A recep-
map. Exp Neurol 217:378387. tor gene polymorphism associated with major
Kawaguchi K, Hickey RW, Rose ME, Zhu L, Chen J, depression and suicide. J Neurosci 23:87888799.
Graham SH (2005) Cyclooxygenase-2 expression Lenck-Santini PP, Holmes GL (2008) Altered
is induced in rat brain after kainate-induced sei- phase precession and compression of tempo-
zures and promotes neuronal death in CA3 hip- ral sequences by place cells in epileptic rats. J
pocampus. Brain Res 1050:130137. Neurosci 28:50535062.
Kirschman LT, Borysiewicz E, Fil D, Konat GW Levin GM, Bowles TM, Ehret MJ, Langaee T, Tan JY,
(2011) Peripheral immune challenge with dsRNA Johnson JA, Millard WJ (2007) Assessment of
enhances kainic acid-induced status epilepticus. human serotonin 1A receptor polymorphisms
Metab Brain Dis 26:9193. and SSRI responsiveness. Mol Diagn Ther
Koh S, Magid R, Chung H, Stine CD, Wilson DN 11:155160.
(2007) Depressive behavior and selective Levin JR, Serrano G, Dingledine R (2012) Reduction
down-regulation of serotonin receptor expres- in delayed mortality and subtle improve-
sion after early-life seizures: Reversal by envi- ment in retrograde memory performance in
ronmental enrichment. Epilepsy Behav 10:2631. pilocarpine-treated mice with conditional neuro-
Kondziella D, Alvestad S, Vaaler A, Sonnewald U nal deletion of cyclooxygenase-2 gene. Epilepsia
(2007) Which clinical and experimental data 53:14111420.
link temporal lobe epilepsy with depression? J Levisohn PM (2007) The autism-epilepsy connec-
Neurochem 103:21362152. tion. Epilepsia 48 Suppl 9:3335.
Krishnadas R, Cavanagh J (2012) Depression: An Librizzi L, No F, Vezzani A, de Curtis M, Ravizza T
inflammatory illness? J Neurol Neurosurg (2012) Seizure-induced brain-borne inflamma-
Psychiatry 83:495502. tion sustains seizure recurrence and blood-brain
Kulkarni SK, Dhir A (2009) Cyclooxygenase in epi- barrier damage. Ann Neurol 72:8290.
lepsy: From perception to application. Drugs Librizzi L, Regondi MC, Pastori C, Frigerio S,
Today (Barc) 45:135154. Frassoni C, de Curtis M (2007) Expression of
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 171

adhesion factors induced by epileptiform activ- McKnight K, Jiang Y, Hart Y, Cavey A, Wroe S,
ity in the endothelium of the isolated guinea pig Blank M, Shoenfeld Y, Vincent A, Palace J,
brain in vitro. Epilepsia 48:743751. Lang B (2005) Serum antibodies in epilepsy
Lowry CA, Hale MW, Evans AK, Heerkens J, Staub and seizure-associated disorders. Neurology
DR, Gasser PJ, Shekhar A (2008) Serotonergic 65:17301736.
systems, anxiety, and affective disorder: Focus Mendez MF, Cummings JL, Benson DF (1986)
on the dorsomedial part of the dorsal raphe Depression in epilepsy: Significance and phe-
nucleus. Ann NY Acad Sci 1148:8694. nomenology. Arch Neurol 43:766770.
Majoie HJ, de Baets M, Renier W, Lang B, Vincent A Michelsen KA, Prickaerts J, Steinbusch HW
(2006) Antibodies to voltage-gated potassium (2008) The dorsal raphe nucleus and sero-
and calcium channels in epilepsy. Epilepsy Res tonin:Implications for neuroplasticity linked to
71:135141. major depression and Alzheimers disease. Prog
Manto M, Dalmau J, Didelot A, Rogemond V, Brain Res 172:233264.
Honnorat J (2010) In vivo effects of antibodies Minghetti L (2005) Role of inflammation in neu-
from patients with anti-NMDA receptor enceph- rodegenerative diseases. Curr Opin Neurol
alitis:Further evidence of synaptic glutamatergic 18:315321.
dysfunction. Orphanet J Rare Dis5:31. Molina-Hernandez M, Tellez-Alcantara NP,
Marchi N, Fan Q, Ghosh C, Fazio V, Bertolini F, Perez-Garcia J, Olivera-Lopez JI, Jaramillo-Jaimes
Betto G, Batra A, Carlton E, Najm I, Granata T, MT (2008) Antidepressant-like actions of
Janigro D (2009) Antagonism of peripheral minocycline combined with several glutamate
inflammation reduces the severity of status epi- antagonists. Prog Neuropsychopharmacol Biol
lepticus. Neurobiol Dis 33:171181. Psychiatry 32:380386.
Maroso M, Balosso S, Ravizza T, Iori V, Wright CI, Moy SS, Nadler JJ (2008) Advances in behav-
French J, Vezzani A (2011) Interleukin-1 bio- ioral genetics: mouse models of autism. Mol
synthesis inhibition reduces acute seizures and Psychiatry 13:426.
drug resistant chronic epileptic activity in mice. Muller CJ, Groticke I, Bankstahl M, Loscher W (2009)
Neurotherapeutics 8:304315. Behavioral and cognitive alterations, spontane-
Maroso M, Balosso S, Ravizza T, Liu J, Aronica E, ous seizures, and neuropathology developing
Iyer AM, Rossetti C, Molteni M, Casalgrandi M, after a pilocarpine-induced status epilepticus in
Manfredi AA, Bianchi ME, Vezzani A (2010) C57BL/6 mice. Exp Neurol 219:284297.
Toll-like receptor 4 and high-mobility group Muller N, Myint AM, Schwarz MJ (2011)
box-1 are involved in ictogenesis and can Inflammatory biomarkers and depression.
be targeted to reduce seizures. Nat Med Neurotox Res 19:308318.
16:413419. National Institutes of Health, National Institute
Mazarati A, Maroso M, Iori V, Vezzani A, Carli M of Neurological Disorders and Stroke (2007)
(2011) High-mobility group box-1 impairs mem- Curing epilepsy 2007: Translating discover-
ory in mice through both toll-like receptor 4 and ies into therapies. http://www.ninds.nih.gov/
receptor for advanced glycation end products. research/epilepsyweb/curingepilepsy/index.htm.
Exp Neurol 232:143148. National Institutes of Health, National Institute
Mazarati AM, Pineda E, Shin D, Tio D, Taylor AN, of Neurological Disorders and Stroke (2013)
Sankar R (2010) Comorbidity between epi- Curing the epilepsies 2013: Pathways forward.
lepsy and depression: Role of hippocampal https://meetingsnindsnihgov/indexcfm?event=e
interleukin-1. Neurobiol Dis 37:461467. xtra&ID=4069&tabID=4087.
Mazarati A, Shin D, Auvin S, Caplan R, Sankar R No FM, Polascheck N, Frigerio F, Bankstahl M,
(2007) Kindling epileptogenesis in immature Ravizza T, Marchini S, Beltrame L, Bandero CR,
rats leads to persistent depressive behavior. Loscher W, Vezzani A (2013) Pharmacological
Epilepsy Behav 10:377383. blockade of IL-1/IL-1 receptor type 1 axis dur-
Mazarati A, Siddarth P, Baldwin RA, Shin D, Caplan R, ing epileptogenesis provides neuroprotection
Sankar R (2008) Depression after status epilep- in two rat models of temporal lobe epilepsy.
ticus: Behavioural and biochemical deficits and Neurobiol Dis 59:183193.
effects of fluoxetine. Brain 131:20712083. Noebels J (2011) Aperfect storm:Converging paths
Mazarati AM, Shin D, Kwon YS, Bragin A, Pineda E, of epilepsy and Alzheimers dementia intersect in
Tio D, Taylor AN, Sankar R (2009) Elevated the hippocampal formation. Epilepsia 52 Suppl
plasma corticosterone level and depressive 1:3946.
behavior in experimental temporal lobe epilepsy. Ordway GA, Schenk J, Stockmeier CA, May W,
Neurobiol Dis 34:457461. Klimek V (2003) Elevated agonist binding to
172 Part II:Homeostatic Control

alpha2-adrenoceptors in the locus coeruleus in Ravizza T, Balosso S, Vezzani A (2011) Inflammation


major depression. Biol Psychiatry 53:315323. and prevention of epileptogenesis. Neurosci Lett
Oskvig DB, Elkahloun AG, Johnson KR, Phillips TM, 497:223230.
Herkenham M (2012) Maternal immune activation Ravizza T, Boer K, Redeker S, Spliet WG, van Rijen
by LPS selectively alters specific gene expression PC, Troost D, Vezzani A, Aronica E (2006a) The
profiles of interneuron migration and oxidative IL-1 system in epilepsy-associated malforma-
stress in the fetus without triggering a fetal immune tions of cortical development. Neurobiol Dis
response. Brain Behav Immun 26:623634. 24:128143.
Pardo CA, Vining EP, Guo L, Skolasky RL, Carson Ravizza T, Gagliardi B, Noe F, Boer K, Aronica E,
BS, Freeman JM (2004) The pathology of Vezzani A (2008a) Innate and adaptive immu-
Rasmussen syndrome:Stages of cortical involve- nity during epileptogenesis and spontaneous sei-
ment and neuropathological studies in 45 hemi- zures: Evidence from experimental models and
spherectomies. Epilepsia 45:516526. human temporal lobe epilepsy. Neurobiol Dis
Parsadaniantz SM, Batsche E, Gegout-Pottie P, 29:142160.
Terlain B, Gillet P, Netter P, Kerdelhue B (1997) Ravizza T, Lucas SM, Balosso S, Bernardino L, Ku
Effects of continuous infusion of interleukin 1 G, Noe F, Malva J, Randle JC, Allan S, Vezzani
on corticotropin-releasing hormone (CRH), CRH A (2006b) Inactivation of caspase-1 in rodent
receptors, proopiomelanocortin gene expression brain:Anovel anticonvulsive strategy. Epilepsia
and secretion of corticotropin, -endorphin and 47:11601168.
corticosterone. Neuroendocrinology 65:5363. Ravizza T, No F, Zardoni D, Vaghi V, Sifringer
Patterson PH (2002) Maternal infection:Window on M, Vezzani A (2008b) Interleukin converting
neuroimmune interactions in fetal brain devel- enzyme inhibition impairs kindling epileptogen-
opment and mental illness. Curr Opin Neurobiol esis in rats by blocking astrocytic IL-1 produc-
12:115118. tion. Neurobiol Dis 31:327333.
Patterson PH (2009) Immune involvement in schizo- Riazi K, Galic MA, Pittman QJ (2010) Contributions
phrenia and autism:Etiology, pathology and ani- of peripheral inflammation to seizure suscepti-
mal models. Behav Brain Res 204:313321. bility:Cytokines and brain excitability. Epilepsy
Patterson PH (2011a) Maternal infection and autism. Res 89:3442
Brain Behav Immun393. Richardson-Jones JW, Craige CP, Guiard BP,
Patterson PH (2011b) Modeling autistic features in Stephen A, Metzger KL, Kung HF, Gardier
animals. Pediatr Res 69:34R40R. AM, Dranovsky A, David DJ, Beck SG, Hen R,
Pernhorst K, Herms S, Hoffmann P, Cichon S, Leonardo ED (2010) 5-HT1A autoreceptor levels
Schulz H, Sander T, Schoch S, Becker AJ, Grote determine vulnerability to stress and response to
A (2013) TLR4, ATF-3 and IL8 inflammation antidepressants. Neuron 65:4052.
mediator expression correlates with seizure fre- Rojas A, Jiang J, Ganesh T, Yang MS, Lelutiu N,
quency in human epileptic brain tissue. Seizure Gueorguieva P, Dingledine R (2014)
22:675678. Cyclooxygenase-2 in epilepsy. Epilepsia 55:1725
Pineda EA, Hensler JG, Sankar R, Shin D, Burke TF, Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg
Mazarati AM (2011) Plasticity of presynaptic AA, Stewart JW, Warden D, Niederehe G, Thase
and postsynaptic serotonin 1A receptors in an ME, Lavori PW, Lebowitz BD, McGrath PJ,
animal model of epilepsy-associated depression. Rosenbaum JF, Sackeim HA, Kupfer DJ, Luther J,
Neuropsychopharmacology 36:13051316. Fava M (2006) Acute and longer-term outcomes
Pineda EA, Hensler JG, Sankar R, Shin D, Burke TF, in depressed outpatients requiring one or sev-
Mazarati AM (2012) Interleukin-1 causes eral treatment steps: A STAR*D report. Am J
fluoxetine resistance in an animal model of Psychiatry 163:19051917.
epilepsy-associated depression. Neurotherapeutics Sankar R, Holmes GL (2004) Mechanisms of
9:477485. action for the commonly used antiepileptic
Pineda E, Shin D, You SJ, Auvin S, Sankar R, drugs:Relevance to antiepileptic drug-associated
Mazarati A (2013) Maternal immune activation neurobehavioral adverse effects. J Child Neurol
promotes hippocampal kindling epileptogenesis 19 Suppl 1:S6S14.
in mice. Ann Neurol 74:1119. Sayyah M, Javad-Pour M, Ghazi-Khansari M
Ponzio NM, Servatius R, Beck K, Marzouk A, Kreider (2003) The bacterial endotoxin lipopolysaccha-
T (2007) Cytokine levels during pregnancy influ- ride enhances seizure susceptibility in mice:
ence immunological profiles and neurobehav- Involvement of proinflammatory factors: Nitric
ioral patterns of the offspring. Ann NY Acad Sci oxide and prostaglandins. Neuroscience
1107:118128. 122:10731080.
Inflammation and Immunomodulation in Epilepsy and Its Comorbidities 173

Scantlebury MH, Galanopoulou AS, Chudomelova L, Vezzani A, Balosso S, Ravizza T (2008) The role of
Raffo E, Bncourth D, Moshe SL (2010) Amodel cytokines in the pathophysiology of epilepsy.
of symptomatic infantile spasms syndrome. Brain Behav Immun 22:797803.
Neurobiol Dis 37:604612. Vezzani A, Balosso S, Ravizza T (2012) Inflammation
Schwartzer JJ, Careaga M, Onore CE, Rushakoff JA, and epilepsy. Handb Clin Neurol 107:163175.
Berman RF, Ashwood P (2013) Maternal immune Vezzani A, Conti M, De Luigi A, Ravizza T,
activation and strain specific interactions in the Moneta D, Marchesi F, De Simoni MG (1999)
development of autism-like behaviors in mice. Interleukin-1 immunoreactivity and microg-
Transl Psychiatry 3:e240. lia are enhanced in the rat hippocampus by
Seidenberg M, Pulsipher DT, Hermann B (2009) focal kainate application: Functional evidence
Association of epilepsy and comorbid condi- for enhancement of electrographic seizures. J
tions. Future Neurol 4:663668. Neurosci 19:50545065.
Shi L, Fatemi SH, Sidwell RW, Patterson PH (2003) Vezzani A, French J, Bartfai T, Baram TZ (2011a) The
Maternal influenza infection causes marked role of inflammation in epilepsy. Nat Rev Neurol
behavioral and pharmacological changes in the 7:3140.
offspring. J Neurosci 23:297302. Vezzani A, Friedman A (2011) Brain inflamma-
Smith SE, Li J, Garbett K, Mirnics K, Patterson PH tion as a biomarker in epilepsy. Biomark Med
(2007) Maternal immune activation alters fetal 5:607614.
brain development through interleukin-6. J Vezzani A, Janigro D (2009) Leukocyte-endothelial
Neurosci 27:1069510702. adhesion mechanisms in epilepsy: Cheers and
Stellwagen D, Beattie EC, Seo JY, Malenka RC (2005) jeers. Epilepsy Curr 9:118121.
Differential regulation of AMPA receptor and Vezzani A, Maroso M, Balosso S, Sanchez MA,
GABA receptor trafficking by tumor necrosis Bartfai T (2011b) IL-1 receptor/Toll-like receptor
factor-alpha. J Neurosci 25:32193228. signaling in infection, inflammation, stress and
Stellwagen D, Malenka RC (2006) Synaptic scal- neurodegeneration couples hyperexcitability and
ing mediated by glial TNF-alpha. Nature seizures. Brain Behav Immun 25:12811289.
440:10541059. Vezzani A, Moneta D, Conti M, Richichi C,
Tarr AJ, McLinden KA, Kranjac D, Kohman RA, Ravizza T, De Luigi A, De Simoni MG, Sperk
Amaral W, Boehm GW (2011) The effects of age G, Andell-Jonsson S, Lundkvist J, Iverfeldt K,
on lipopolysaccharide-induced cognitive deficits Bartfai T (2000) Powerful anticonvulsant action
and interleukin-1 expression. Behav Brain Res of IL-1 receptor antagonist on intracerebral
217:481485. injection and astrocytic overexpression in mice.
Teather LA, Packard MG, Bazan NG (2002) Proc Natl Acad Sci USA 97:1153411539.
Post-training cyclooxygenase-2 (COX-2) inhibi- Vezzani A, Moneta D, Richichi C, Aliprandi M,
tion impairs memory consolidation. Learn Mem Burrows SJ, Ravizza T, Perego C, De Simoni
9:4147. MG (2002) Functional role of inflammatory
Thieben MJ, Lennon VA, Boeve BF, Aksamit AJ, cytokines and antiinflammatory molecules in
Keegan M, Vernino S (2004) Potentially revers- seizures and epileptogenesis. Epilepsia 43 Suppl
ible autoimmune limbic encephalitis with neu- 5:3035.
ronal potassium channel antibody. Neurology Vezzani A, Viviani B (2014) Neuromodulatory prop-
62:11771182. erties of inflammatory cytokines and their impact
Tuchman R, Rapin I (2002) Epilepsy in autism. on neuronal excitability. Neuropharmacology.
Lancet Neurol 1:352358. doi: 10.1016/j.neuropharm.2014.10.027. [Epub
Varadkar S, Bien CG, Kruse CA, Jensen FE, Bauer J, ahead ofprint]
Pardo CA, Vincent A, Mathern GW, Cross JH Vianello M, Bisson G, Dal Maschio M, Vassanelli S,
(2014) Rasmussens encephalitis: Clinical fea- Girardi S, Mucignat C, Fountzoulas K,
tures, pathobiology, and treatment advances. Giometto B (2008) Increased spontaneous
Lancet Neurol 13:195205 activity of a network of hippocampal neurons
Vargas DL, Nascimbene C, Krishnan C, Zimmerman in culture caused by suppression of inhibitory
AW, Pardo CA (2005) Neuroglial activation and potentials mediated by anti-gad antibodies.
neuroinflammation in the brain of patients with Autoimmunity 41:6673.
autism. Ann Neurol 57:6781. Vincent A, Buckley C, Schott JM, Baker I,
Vezzani A, Aronica E, Mazarati A, Pittman QJ Dewar BK, Detert N, Clover L, Parkinson
(2013) Epilepsy and brain inflammation. Exp A, Bien CG, Omer S, Lang B, Rossor
Neurol 244:1121. MN, Palace J (2004) Potassium channel
174 Part II:Homeostatic Control

antibody-associated encephalopathy:Apoten- Yamagata K, Andreasson KI, Kaufmann WE,


tially immunotherapy-responsive form of lim- Barnes CA, Worley PF (1993) Expression of a
bic encephalitis. Brain 127:701712. mitogen-inducible cyclooxygenase in brain neu-
Vincent A, Crino PB (2011) Systemic and neurologic rons:Regulation by synaptic activity and gluco-
autoimmune disorders associated with seizures corticoids. Neuron 11:371386.
or epilepsy. Epilepsia 52 Suppl 3:1217. Yin P, Liu J, Li Z, Wang YY, Qiao NN, Huang
Vincent A, Irani SR (2010) Caspr2 antibodies SY, Li BM, Sun RP (2013) Prenatal immune
in patients with thymomas. J Thorac Oncol challenge in rats increases susceptibility to
5:S277280. seizure-induced brain injury in adulthood.
Vincent A, Irani SR, Lang B (2011) Potentially patho- Brain Res 1519:7886.
genic autoantibodies associated with epilepsy Zattoni M, Mura ML, Deprez F, Schwendener RA,
and encephalitis in children and adults. Epilepsia Engelhardt B, Frei K, Fritschy JM (2011) Brain
52 Suppl 8:811. infiltration of leukocytes contributes to the
Vincent A, Lily O, Palace J (1999) Pathogenic auto- pathophysiology of temporal lobe epilepsy. J
antibodies to neuronal proteins in neurological Neurosci 31:40374050.
disorders. J Neuroimmunol 100:169180. Zurolo E, Iyer A, Maroso M, Carbonell C, Anink
Viviani B, Bartesaghi S, Gardoni F, Vezzani A, Behrens JJ, Ravizza T, Fluiter K, Spliet GWM, van Rijen
MM, Bartfai T, Binaglia M, Corsini E, Di Luca M, PC, Vezzani A, Aronic E (2011) Activation of
Galli CL, Marinovich M (2003) Interleukin-1 TLR, RAGE and HMGB1 signaling in mal-
enhances NMDA receptor-mediated intracellu- formations of cortical development. Brain
lar calcium increase through activation of the Src 134:10151032.
family of kinases. J Neurosci 23:86928700. Wang N, Mi X, Gao B, Gu J, Wang W, Zhang Y, Wang
Weinberg MS, Blake BL, McCown TJ (2013) X (2015) Minocycline inhibits brain inflam-
Opposing actions of hippocampus TNFalpha mation and attenuates spontaneous recurrent
receptors on limbic seizure susceptibility. Exp seizures following pilocarpine-induced status
Neurol 247:429437. epilepticus. Neuroscience 287:144156.
11
Neuroplasticity
HIROYUKIOKUNO

OVERVIEW et al., 2004; Kandel, 2001). At the most micro-


The neural networks of the brain are not static scopic level, neuroplasticity is achieved at the
but rather change dynamically throughout an synapse and is referred to as synaptic plasticity.
individuals lifetime. Neuroplasticity is the abil- Synaptic plasticity, which modifies the efficiency
ity of a neural network to change in response to of synaptic transmission (synaptic weight) accord-
the environment. At each synapse, the efficiency ing to the history of synaptic input, can be clas-
of synaptic transmission is adaptively modulated sified into several types depending on the mode
based on the historic pattern of synaptic activity. of action (Bliss and Collingridge, 2013; Huganir
The overall synaptic weight of individual neurons and Nicoll, 2014). Long-term potentiation (LTP)
is homeostatically regulated by compensatory and long-term depression (LTD) are the two
mechanisms to keep cellular excitability at stable most-studied types of synaptic plasticity (see fol-
levels. Such synaptic plasticity serves to maintain lowing discussion). Whereas LTP and LTD act on
the excitation-inhibition balance within a neu- specific synapses that receive plasticity-inducing
ral network and provides pivotal mechanisms synaptic inputs, other types of synaptic plastic-
underlying higher cognitive functions includ- ity affect all synapses of the cell. One type of
ing learning and memory. At the network level, cell-wide plasticity is homeostatic synaptic scal-
neuroplasticity is critically involved in functional ing. It is now widely thought that at each synapse,
recovery from brain damage or loss of sensory the net synaptic weight is determined by the
information. Malfunction of neuroplasticity may summation of input-specific synaptic plasticity
lead to various types of brain disorders, includ- and homeostatic synaptic scaling (Davis, 2006;
ing addiction, psychiatric abnormalities, and Turrigiano, 2008). At more macroscopic levels,
cognitive impairment. neuroplasticity involves changes in cellular and
network excitability, as well as neural network
INTRODUCTION connectivity, that lead to cortical reorganization
The brain shows a huge capacity to coordinate and remapping (Buonomano and Merzenich,
complex body movements during exercise, to 1998; Hensch, 2004; Shah etal.,2010).
store personal experiences and knowledge dur- In this chapter I first describe several cat-
ing life, and to manage functional recovery from egories of synaptic plasticity and its molecular
brain damage caused by injury or stroke. The mechanisms. Ithen present an overview of neu-
mechanisms underlying such plastic and homeo- roplasticity at the cellular/circuit levels. Finally,
static abilities of the brain are of interest not only I discuss malfunctions of neuroplasticity as a
to researchers but also to the general public, and possible cause of brain disorders and neuropa-
understanding the mechanisms underlying this thology. Although neuroplasticity is fundamen-
plasticity has been one of the central themes in tal and observed in a wide variety of species, here
neuroscience. Neuroplasticity is a comprehensive I focus on vertebrate/mammalian nervous sys-
term that encompasses changes in synaptic func- tems, unless otherwise mentioned.
tion, cellular excitability, and network connectiv-
ity in response to information about changes in NEUROPLASTICITY
the internal and external environment (Cooke AT S Y N A P T I C L E V E L S
and Bliss, 2006). Neuroplasticity occurs in The efficiency of synaptic transmission can be
various areas and at various levels of the brain either strengthened or weakened in response to
(Buonomano and Merzenich, 1998; Chklovskii particular patterns of synaptic inputs, and such
176 Part II:Homeostatic Control

alternations can be sustained for a short time (A) Synaptic


(second to minutes) or can be long-lasting (hours Long-term potentiation vesicles
HFS (LTP) AMPA-R
to days, weeks, or months). Synaptic plasticity
can also be classified by local (input-specific) Pre
or global (cell-wide) phenomena. Although EPSP
each type of synaptic plasticity involves distinct
molecular and cellular mechanisms, it is gener- Post
ally accepted that the efficiency of synaptic trans- (B)
Long-term depression
mission is primarily determined by the number LFS (LTD)
of receptors for neurotransmitters that are pre-
EPSP
sent on the postsynaptic membrane (Huganir
and Nicoll, 2014). Here Isummarize several types
of input-specific and cell-wide synaptic plasticity.
(C)
Long-Term Potentiation HFS
Heterosynaptic LTD
LTP is one of the most-studied input-specific
synaptic modifications (Bliss and Collingridge, EPSP
1993, 2013). LTP is an enhancement of synap-
tic transmission efficiency that endures for a
long time (Bliss and Gardner-Medwin, 1973;
Bliss and Lomo, 1973; Douglas and Goddard, EPSP
1975; Figure 11.1A). As Hebb (1949) proposed,
when a presynaptic cell Arepeatedly or persis-
FIGURE 11.1: Several types of input-specific
tently takes part in firing a postsynaptic cell B,
long-term synaptic plasticity. (A) Long-term
the synaptic efficiency of cell B from cell A is
potentiation (LTP): Strong high-frequency synaptic
increased. This coincidence of pre- and post-
stimulation (HFS) (e.g., 100 Hz, 100 pulses) to presyn-
synaptic activation ensures input specificity of
apses evokes long-lasting (> hours) enhancement of
LTP because the synaptic efficiencies of all other
excitatory postsynaptic potentials (EPSP) at the postsyn-
postsynapses connected with nonstimulated
apses. The coincidence of presynaptic and postsynaptic
cells are unchanged. The temporal order and
activation is required for induction of LTP (Hebbs rule)
timing of presynaptic and postsynaptic activa-
and the synaptic changes are spatially restricted to the
tion are critical determinants for the synaptic
site of activation (input-specificity, i.e., homosynaptic
efficiency; the sign and amplitude of changes in
expression). (B)Long-term depression (LTD):In contrast
synaptic transmission are precisely controlled
to LTP, low-frequency synaptic stimulation (LFS) (e.g.,
with the range of tens of milliseconds, a mecha-
1 Hz, 900 pulses) to presynapses induces long-lasting
nism known as spike-timing dependent plastic-
(> hours) reduction of EPSP at the postsynapses. This
ity (Magee and Johnston, 1997; Markram etal.,
type of LTD also follows Hebbs rule and is homosynap-
1997). Interestingly, while LTP is primarily
tic. (C)Heterosynaptic LTD:In some cases, homosynap-
expressed at the site that receives strong repeti-
tic synaptic plasticity also involves changes in synaptic
tive synaptic inputs, it can also be established
efficiency at the synapses that are not directly stimulated.
at other synaptic sites if the synapses receive
This is called heterosynaptic plasticity. Heterosynaptic
weak synaptic stimulation that per se is insuf-
LTD is observed in association with homosynaptic LTP
ficient to induce LTP concurrently with the
in several brain areas and neural networks. AMPA-R;
LTP-inducing strong stimuli. This property is
AMPA-type glutamate receptor.
called associativity of LTP. LTP also is coop-
erative in the sense that, even though a single
presynaptic fiber with weak stimulation fails to focal high-frequency stimulation with a micro-
elicit LTP, many of such weakly activated fibers electrode to presynaptic fibers (Bliss and Lomo,
can cooperatively induce LTP if the presynaptic 1973; Gustafsson etal., 1987; Kauer etal., 1988),
inputs converge on a postsynaptic region to pro- spike-timing dependent plasticity protocols
vide sufficient postsynaptic depolarization. The (Magee and Johnston, 1997; Markram et al.,
associativity and cooperativity together govern 1997; Zhang etal., 1998)or postsynaptic depo-
input specificity of LTP. Selective establishment larization with optically controlled repeated
of LTP at a subset of synapses, but not at other glutamate application (Harvey and Svoboda,
sets of synapses, can be induced by applying 2007; Matsuzaki etal.,2004).
Neuroplasticity 177

There are various types of LTP: these oper- the hippocampus interferes with spatial mem-
ate by distinct molecular mechanisms, depend- ory formation in the Morris water-maze task
ing on the cell type, inputs, and brain areas. (Barnes etal., 1994; Moser etal., 1998). Conversely,
Likely the most-studied LTP is N-Methyl- learning-induced LTP occludes subsequent induc-
d-aspartate (NMDA)-type glutamate recep- tion of LTP in the hippocampus (Whitlock
tor (NMDA-R)-dependent LTP, which occurs et al., 2006) and in the primary motor cortex
at excitatory synapses in the CA1 hippocam- (Rioult-Pedotti et al., 2000). Furthermore, many
pal region (Shaffer collateral-CA1 pyramidal genetically modified mice that are deficient in LTP
cell connections; Bliss and Collingridge, 2013). show reduced/impaired memory formation (Abel
This Hebbian-type synaptic plasticity is now etal., 1997; Bourtchuladze etal., 1994; Grant etal.,
understood as follows: high-frequency stimu- 1992; Lee etal., 2003; Plath etal., 2006; Silva etal.,
lation, such as 100 pulses at 100 Hz, induces 1992a). These observations, together with theoreti-
glutamate release from presynaptic sites. The neu- cal considerations and modeling, strongly support
rotransmitter activates postsynaptic -amino- the idea that LTP is a cellular mechanism that is cru-
3-hydroxy-5-methyl-4-isoxazolepropionic acid cial for learning and memory (Neves etal.,2008).
(AMPA)-type glutamate receptors (AMPA-Rs),
as well as NMDA-Rs. The activated NMDA-Rs Long-Term Depression
permit Ca 2+ influx, which subsequently activates LTD is a synaptic plasticity mechanism that func-
chemical reaction cascades at the postsynaptic tions in the opposite fashion to LTP. The syn-
site to enhance the number of AMPA-Rs on the apses that receive a particular frequency range
postsynaptic surface. Anumber of pathways and of stimulation show long-lasting decreases in
molecules have been suggested to contribute to synaptic transmission efficiency (Dudek and
the NMDA-dependent LTP process (Bliss and Bear, 1992; Luscher and Huber, 2010; Malinow
Collingridge, 2013; Huganir and Nicoll, 2014). and Malenka, 2002; Mulkey and Malenka,
Among them, Ca2+/Calmodulin-dependent pro- 1992). For example, low-frequency stimulation,
tein kinase II (CaMKII) is one of the most prom- such as 1 Hz for 15 min, induces LTD in Shaffer
ising key molecules. Pharmacological inhibition collateral-CA1 pyramidal cell synapses (Dudek
or genetic disruption of CaMKII impairs hip- and Bear, 1993) (Figure 11.1B). This synaptic
pocampal LTP (Malinow etal., 1988; Otmakhov modification also requires coincident activa-
et al., 1997; Otmakhov et al., 2004; Silva et al., tion of pre- and postsynapses (i.e., Hebbian-type
1992b). While many works dealing with LTP plasticity). As in the case of LTP, several types
focus on excitatory synapses between excita- of LTD mechanisms have been identified. Two
tory neurons (glutamatergic neurons), others major types of LTD mechanisms are NMDA-R
have investigated LTP at excitatory synapses of dependent and metabotropic glutamate recep-
inhibitory neurons (gamma-aminobutyric acid tor (mGluR) dependent (Bear and Abraham,
(GABA)-ergic neurons). Interestingly, in contrast 1996). In both cases, a reduction in AMPA-Rs at
to previously described NMDA-R-dependent the postsynaptic membrane is thought to be the
Hebbian LTP, which requires postsynaptic depo- principal molecular mechanism of LTD expres-
larization for its induction, some forms of LTP sion at excitatory synapses in the central nervous
in inhibitory neurons critically depend on post- system. Although the mechanism of the AMPA-R
synaptic hyperpolarization, instead of depolari- reduction in LTD is not fully understood, several
zation. Because this type of LTP is blocked by possible mechanisms, including the regulation
conventional postsynaptic activation during its of endocytosis and diffusion of AMPA-Rs on the
induction, it is called anti-Hebbian LTP (Lamsa plasma membrane, have been proposed (Man
etal., 2007; Kullmann and Lamsa,2007). etal., 2000; Wang and Linden, 2000; Waung etal.,
Theoretical studies have proposed that LTP 2008; Xia etal.,2000).
is the cellular substrate of memory (Morris In the cerebellum, a center of motor coordi-
et al., 2003). Several lines of experimental evi- nation and postural control, LTD occurs at excit-
dence have also demonstrated the involvement atory synapses between parallel fibers (PFs) and
of LTP in memory-formation processes. First, Purkinje cells (Ito, 1989). Purkinje cells receive
learning-induced LTP has been observed in two types of inputs:one from PFs from cerebel-
the CA1 region during a passive avoidance task lar granule cells and the other from the climb-
(Whitlock et al., 2006) and the amygdala after ing fibers from the inferior olive nucleus. When
a fear-conditioning paradigm (Nabavi et al., Purkinje cells receive simultaneous inputs from
2014) in free-moving rats. Saturation of LTP in PFs and climbing fibers, PF-mediated synaptic
178 Part II:Homeostatic Control

responses decrease in an mGluR1-dependent likely have impacts on areas beyond single syn-
manner (Aiba et al., 1994; Kano et al., 1997). apses. Indeed, D-serine and ATP released from
Because mGluR1 is a G-protein-coupled metabo- glial cells have been implicated in LTP on multi-
tropic receptor and is coupled with phospholipase ple neurons (Henneberger etal., 2010)and heter-
C, the mGluR1 activation results in an intracel- osynaptic LTD (Pascual etal., 2005), respectively.
lular Ca2+ increase as well as in protein kinase
C (PKC) activation via IP3/diacyl-glycerol second Synaptic Tagging and
messenger systems. Cerebellar LTD is thought to Inverse Tagging
play critical roles in the vestibulo-ocular reflex The maintenance of long-term synaptic plastic-
and in motor learning (Ito, 2000; Lee etal., 2009; ity requires new synthesis of transcripts and pro-
Shibuki etal., 1996; Shutoh etal.,2006). teins (Costa-Mattioli et al., 2009; Kandel, 2001;
Kelleher et al., 2004; Okuno, 2011). This is espe-
Heterosynaptic LTP and cially true during a particular time window after
HeterosynapticLTD synapses receive plasticity-inducing stimuli, when
To date, the majority of studies that have newly synthesized plasticity-related proteins may
addressed long-term synaptic plasticity have functionally interact with local synaptic tags. The
focused on LTP and LTD at the synaptic site synaptic tagging and capture theory adopts a con-
where the plasticity-inducing stimuli were origi- ceptual framework in which activity-triggered
nally given. Because the site of change corre- local changes at synaptic sites (i.e., synaptic tagging)
sponds to the site of stimulation, this mode of permit the use of activity-induced plasticity-related
synaptic plasticity is called homosynaptic plastic- proteins at the cell body and dendrites (i.e.,
ity (homosynaptic LTP and homosynaptic LTD). plasticity-related protein capture) to stabilize and
However, neurons also have the ability to induce maintain changes in synaptic efficacy (Frey and
synaptic changes at sites other than the site of Morris, 1997; Martin et al., 2000; Figure 11.2A).
homosynaptic plasticity, in order to balance the Several expanded versions of this hypothesis have
initial input-specific synaptic change. For exam- been proposed and are experimentally supported
ple, when strong synaptic inputs induce LTP in (Fonseca etal., 2004; Sajikumar etal., 2007). The
a subset of synapses, another subset of synapses synaptic tagging and capture hypothesis explains
can undergo LTD accordingly (Abraham and how protein-synthesis-dependent LTP recon-
Goddard, 1983; Barry etal., 1996). Because this ciles input-specific synaptic modifications with
type of LTD occurs at synapses that have not the broad cellular distribution of newly synthe-
received plasticity-inducing inputs, it is called het- sized proteins that are required for LTP mainte-
erosynaptic LTD (Figure 11.1C). Heterosynaptic nance (Govindarajan et al., 2006; Redondo and
LTD can be envisioned as a homeostatic process Morris,2011).
to maintain local synaptic inputs at a constant An opposite, but nonmutually exclusive,
level after a set of synapses are potentiated by hypothesis is the inverse synaptic tagging model,
LTP-inducing stimuli (Abraham, 1996). This het- which assumes the existence of synaptic tags that
erosynaptic mechanism could also play a role in mark nonpotentiated synapses and play a role in
accentuating the contrast of synapses undergoing a process to prevent these synapses from being
LTP with surrounding unpotentiated synapses. strengthened (Figure 11.2B). A recent study has
Heterosynaptic LTP after induction of homosyn- indicated that an activity-dependent protein,
aptic LTD has also been observed in some brain called Arc, may be involved in such an inverse
areas (Nguyen, 2001; Royer and Pare,2002). synaptic tagging process (Okuno et al., 2012).
It is known that glial cells release diffusible When Arc is induced by LTP-eliciting stimuli in
factors, including D-serine, adenosine triphos- neurons, the newly synthesized Arc mainly accu-
phate (ATP), and tumor necrosis factor (TNF)- mulates at nonpotentiated synapses rather than
for cellcell communication (Ben Achour and at potentiated synapses. The levels of synaptic
Pascual, 2010; Fields and Burnstock, 2006; Perea Arc accumulation were found to be negatively
et al., 2009; Stellwagen and Malenka, 2006). correlated with the surface expression levels of
Those factors, called gliotransmitters, act not only AMPA-Rs (Chowdhury etal., 2006; Okuno etal.,
on glia themselves but also on neighboring neu- 2012; Shepherd etal., 2006). Because Arc induc-
rons to modulate synaptic functions (Perea etal., tion has been shown to correlate with ongoing
2009). Because individual astrocytes contact cognitive activity in the hippocampus and the
with hundreds of synapses of neighboring neu- cortex (Guzowski et al., 1999), and because Arc
rons, gliotransmitters released from an astrocyte absence causes severe memory disorders (Plath
Neuroplasticity 179

(A) Synaptic tagging where synapses receive plasticity-inducing stim-


uli. In contrast, other types of synaptic plastic-
ity involve synaptic modification in a cell-wide
manner (Burrone etal., 2002; Thiagarajan etal.,
Potentiated
synapse EPSP 2005). Synaptic scaling (also see later discussion)
is one such cell-wide mechanism that homeo-
statically modifies the strength of all synapses
Un-potentiated Un-potentiated in response to global changes in synaptic inputs
synapses synapses (Desai et al., 2002; Turrigiano et al., 1998). For
example, when the synaptic activity of cultured
neurons is chronically suppressed by the sodium
(B) Inverse synaptic tagging channel blocker tetrodotoxin, all synapses in the
neurons gain synaptic strength. In the opposite
scenario, chronic enhancement of synaptic activ-
ity with GABA receptor blockers in neuronal cul-
EPSP
Un-potentiated ture results in the reduction of synaptic efficiency
or
synapses at all synapses. This bidirectional change in each
synapse occurs in proportion to its initial strength
(i.e., a multiplicative function); thus this phe-
Potentiated nomenon is called synaptic scaling (Figure 11.3 ;
synapse Turrigiano, 2008). Synaptic scaling involves
several cell-wide mechanisms associated with
transcription and translation in the cell body,
FIGURE 11.2: Synaptic tagging and capture but it also includes synapse-specific components
model. (A) Synaptic tagging: During the induction
phase of long-term potentiation (LTP), plasticity-
inducing stimuli develop a putative signature (tag)
within the activated synapses (a red flag). This synap- EPSP

tic tag serves as a mark to distinguish the synapses to


be potentiated from other nonstimulated synapses and
stimulus-induced plasticity-related proteins (PRPs; red
circles) are then captured based on the presence of the
synaptic tags. The PRPs are required to maintain the
EPSP enhancement during the late phase of LTP. (B)
Inverse synaptic tagging: In contrast to the synaptic
tagging, the inverse synaptic tag (blue flags) is created EPSP
in synapses other than the synapse to be potentiated. Chronically
low activity
The stimulus-induced negative PRPs (blue circles) are
captured in the nonpotentiated synapses and play a role
in preventing undesired synaptic augmentation during Normal neuronal
the late phase ofLTP. activity

etal., 2006), the inverse synaptic tagging of Arc


may subserve memory consolidation by prevent- Chronically
high activity
ing undesired synaptic enhancement at weak
synapses while sparing potentiated synapses FIGURE 11.3: Homeostatic synaptic scaling.
(Kim et al., 2012; Okuno et al., 2012). Such an Contrary to homosynaptic plasticity, synaptic scaling
inverse synaptic tagging mechanism may also is a mechanism that influences all synapses in the cell.
participate in some forms of heterosynaptic LTD Through this homeostatic mechanism, the surface
(see previous discussion). expression levels of AMPA receptors are upregulated
at all synapses under conditions of chronically low
Synaptic Scaling neuronal activity (left), while surface AMPA receptor
The types of synaptic plasticity described pre- levels are downregulated with persistent high neuronal
viously are expressed in spatially limited areas activity (right).
180 Part II:Homeostatic Control

(Beique etal., 2011; Sutton etal., 2006). In gen- on parvalbumin-expressing inhibitory neurons
eral, synaptic scaling is thought to be advanta- (Chang et al., 2010). The Narp-mediated effect
geous for keeping neuronal excitability within a on parvalbumin neurons results in a homeostatic
certain dynamic range without affecting the rela- increase in inhibitory inputs within the neuronal
tive balance between strong and weak synapses network, suggesting that Narp mediates cell
(Turrigiano,2008). nonautonomous homeostatic regulation of the
Because activity-dependent gene expres- excitatory-inhibitory balance (Chang etal.,2010).
sion is critical for synaptic scaling and other
forms of synaptic homeostasis (Ibata et al., NEUROPLASTICITY
2008; Sutton et al., 2006), a number of labo- AT T H E N E T W O R K L E V E L
ratories have attempted to identify the genes In addition to modulations in synaptic weight and
involved. Several neuronal immediate-early cellular excitability, there are several additional
genes, a class of genes that is rapidly and mechanisms for changing network connectivity,
transiently induced by synaptic activity, have by generating new synapses and/or by pruning
been shown to critically contribute to syn- existing synapses. Such changes in connectivity
aptic homeostatic changes. The immediate can be most dynamically observed in the brain
early gene Arc is rapidly induced by synaptic after a drastic perturbation of sensoryinput.
activation in an NMDA-R-dependent man-
ner (Link etal., 1995; Lyford etal., 1995). This Cortical Reorganization
activity-regulated Arc expression and the role and Remapping
of Arc in promoting AMPA-R endocytosis con- The somatosensory area of the cerebral cor-
stitutes a cell-autonomous loop that accounts tex receives tactile information from each part
for the homeostatic control of AMPA-R sur- of the body in a topologically organized man-
face expression levels (Rial Verde et al., 2006; ner. When one part of the body is severely
Shepherd etal., 2006). When the levels of excit- damagedfor example, finger amputationthe
atory inputs increase, neurons express more area representing the finger suddenly loses sen-
Arc. The induced Arc then promotes AMPA-R sory input. However, as time goes by, the area
endocytosis at postsynaptic sites, resulting recovers neural activity by receiving sensory
in a reduction in responsiveness to excitatory input from the remaining fingers (Merzenich
inputs (Shepherd etal., 2006). By contrast, the et al., 1984). This phenomenon is called corti-
cells with suppressed synaptic inputs cease cal reorganization or cortical remapping and is
to express Arc; thus Arc-induced AMPA-R thought to be a compensatory adaptive response
endocytosis is reduced, resulting in increased to the loss of a bodypart.
cellular responsiveness. In this way, cellular Similarly, cortical reorganization is observed
responsiveness is homeostatically controlled in other sensory cortices. One of the most-studied
by activity-regulated Arc expression. Indeed, areas is the primary visual cortex. In primates,
Arc knockout neurons do not show homeostatic including humans, as well as many carnivorous
synaptic scaling both in vitro and in vivo (Gao mammals, visual inputs from two eyes are sepa-
et al., 2010; Shepherd et al., 2006). Consistent rately transmitted to and segregated within the
with this, a recent work has demonstrated that primary visual cortex (Hubel etal., 1976). When
Arc expression during synaptic scaling is reg- one eye is occluded for days to weeks in a par-
ulated by a protein kinase called MSK1, and, ticular time window during development (i.e., a
importantly, knockout of MSK1 results in loss critical period), neural activity in the visual areas
of synaptic scaling (Correa etal., 2012). Asimi- corresponding to each of the two eyes becomes
lar but distinct activity-dependent mechanism unbalanced. The area responding to the intact
with another immediate-early gene product, eye expands, while the cortical area correspond-
Homer1a, has been reported (Hu etal.,2010). ing to the occluded eye shrinks (Figure 11.4). This
Neuronal activity-regulated pentraxin (Narp) phenomenon is known as ocular dominance plas-
is also a major neuronal immediate-early gene ticity and was originally demonstrated in ani-
product. Unlike Arc and Homer1a, which are mal models, such as with cats and monkeys, but
postsynaptic proteins, Narp is a secreted protein has now been observed in many other species,
that localizes to excitatory synapses (OBrien including mice (Gordon and Stryker, 1996; LeVay
etal., 1999). Interestingly, activity-induced Narp etal., 1980; Shatz and Stryker, 1978). The molec-
predominantly accumulates, and facilitates the ular and cellular mechanisms underlying ocu-
recruitment of AMPA-Rs, at excitatory synapses lar dominance plasticity have been extensively
Neuroplasticity 181

plasticity (Keck etal., 2013). Hengen etal. (2013)


performed chronic multiunit recording from
the rat visual cortex and revealed that cellular
activity transiently dropped after eye-lid suture
(i.e., monocular deprivation) but that normal
Right
activity resumed by six days after the depriva-
tion (Hengen etal., 2013). The authors also dem-
Left Monocular onstrated that mEPSC amplitude was enhanced
deprivation at six days after deprivation in ex vivo prepa-
rations, indicating that synaptic scaling con-
trols the homeostatic process in this phase. It is
worth noting that such homeostatic responses
in the visual cortex after manipulation of vision
are mediated by Arc. Arc knockout mice fail
to exhibit visual experience-induced homeo-
static plasticity of mEPSC amplitude in the
Right visual cortex (Gao etal., 2010), as well as ocular
dominance plasticity that normally takes place
Left after monocular deprivation in wild-type mice
(McCurry etal.,2010).
FIGURE 11.4: Cortical plasticity after sensory-
input deprivation. Ocular dominance plasticity in NEUROPLASTICITY AND
the visual cortex. In many mammals such as primates N E U R O L O G I C A L / P S Y C H I AT R I C
and cats, the primary visual cortex is organized to DISORDERS
create the alternating columnar structures (blue and The cognitive and executive functions of the
red stripes) corresponding to each eye during normal brain largely rely on the proper control of syn-
development (top). When visual information from one apse efficiency and connectivity. Such synaptic
eye is deprived by eye-lid suture or injury, especially properties are controlled by the balance between
in a particular time window during development (criti- cell-wide (e.g., homeostatic synaptic scaling)
cal period), the cortical areas of the nondeprived eye and local (e.g., LTP/LTD) synaptic plasticity. So
become larger than those of the deprived eye (bottom). what impact does synaptic plasticity impairment
have on brain functions? A number of studies
have demonstrated that gene-knockout animals
investigated using a combination of electrophysi- that show LTP deficits in the hippocampus also
ology, optical imaging, genetics, and molecular exhibit impaired long-term memory forma-
biology in recent decades (Hensch,2004). tion (Abel et al., 1997; Bourtchuladze et al.,
1994; Grant et al., 1992; Lee et al., 2003; Plath
Homeostatic Response Recovery et al., 2006; Silva et al., 1992a). Hippocampal
Recent in vivo live imaging and electrophysi- LTP is also impaired in many animal models of
ological studies have demonstrated the dynam- Alzheimers disease (Auffret et al., 2009; Gong
ics of homeostatic responses after sensory input et al., 2004; Oddo et al., 2003). In the striatum,
manipulation in the visual cortex. Keck et al exaggerated LTP in dopaminergic neurons is
(2013), using in vivo 2-photon Ca 2+ imaging, associated with drug addiction (Luscher and
reported that neuronal activity in the visual cor- Malenka, 2011; Nestler et al., 1993). Moreover,
tex of mice transiently decreased after retinal dysregulation or malfunction of homeostatic
lesions deprived the cortex of visual input, but synaptic scaling is often associated with neuro-
the cortical activity gradually recovered over one pathological processes, including drug addiction
to two days after the lesioning (Keck etal., 2013). (Boudreau and Wolf, 2005), epilepsy(Chang etal.,
This homeostatic response was accompanied by 2010; Savin etal., 2009), and neurodevelopmen-
synaptic scaling (increases in mEPSC ampli- tal disorders such as fragile X (Soden and Chen,
tude) and spine-head enlargement in acute brain 2010)and Rett syndromes (Blackman etal., 2012;
preparations. Furthermore, although synaptic Qiu etal., 2012). These facts suggest that normal
scaling was observed during the early phase, brain functions require the appropriate balance
the authors also observed a reduction in inhibi- of cell-wide homeostatic and input-specific syn-
tory inputs during the late phase of homeostatic aptic plasticity.
182 Part II:Homeostatic Control

CONCLUDING REMARKS Aiba, A., Kano, M., Chen, C., Stanton, M.E.,
Brain flexibility is involved in numerous pro Fox, G.D., Herrup, K., Zwingman, T.A., and
cesses, such as learning and memory, motor Tonegawa, S. (1994). Deficient cerebellar
coordination, and recovery from injury, and this long-term depression and impaired motor learn-
flexibility is based mainly on neuroplastic mecha- ing in mGluR1 mutant mice. Cell 79, 377388.
nisms. In this chapter I described several repre- Auffret, A., Gautheron, V., Repici, M., Kraftsik, R.,
sentative forms of synaptic plasticity and cortical Mount, H.T., Mariani, J., and Rovira, C. (2009).
reorganization. Neuroplasticity is a broad term Age-dependent impairment of spine morphol-
that covers a variety of synaptic and nonsynaptic ogy and synaptic plasticity in hippocampal
plasticity mechanisms. Other neuroplastic mecha- CA1 neurons of a presenilin 1 transgenic mouse
nisms include structural plasticity, which modifies model of Alzheimers disease. J Neurosci 29,
the shape and size of synapses (Kasai etal., 2003; 1014410152.
Barnes,C.A.,Jung,M.W.,McNaughton,B.L.,Korol,D.L.,
Luo, 2002), intrinsic plasticity, which controls the
Andreasson, K., and Worley, P.F. (1994). LTP satu-
biophysical properties of the plasma membrane
ration and spatial learning disruption: Effects of
(Beck and Yaari, 2008), and metaplasticity, by
task variables and saturation levels. J Neurosci 14,
which the rules governing synaptic plasticity per se
57935806.
are dynamically modulated by prior synaptic activ-
Barres, B.A. (2008). The mystery and magic of glia: A
ity (Abraham and Tate, 1997; Hulme etal., 2013). perspective on their roles in health and disease.
Currently, however, the relationship between the Neuron 60, 430440.
different types of plasticity remains unclear. This Barry, M.F., Vickery, R.M., Bolsover, S.R., and
relationship should be clarified experimentally Bindman, L.J. (1996). Intracellular studies of het-
and theoretically in the near future. Furthermore, erosynaptic long-term depression (LTD) in CA1
accumulating evidence is now shedding light on of hippocampal slices. Hippocampus 6, 38.
the fundamental connections between neuroplas- Bear, M.F., and Abraham, W.C. (1996). Long-term
ticity and other research fields, such as epigenet- depression in hippocampus. Annu Rev Neurosci
ics (Guzman-Karlsson et al., 2014), glia (Barres, 19, 437462.
2008), and metabolism (Liu et al., 2013). In par- Beck, H., and Yaari, Y. (2008). Plasticity of intrinsic
ticular, adult neurogenesis recently emerged as neuronal properties in CNS disorders. Nat Rev
one of the central topics in neuroscience (Gage Neurosci 9, 357369.
and Temple, 2013). Several reports have indicated Beique, J.C., Na, Y., Kuhl, D., Worley, P.F.,
the involvement of adult neurogenesis in memory and Huganir, R.L. (2011). Arc-dependent
(Imayoshi etal., 2008; Kitamura etal., 2009; Sahay synapse-specific homeostatic plasticity. Proc
et al., 2011). Further studies of neuroplasticity Natl Acad Sci USA 108, 816821.
might lead to a comprehensive understanding of Ben Achour, S., and Pascual, O. (2010). Glia: The
our brains from the perspective of basic research many ways to modulate synaptic plasticity.
and provide clues to novel therapeutic approaches Neurochem Int 57, 440445.
to the many clinically unmet needs, including Blackman, M.P., Djukic, B., Nelson, S.B., and
memory impairment, addiction, psychiatric dis- Turrigiano, G.G. (2012). A critical and
orders, and rehabilitation after stroke. cell-autonomous role for MeCP2 in synaptic
scaling up. J Neurosci 32, 1352913536.
References Bliss, T.V., and Collingridge, G.L. (1993). A synaptic
Abel, T., Nguyen, P.V., Barad, M., Deuel, T.A., model of memory:Long-term potentiation in the
Kandel, E.R., and Bourtchouladze, R. (1997). hippocampus. Nature 361,3139.
Genetic demonstration of a role for PKA in the Bliss, T.V., and Collingridge, G.L. (2013). Expression
late phase of LTP and in hippocampus-based of NMDA receptor-dependent LTP in the hippo-
long-term memory. Cell 88, 615626. campus: Bridging the divide. Mol Brain6,5.
Abraham, W.C. (1996). Induction of heterosyn- Bliss, T.V., and Gardner-Medwin, A.R. (1973).
aptic and homosynaptic LTD in hippocampal Long-lasting potentiation of synaptic transmis-
sub-regions in vivo. J Physiol Paris 90, 305306. sion in the dentate area of the unanaestetized
Abraham, W.C., and Goddard, G.V. (1983). rabbit following stimulation of the perforant
Asymmetric relationships between homosyn- path. J Physiol 232, 357374.
aptic long-term potentiation and heterosynaptic Bliss, T.V., and Lomo, T. (1973). Long-lasting poten-
long-term depression. Nature 305, 717719. tiation of synaptic transmission in the den-
Abraham, W.C., and Tate, W.P. (1997). Metaplasticity: tate area of the anaesthetized rabbit following
Anew vista across the field of synaptic plasticity. stimulation of the perforant path. J Physiol 232,
Prog Neurobiol 52, 303323. 331356.
Neuroplasticity 183

Boudreau, A.C., and Wolf, M.E. (2005). Behavioral Dudek, S.M., and Bear, M.F. (1993). Bidirectional
sensitization to cocaine is associated with long-term modification of synaptic effective-
increased AMPA receptor surface expression in ness in the adult and immature hippocampus. J
the nucleus accumbens. J Neurosci 25, 91449151. Neurosci 13, 29102918.
Bourtchuladze, R., Frenguelli, B., Blendy, J., Cioffi, Fields, R.D., and Burnstock, G. (2006). Purinergic
D., Schutz, G., and Silva, A.J. (1994). Deficient signalling in neuron-glia interactions. Nat Rev
long-term memory in mice with a targeted muta- Neurosci 7, 423436.
tion of the cAMP-responsive element-binding Fonseca, R., Nagerl, U.V., Morris, R.G., and
protein. Cell 79,5968. Bonhoeffer, T. (2004). Competing for mem-
Buonomano, D.V., and Merzenich, M.M. (1998). ory:Hippocampal LTP under regimes of reduced
Cortical plasticity: From synapses to maps. protein synthesis. Neuron 44, 10111020.
Annu Rev Neurosci 21, 149186. Frey, U., and Morris, R.G. (1997). Synaptic tag-
Burrone, J., OByrne, M., and Murthy, V.N. (2002). ging and long-term potentiation. Nature 385,
Multiple forms of synaptic plasticity triggered 533536.
by selective suppression of activity in individual Gage, F.H., and Temple, S. (2013). Neural stem
neurons. Nature 420, 414418. cells: Generating and regenerating the brain.
Chang, M.C., Park, J.M., Pelkey, K.A., Grabenstatter, Neuron 80, 588601.
H.L., Xu, D., Linden, D.J., Sutula, T.P., McBain, C.J., Gao, M., Sossa, K., Song, L., Errington, L., Cummings,
and Worley, P.F. (2010). Narp regulates L., Hwang, H., Kuhl, D., Worley, P., and Lee, H.K.
homeostatic scaling of excitatory synapses on (2010). A specific requirement of Arc/Arg3.1 for
parvalbumin-expressing interneurons. Nat visual experience-induced homeostatic synap-
Neurosci 13, 10901097. tic plasticity in mouse primary visual cortex. J
Chklovskii, D.B., Mel, B.W., and Svoboda, K. (2004). Neurosci 30, 71687178.
Cortical rewiring and information storage. Gong, B., Vitolo, O.V., Trinchese, F., Liu, S., Shelanski,
Nature 431, 782788. M., and Arancio, O. (2004). Persistent improve-
Chowdhury, S., Shepherd, J.D., Okuno, H., Lyford, G., ment in synaptic and cognitive functions in an
Petralia, R.S., Plath, N., Kuhl, D., Huganir, R.L., Alzheimer mouse model after rolipram treat-
and Worley, P.F. (2006). Arc/Arg3.1 interacts ment. J Clin Invest 114, 16241634.
with the endocytic machinery to regulate AMPA Gordon, J.A., and Stryker, M.P. (1996).
receptor trafficking. Neuron 52, 445459. Experience-dependent plasticity of binocular
Cooke, S.F., and Bliss, T.V. (2006). Plasticity in the responses in the primary visual cortex of the
human central nervous system. Brain 129, mouse. J Neurosci 16, 32743286.
16591673. Govindarajan, A., Kelleher, R.J., and Tonegawa, S.
Correa, S.A., Hunter, C.J., Palygin, O., Wauters, S.C., (2006). A clustered plasticity model of long-term
Martin, K.J., McKenzie, C., McKelvey, K., memory engrams. Nat Rev Neurosci 7, 575583.
Morris, R.G., Pankratov, Y., Arthur, J.S., eta l. (2012). Grant, S.G., ODell, T.J., Karl, K.A., Stein, P.L.,
MSK1 regulates homeostatic and experience- Soriano, P., and Kandel, E.R. (1992). Impaired
dependent synaptic plasticity. J Neurosci 32, long-term potentiation, spatial learning, and
1303913051. hippocampal development in fyn mutant mice.
Costa-Mattioli, M., Sossin, W.S., Klann, E., and Science 258, 19031910.
Sonenberg, N. (2009). Translational control of Gustafsson, B., Wigstrom, H., Abraham, W.C., and
long-lasting synaptic plasticity and memory. Huang, Y.Y. (1987). Long-term potentiation in
Neuron 61,1026. the hippocampus using depolarizing current
Davis, G.W. (2006). Homeostatic control of neural pulses as the conditioning stimulus to single vol-
activity: From phenomenology to molecular ley synaptic potentials. J Neurosci 7, 774780.
design. Annu Rev Neurosci 29, 307323. Guzman-Karlsson, M.C., Meadows, J.P., Gavin,
Desai, N.S., Cudmore, R.H., Nelson, S.B., and C.F., Hablitz, J.J., and Sweatt, J.D. (2014).
Turrigiano, G.G. (2002). Critical periods for Transcriptional and epigenetic regula-
experience-dependent synaptic scaling in visual tion of Hebbian and non-Hebbian plasticity.
cortex. Nat Neurosci 5, 783789. Neuropharmacology 80,317.
Douglas, R.M., and Goddard, G.V. (1975). Long-term Guzowski, J.F., McNaughton, B.L., Barnes, C.A., and
potentiation of the perforant path-granule cell syn- Worley, P.F. (1999). Environment-specific expres-
apse in the rat hippocampus. Brain Res 86, 205215. sion of the immediate-early gene Arc in hippocam-
Dudek, S.M., and Bear, M.F. (1992). Homosynaptic pal neuronal ensembles. Nat Neurosci 2, 11201124.
long-term depression in area CA1 of hippocam- Harvey, C.D., and Svoboda, K. (2007). Locally
pus and effects of N-methyl-D-aspartate receptor dynamic synaptic learning rules in pyramidal
blockade. Proc Natl Acad Sci USA 89, 43634367. neuron dendrites. Nature 450, 11951200.
184 Part II:Homeostatic Control

Hebb, D.O. (1949). The organization of behaviour Keck, T., Keller, G.B., Jacobsen, R.I., Eysel, U.T.,
(NewYork:Wiley). Bonhoeffer, T., and Hubener, M. (2013). Synaptic
Hengen, K.B., Lambo, M.E., Van Hooser, S.D., Katz, scaling and homeostatic plasticity in the mouse
D.B., and Turrigiano, G.G. (2013). Firing rate visual cortex in vivo. Neuron 80, 327334.
homeostasis in visual cortex of freely behaving Kelleher, R.J., 3rd, Govindarajan, A., and Tonegawa, S.
rodents. Neuron 80, 335342. (2004). Translational regulatory mechanisms in
Henneberger, C., Papouin, T., Oliet, S.H., and persistent forms of synaptic plasticity. Neuron
Rusakov, D.A. (2010). Long-term potentiation 44,5973.
depends on release of D-serine from astrocytes. Kim, R., Okuno, H., and Bito, H. (2012). Deciphering
Nature 463, 232236. the molecular rules governing synaptic targeting
Hensch, T.K. (2004). Critical period regulation. of the memory-related protein Arc. Commun
Annu Rev Neurosci 27, 549579. Integr Biol 5, 496498.
Hu, J.H., Park, J.M., Park, S., Xiao, B., Dehoff, M.H., Kitamura, T., Saitoh, Y., Takashima, N., Murayama, A.,
Kim, S., Hayashi, T., Schwarz, M.K., Huganir, R.L., Niibori, Y., Ageta, H., Sekiguchi, M., Sugiyama, H.,
Seeburg, P.H., etal. (2010). Homeostatic scaling and Inokuchi, K. (2009). Adult neurogenesis
requires group ImGluR activation mediated by modulates the hippocampus-dependent period
Homer1a. Neuron 68, 11281142. of associative fear memory. Cell 139, 814827.
Hubel, D.H., Wiesel, T.N., and LeVay, S. (1976). Kullmann, D.M., and Lamsa, K.P. (2007). Long-term
Functional architecture of area 17 in normal and synaptic plasticity in hippocampal interneurons.
monocularly deprived macaque monkeys. Cold Nat Rev. Neurosci. 8, 687699.
Spring Harbor Symposia on Quantitative Biol., Lamsa, K.P., Heeroma, J.H., Somogyi, P., Rusakov,
40, 581589. D.A., and Kullmann, D.M. (2007). Anti-
Huganir, R.L., and Nicoll, R.A. (2014). AMPARs and Hebbian long-term potentiation in the hip-
synaptic plasticity:The last 25years. Neuron 80, pocampal feedback inhibitory circuit. Science,
704717. 315, 12621266.
Hulme, S.R., Jones, O.D., and Abraham, W.C. (2013). Lee, H.K., Takamiya, K., Han, J.S., Man, H., Kim, C.H.,
Emerging roles of metaplasticity in behaviour Rumbaugh, G., Yu, S., Ding, L., He, C., Petralia,
and disease. Trends Neurosci 36, 353362. R.S., etal. (2003). Phosphorylation of the AMPA
Ibata, K., Sun, Q., and Turrigiano, G.G. (2008). Rapid receptor GluR1 subunit is required for synaptic
synaptic scaling induced by changes in postsyn- plasticity and retention of spatial memory. Cell
aptic firing. Neuron 57, 819826. 112, 631643.
Imayoshi, I., Sakamoto, M., Ohtsuka, T., Takao, K., Lee, K.H., Chatila, T.A., Ram, R.A., and Thompson, R.F.
Miyakawa, T., Yamaguchi, M., Mori, K., Ikeda, T., (2009). Impaired memory of eyeblink condition-
Itohara, S., and Kageyama, R. (2008). Roles of ing in CaMKIV KO mice. Behav Neurosci 123,
continuous neurogenesis in the structural and 438442.
functional integrity of the adult forebrain. Nat LeVay, S., Wiesel, T.N., and Hubel, D.H. (1980). The
Neurosci 11, 11531161. development of ocular dominance columns in
Ito, M. (1989). Long-term depression. Annu Rev normal and visually deprived monkeys. J Comp
Neurosci 12, 85102. Neurol 191,151.
Ito, M. (2000). Mechanisms of motor learning in the Link, W., Konietzko, U., Kauselmann, G., Krug,
cerebellum. Brain Res 886, 237245. M., Schwanke, B., Frey, U., and Kuhl, D. (1995).
Kandel, E.R. (2001). The molecular biology of mem- Somatodendritic expression of an immediate
ory storage:Adialogue between genes and syn- early gene is regulated by synaptic activity. Proc
apses. Science 294, 10301038. Natl Acad Sci USA 92, 57345738.
Kano, M., Hashimoto, K., Kurihara, H., Watanabe, M., Liu, C.C., Kanekiyo, T., Xu, H., and Bu, G. (2013).
Inoue, Y., Aiba, A., and Tonegawa, S. (1997). Apolipoprotein E and Alzheimer disease: Risk,
Persistent multiple climbing fiber innerva- mechanisms and therapy. Nat Rev Neurol 9,
tion of cerebellar Purkinje cells in mice lacking 106118.
mGluR1. Neuron 18,7179. Luo, L. (2002). Actin cytoskeleton regulation in neu-
Kasai, H., Matsuzaki, M., Noguchi, J., Yasumatsu, ronal morphogenesis and structural plasticity.
N., and Nakahara, H. (2003). Structure- Annu Rev Cell Dev Biol 18, 601635.
stability-function relationships of dendritic Luscher, C., and Huber, K.M. (2010). Group 1
spines. Trends Neurosci 26, 360368. mGluR-dependent synaptic long-term depres-
Kauer, J.A., Malenka, R.C., and Nicoll, R.A. (1988). sion:Mechanisms and implications for circuitry
A persistent postsynaptic modification medi- and disease. Neuron 65, 445459.
ates long-term potentiation in the hippocampus. Luscher, C., and Malenka, R.C. (2011). Drug-evoked
Neuron 1, 911917. synaptic plasticity in addiction:From molecular
Neuroplasticity 185

changes to circuit remodeling. Neuron 69, depression in area CA1 of the hippocampus.
650663. Neuron 9, 967975.
Lyford, G.L., Yamagata, K., Kaufmann, W.E., Nabavi, S., Fox, R., Proulx, C.D., Lin, J.Y., Tsien, R.Y.,
Barnes, C.A., Sanders, L.K., Copeland, N.G., and Malinow, R. (2014). Engineering a memory
Gilbert, D.J., Jenkins, N.A., Lanahan, A.A., with LTD and LTP. Nature 511, 348352.
and Worley, P.F. (1995). Arc, a growth factor Nestler, E.J., Hope, B.T., and Widnell, K.L. (1993).
and activity-regulated gene, encodes a novel Drug addiction:Amodel for the molecular basis
cytoskeleton-associated protein that is enriched of neural plasticity. Neuron 11, 9951006.
in neuronal dendrites. Neuron 14, 433445. Neves, G., Cooke, S.F., and Bliss, T.V. (2008).
Magee, J.C., and Johnston, D. (1997). A synaptically Synaptic plasticity, memory and the hippocam-
controlled, associative signal for Hebbian plastic- pus: A neural network approach to causality. Nat
ity in hippocampal neurons. Science 275, 209213. Rev Neurosci 9,6575.
Malinow, R., Madison, D.V., and Tsien, R.W. (1988). Nguyen, P.V. (2001). Heterosynaptic strengthening of
Persistent protein kinase activity underlying hippocampal LTP. Trends Neurosci 24, 502503.
long-term potentiation. Nature 335, 820824. OBrien, R.J., Xu, D., Petralia, R.S., Steward, O.,
Malinow, R., and Malenka, R.C. (2002). AMPA Huganir, R.L., and Worley, P. (1999). Synaptic
receptor trafficking and synaptic plasticity. clustering of AMPA receptors by the extracellu-
Annu Rev Neurosci 25, 103126. lar immediate-early gene product Narp. Neuron
Man, H.Y., Lin, J.W., Ju, W.H., Ahmadian, G., Liu, L., 23, 309323.
Becker, L.E., Sheng, M., and Wang, Y.T. (2000). Oddo, S., Caccamo, A., Shepherd, J.D., Murphy, M.P.,
Regulation of AMPA receptor-mediated synap- Golde, T.E., Kayed, R., Metherate, R., Mattson,
tic transmission by clathrin-dependent receptor M.P., Akbari, Y., and LaFerla, F.M. (2003).
internalization. Neuron 25, 649662. Triple-transgenic model of Alzheimers disease
Markram, H., Lubke, J., Frotscher, M., and Sakmann, B. with plaques and tangles: Intracellular Abeta
(1997). Regulation of synaptic efficacy by coin- and synaptic dysfunction. Neuron 39, 409421.
cidence of postsynaptic APs and EPSPs. Science Okuno, H. (2011). Regulation and function of
275, 213215. immediate-early genes in the brain:Beyond neu-
Martin, S.J., Grimwood, P.D., and Morris, R.G. ronal activity markers. Neurosci Res 69, 175186.
(2000). Synaptic plasticity and memory:An eval- Okuno, H., Akashi, K., Ishii, Y., Yagishita-Kyo, N.,
uation of the hypothesis. Annu Rev Neurosci 23, Suzuki, K., Nonaka, M., Kawashima, T., Fujii,
649711. H., Takemoto-Kimura, S., Abe, M., etal. (2012).
Matsuzaki, M., Honkura, N., Ellis-Davies, G.C., and Inverse synaptic tagging of inactive synapses
Kasai, H. (2004). Structural basis of long-term via dynamic interaction of Arc/Arg3.1 with
potentiation in single dendritic spines. Nature CaMKIIbeta. Cell 149, 886898.
429, 761766. Otmakhov, N., Griffith, L.C., and Lisman, J.E.
McCurry, C.L., Shepherd, J.D., Tropea, D., Wang, (1997). Postsynaptic inhibitors of calcium/
K.H., Bear, M.F., and Sur, M. (2010). Loss of calmodulin-dependent protein kinase type
Arc renders the visual cortex impervious to the II block induction but not maintenance of
effects of sensory experience or deprivation. Nat pairing-induced long-term potentiation. J
Neurosci 13, 450457. Neurosci 17, 53575365.
Merzenich, M.M., Nelson, R.J., Stryker, M.P., Otmakhov, N., Tao-Cheng, J.H., Carpenter, S.,
Cynader, M.S., Schoppmann, A., and Zook, J.M. Asrican, B., Dosemeci, A., Reese, T.S., and
(1984). Somatosensory cortical map changes Lisman, J. (2004). Persistent accumulation of
following digit amputation in adult monkeys. J calcium/calmodulin-dependent protein kinase
Comp Neurol 224, 591605. II in dendritic spines after induction of NMDA
Morris, R.G., Moser, E.I., Riedel, G., Martin, S.J., receptor-dependent chemical long-term poten-
Sandin, J., Day, M., and OCarroll, C. (2003). tiation. J Neurosci 24, 93249331.
Elements of a neurobiological theory of the hip- Pascual, O., Casper, K.B., Kubera, C., Zhang, J.,
pocampus: The role of activity-dependent synap- Revilla-Sanchez, R., Sul, J.Y., Takano, H., Moss,
tic plasticity in memory. Philos Trans R Soc Lond S.J., McCarthy, K., and Haydon, P.G. (2005).
B Biol Sci 358, 773786. Astrocytic purinergic signaling coordinates syn-
Moser, E.I., Krobert, K.A., Moser, M.B., and Morris, aptic networks. Science 310, 113116.
R.G. (1998). Impaired spatial learning after satu- Perea, G., Navarrete, M., and Araque, A. (2009).
ration of long-term potentiation. Science 281, Tripartite synapses:Astrocytes process and control
20382042. synaptic information. Trends Neurosci 32, 421431.
Mulkey, R.M., and Malenka, R.C. (1992). Mechanisms Plath, N., Ohana, O., Dammermann, B., Errington,
underlying induction of homosynaptic long-term M.L., Schmitz, D., Gross, C., Mao, X., Engelsberg,
186 Part II:Homeostatic Control

A., Mahlke, C., Welzl, H., et al. (2006). Arc/ learning shifts transsynaptically from cerebellar
Arg3.1 is essential for the consolidation of synap- cortex to nuclei for consolidation. Neuroscience
tic plasticity and memories. Neuron 52, 437444. 139, 767777.
Qiu, Z., Sylwestrak, E.L., Lieberman, D.N., Zhang, Silva, A.J., Paylor, R., Wehner, J.M., and Tonegawa, S.
Y., Liu, X.Y., and Ghosh, A. (2012). The Rett syn- (1992a). Impaired spatial learning in
drome protein MeCP2 regulates synaptic scal- alpha-calcium-calmodulin kinase II mutant
ing. J Neurosci 32, 989994. mice. Science 257, 206211.
Redondo, R.L., and Morris, R.G. (2011). Making Silva, A.J., Stevens, C.F., Tonegawa, S., and Wang, Y.
memories last: The synaptic tagging and capture (1992b). Deficient hippocampal long-term poten-
hypothesis. Nat Rev Neurosci 12,1730. tiation in alpha-calcium-calmodulin kinase II
Rial Verde, E.M., Lee-Osbourne, J., Worley, P.F., mutant mice. Science 257, 201206.
Malinow, R., and Cline, H.T. (2006). Increased Soden, M.E., and Chen, L. (2010). Fragile X protein
expression of the immediate-early gene arc/ FMRP is required for homeostatic plasticity and
arg3.1 reduces AMPA receptor-mediated synap- regulation of synaptic strength by retinoic acid. J
tic transmission. Neuron 52, 461474. Neurosci 30, 1691016921.
Rioult-Pedotti, M.S., Friedman, D., and Donoghue, Stellwagen, D., and Malenka, R.C. (2006). Synaptic
J.P. (2000). Learning-induced LTP in neocortex. scaling mediated by glial TNF-alpha. Nature
Science 290, 533536. 440, 10541059.
Royer, S., and Pare, D. (2002). Bidirectional synap- Sutton, M.A., Ito, H.T., Cressy, P., Kempf, C., Woo,
tic plasticity in intercalated amygdala neurons J.C., and Schuman, E.M. (2006). Miniature neu-
and the extinction of conditioned fear responses. rotransmission stabilizes synaptic function via
Neuroscience 115, 455462. tonic suppression of local dendritic protein syn-
Sahay, A., Scobie, K.N., Hill, A.S., OCarroll, C.M., thesis. Cell 125, 785799.
Kheirbek, M.A., Burghardt, N.S., Fenton, A.A., Thiagarajan, T.C., Lindskog, M., and Tsien, R.W.
Dranovsky, A., and Hen, R. (2011). Increasing (2005). Adaptation to synaptic inactivity in hip-
adult hippocampal neurogenesis is sufficient to pocampal neurons. Neuron 47, 725737.
improve pattern separation. Nature 472, 466470. Turrigiano, G.G. (2008). The self-tuning neu-
Sajikumar, S., Navakkode, S., and Frey, J.U. ron:Synaptic scaling of excitatory synapses. Cell
(2007). Identification of compartment- and 135, 422435.
process-specific molecules required for synap- Turrigiano, G.G., Leslie, K.R., Desai, N.S.,
tic tagging during long-term potentiation and Rutherford, L.C., and Nelson, S.B. (1998).
long-term depression in hippocampal CA1. J Activity-dependent scaling of quantal amplitude
Neurosci 27, 50685080. in neocortical neurons. Nature 391, 892896.
Savin, C., Triesch, J., and Meyer-Hermann, M. Wang, Y.T., and Linden, D.J. (2000). Expression of
(2009). Epileptogenesis due to glia-mediated syn- cerebellar long-term depression requires post-
aptic scaling. J R Soc Interface 6, 655668. synaptic clathrin-mediated endocytosis. Neuron
Shah, M.M., Hammond, R.S., and Hoffman, D.A. 25, 635647.
(2010). Dendritic ion channel trafficking and Waung, M.W., Pfeiffer, B.E., Nosyreva, E.D.,
plasticity. Trends Neurosci 33, 307316. Ronesi, J.A., and Huber, K.M. (2008). Rapid
Shatz, C.J., and Stryker, M.P. (1978). Ocular domi- translation of Arc/Arg3.1 selectively mediates
nance in layer IV of the cats visual cortex and mGluR-dependent LTD through persistent
the effects of monocular deprivation. J Physiol increases in AMPAR endocytosis rate. Neuron
281, 267283. 59,8497.
Shepherd, J.D., Rumbaugh, G., Wu, J., Chowdhury, Whitlock, J.R., Heynen, A.J., Shuler, M.G., and
S., Plath, N., Kuhl, D., Huganir, R.L., and Worley, Bear, M.F. (2006). Learning induces long-term
P.F. (2006). Arc/Arg3.1 mediates homeostatic potentiation in the hippocampus. Science 313,
synaptic scaling of AMPA receptors. Neuron 52, 10931097.
475484. Xia, J., Chung, H.J., Wihler, C., Huganir, R.L.,
Shibuki, K., Gomi, H., Chen, L., Bao, S., Kim, J.J., and Linden, D.J. (2000). Cerebellar long-term
Wakatsuki, H., Fujisaki, T., Fujimoto, K., Katoh, depression requires PKC-regulated interactions
A., Ikeda, T., et al. (1996). Deficient cerebellar between GluR2/3 and PDZ domain-containing
long-term depression, impaired eyeblink con- proteins. Neuron 28, 499510.
ditioning, and normal motor coordination in Zhang, L.I., Tao, H.W., Holt, C.E., Harris, W.A., and
GFAP mutant mice. Neuron 16, 587599. Poo, M. (1998). A critical window for coopera-
Shutoh, F., Ohki, M., Kitazawa, H., Itohara, S., tion and competition among developing retino-
and Nagao, S. (2006). Memory trace of motor tectal synapses. Nature 395,3744.
12
Epigenetics
K AT J A K O B O W A N D I N G M A R B L M C K E

INTRODUCTION as MBD1, MBD2, MBD3, MBD4, MeCP2, and


Epigenetic mechanisms are self-perpetuating, KAISO (Buck-Koehntop and Defossez, 2013;
functionally relevant modifications to the Fournier etal., 2012; Lunyak etal.,2002).
genome not involving changes in nucleotide Present in all higher eukaryotes including
sequence. They include covalent modifications humans, DNA methylation is mainly confined to
of histone proteins and DNA, chromatin remod- CpG dinucleotides (Dulac, 2010). However, meth-
eling, as well as the action of noncoding RNAs. ylation of non-CpG sites has also been reported
The number and variability of epigenetic marks, for embryonic stem cells, induced pluripotent
their flexibility over time, their interdependence, stem cells, oocytes, and the brain (Barres etal.,
and potential synergistic effects may provide the 2009; Clark et al., 1995; Grandjean et al., 2007;
molecular basis for any phenotypic variation in Ichiyanagi etal., 2013; Lister etal., 2013; Varley
physiological and pathological conditions. This etal., 2013; Ziller etal., 2011). While CpG meth-
is particularly interesting with respect to post- ylation is mainly thought to lock the genome and
mitotic neurons in the brain. Experimental data provide a basal repressive mark, the biological
showed evidence for epigenetic mechanisms as impact of non-CpG methylation remains poorly
fundamental regulatory processes in neurogen- understood. Given that this mark is highly pre-
esis and central nervous system (CNS) develop- sent in the adult mouse and human brain but
ment, cellular plasticity, and higher order brain rare or absent in other differentiated cell types,
function. There is further compelling evidence a unique role in mammalian brain development
that dysfunctional epigenetic processes are and function can be assumed (Lister etal.,2013).
involved in the pathobiology of neurologic dis- DNA methylation is usually high within
eases and may serve as the molecular arbiter for sequences of low CpG content, for example at
integrating the effects of inherited and acquired noncoding regions (e.g., centromeric hetero-
etiological factors and thus for modulating clini- chromatin) and interspersed highly repetitive
cal manifestations of a specific disease. elements (endogenous retroviruses and transpo-
sons). Therefore it has been suggested to prevent
BUILDING BLOCKS genomic instability phenomena, making genome
OFTHE EPIGENOME stabilization a primary function of DNA meth-
ylation (Bestor, 2000). In contrast, regions of high
DNA Methylation CpG density, referred to as CpG islands (CGIs),
DNA methylation is a covalent chromatin modifi- are frequently found in the 5 end of genes and
cation, characterized by the biochemical addition have been identified as sites of transcription ini-
of a methyl group (CH3) to cytosine nucleo- tiation (Eckhardt etal., 2006; Weber etal., 2007).
tides. S-adenosyl methionine (SAM) serves as CGIs are very frequently located within pro-
the methyl donor that is converted to S-adenosyl moters, often in the direct vicinity of transcrip-
homocysteine following methyl group trans- tion start sites. Some CGIs are not associated
fer (Figure 12.1). DNA methylation patterns are with known genes, but we think there are still
directed and preserved by the action of the DNA unknown genes hidden in the genome. In favor
methyltransferase (DNMT) family, whereas the of this hypothesis respective CGIs show features
effects of DNA methylation are mediated by of standard gene promoters and active genes
recruitment of the reader methyl-CpG-binding including the binding of specific proteins (PolII),
domain (MBD) family, containing proteins such chromatin modifications that support active
188 Part II:Homeostatic Control

Other studies suggested a role of intragenic DNA


methylation in controlling alternative promoter
usage, particularly in the brain (Maunakea etal.,
2010). It is generally anticipated that downstream
effects of DNA methylation may result from
(a)interference with transcription factor binding,
(b) recruitment of methyl-binding proteins and
their associated regulatory complexes, and/or
(c)induced chromatin remodeling. Interestingly,
mammalian transcription factor binding sites
are more CG rich than the bulk genome, and
many contain CpG within their recognition
FIGURE 12.1: DNA methylation and demeth- sequence (Deaton and Bird, 2011), suggesting a
ylation dynamics in the mammalian brain. strong interdependence.
Potential mechanisms of DNA demethylation. Passive DNA methylation is present in all higher
DNA demethylation is thought to occur by a reduc- eukaryotes, including humans (Dulac,
tion in activity or absence of DNA methyltransferases 2010) and, besides general stabilization of the
(DNMT) during DNA replication, whereas active genome, is associated with parent-of-origin
DNA demethylation involves DNA repair mecha- imprinting (Edwards and Ferguson-Smith,
nisms: 5-methylcytosine (5-mC) is hydroxylated by 2007), X-chromosome inactivation in females
ten-eleven translocation (TET) to form the key inter- (Minkovsky et al., 2012), during the process of
mediate 5-hydroxymethylcytosine (5-hmC). 5-mC or aging (Madrigano et al., 2012; Numata et al.,
5-hmC are deaminated by activation-induced cyti- 2012), in lineage commitment (e.g., during neu-
dine deaminase (AID) to form thymine (not shown) rogenesis; Ma etal., 2010), and in neural plastic-
or 5-hydroxymethyluracil (5-hmU), which are then ity (Feng etal., 2010; Guo etal., 2011a; Levenson
removed by thymine DNA glycosylases (TDG) and etal., 2006; Miller and Sweatt, 2007; Nelson etal.,
exchanged by unmethylated C through the BER path- 2008). Despite previous hypotheses, DNA meth-
way. Alternatively, 5-hmC may be further oxidized by ylation has proven to be highly dynamic not only
TET to form other intermediates, which are converted during development and cellular differentiation
to cytosine or 5-hmC may be lost during replication. (Jaenisch and Bird, 2003; Reik etal., 2001; Reik
Double arrowheads indicate chemical chain reactions, and Walter, 2001)but also in adult and postmi-
where intermediates were not shown for simplicity. totic cells (Guo etal., 2011a; Reik, 2007; Wu and
Other abbreviations: SAM = S-adenosyl methionine; Zhang, 2010). Genomic or locus-dependent DNA
SAH =S-adenosyl homocysteine; Gadd45 = growth methylation loss were observed in both physi-
arrest and DNA-damage-inducible. ologic and pathologic conditions, but no DNA
demethylase has been identified so far (Ooi and
Bestor, 2008). DNA methylation may be pas-
gene expression and RNA that is obviously tran- sively lost or diluted during the cell cycle, when
scribed from these gene loci. All of these findings DNA methylation enzymes and/or their com-
point to orphan CGIs as sites of transcription plexes are denied access to the newly replicated
initiation (Illingworth etal.,2010). DNA (Figure 12.1, left panel). In addition, active
The great majority of CGIs are unmethyl- DNA demethylation may be facilitated in non-
ated at all stages of development in all normal, dividing cells through the selective recruitment
nondiseased tissue types (Bird, 2002; Edwards of cytidine deaminases and DNA glycosylases
etal., 2010), thereby retaining an open chromatin (Figure 12.1, right panel; Gong and Zhu,
structure for potential accession of transcription 2011). A common intermediate in the pro-
factors and dynamic regulation of gene expres- cess of DNA demethylation seems to be
sion (Bergman and Cedar, 2013). However, when 5-hydroxymethyl-Cytosine (5-hmC). The func-
present, promoter methylation is generally cor- tion of 5-hmC is still matter of debate, but there
related with gene repression. In contrast to pro- is some evidence for 5-hmC-mediated epige-
moter regions, gene body methylation shows less netic dynamics during postnatal development
stringent associations with gene silencing. There and aging in the rodent as well as the human
are reports that intragenic methylation is indica- brain (Hahn et al., 2013; Szulwach et al., 2011;
tive for active transcription, including the regu- Wang et al., 2012). Oxidative deamination by
lation of alternative splicing (Sati et al., 2012). the activation-induced cytidine deaminase/
Epigenetics 189

apolipoprotein B messanger RNA (mRNA)- phosphorylation, methylation, ubiquitination,


editing catalytic component family of deaminases and ADP-ribosylation (Khorasanizadeh, 2004).
converts 5-hmC to 5-hmU or other interme- Intriguingly, multiple posttranslational histone
diates, which may all be further processed by modifications may function combinatorially and/
base excision repair enzymes (Guo etal., 2011b). or sequentially to regulate downstream function
Another enzyme considered to facilitate active (Fischle, 2008; Wang et al., 2008). Through its
DNA demethylation is the growth arrest and complexity, the so-called histone code ensures
DNA damage 45 (Gadd45) family of genes (Engel nuclear processes (transcription, replication,
etal., 2009). Gadd45 has been suggested to act as DNA damage response) to be directed to the
an adapter protein for nucleotide excision repair required region of the genome at the appropriate
and/or base excision repair enzymes promot- time, mediating unique cellular responses and
ing removal of 5-methylcytosine from DNA at biological outcomes (Figure12.2).
selected gene-specific loci (Schafer,2013). In histone acetyltransferase, a negatively
charged acetyl group is added to lysine resi-
Posttranslational Histone dues on histone proteins. An acetylated lysine
Modification has no longer a basic side chain and interferes
DNA is wrapped around histone octamers con- with the usual electrostatic affinity between
taining two H2A, H2B, H3, and H4 proteins, histone proteins and the DNA backbone. It is a
each forming the basic unit of chromatinthe hypothesis structure more permissive to gene
nucleosome. Each histone protein is composed expression (Graff and Tsai, 2013). Histone meth-
of a central globular domain and an N-terminal ylation can target lysine or arginine residues
tail that contains multiple sites for posttrans- (Di Lorenzo and Bedford, 2011; Lachner and
lational modification, including acetylation, Jenuwein, 2002). Of note, different methylation

NH2
NUCLEOSOME

C K119

Ub NH2
H2A H2B
Ub
Ub

H4 H3

P A
NH2 K36
K20 S10
K16 K27 K9
M

M M M M M M
M M
M

A
M

M M NH2
K4
M

FIGURE 12.2: Nucleosome structure and histonecode. Simplified scheme of nucleosome structure and
histone code. All histones are subject to posttranscriptional modifications, which mainly occur in N-terminal
histone tails. Posttranscriptional histone modifications include acetylation (Ac), methylation (M), phosphoryl-
ation (P), and ubiquitination (Ub), among others. Each modification has specific functions in gene regulation
(green=activation, red=inhibition), whereby interdependence of signals and crosstalk between different signals
allows very fine-tuning of gene expression. The sum of all activating and inhibiting signals define the activity state
of a specific gene. Histone acetylation has long been associated with increased gene expression. More recently,
histone lysine methylation has been linked to both gene activation and gene repression, depending on the site
and extent of methylation. The number associated with each amino acid represents its position in the sequence.
K=lysine; S=serine; NH2=N-terminal histone tail; C=C-terminaltail.
190 Part II:Homeostatic Control

marks are established by specific enzymes with In addition to the well-known acetylation or
relatively little overlap in substrate and function methylation, histones can also be ubiquitinated
(Kouzarides, 2007; Wu et al., 2010). They may (Zhang, 2003). This modification was long con-
further have opposing effects on gene expres- sidered to be associated only with the protein
sion, dependent on the targeted amino acid. degradation system, whereas its contribution to
Methylation of H3K9, H3K27, H4K20 has been transcriptional regulation remained uncertain.
associated with condensed chromatin states Proteasome-dependent degradation of polyubiq-
and gene repression (Martin and Zhang, 2005), uitinated transcription factors or linker protein
although different effects could be observed H1 are important regulatory mechanisms, but
with mono-, di-, or trimethylation of the same now proteasome-independent mechanisms have
amino acid (Barski et al., 2007). H3K9 meth- also come into focus. The carefully orchestrated
ylation has been particularly implicated in the addition and removal of single ubiquitin moieties
induction and propagation of heterochroma- to histones has been associated with gene regu-
tin formation, whereas H4K20 methylation is a lation (Davie and Murphy, 1990). There is fur-
key regulator of biological processes that ensure ther evidence for an interdependence of histone
genome integrity, such as DNA damage repair, ubiquitination and other epigenetic mechanisms.
DNA replication, and chromatin compaction Intriguingly, H2B monoubiquitination appears
during cell cycles (Jrgensen etal., 2013; Tardat to be implicated in the recruitment of some
et al., 2007). H3K27 methylation contributes methyltransferases mediating di- and trimeth-
to the inactivation of gene expression. Given ylation of H3K4 and H3K79 commonly associ-
the critical role of H3K27 methylation in the ated with transcriptionally active chromatin (Lee
balance of gene activity, it is not surprising to etal., 2007). Moreover, ubiquitination of histones
find anomalies of this system in different types may regulate histone acetylation and facilitate
of cancer (Martinez-Garcia and Licht, 2010). histone eviction, as the sequential acetylation
Both H3K9- and H3K27-trimethylation have and ubiquitination of H2AX has been shown to
been further described as playing a crucial role promote histone dynamics (Ikura et al., 2007).
in regulating DNA methylation, thereby link- An interdependence of H2A ubiquitination, his-
ing different epigenetic repression systems tone, and DNA methylation has been established
(Lehnertz et al., 2003; Vire et al., 2006). This and linked to X-chromosome inactivation as well
interdependence seems to hold true for most of as polycomb silencing (de Napoles et al., 2004;
the genome, except CpG islands in postmitotic Sarcinella et al., 2007; Wang et al., 2004; Wu
neurons, where more recent evidence showed et al., 2008). These findings promote an impor-
that H3K27-trimethylation and DNA methyla- tant role for histone ubiquitination in transcrip-
tion are mutually exclusive and work antagonis- tional regulation. However, most findings were
tically to silence genes (Brinkman etal.,2012). obtained in yeast and flies and await experimen-
Other methylation marks seem to be restricted tal confirmation in higher organisms. An explicit
to active gene promoters (e.g., H3K4 methylation role in the homeostasis of brain function has not
associated with transcription initiation), while been shownyet.
H3K36 methylation may support transcription
elongation (Rando, 2012). Monomethylation of NoncodingRNAs
H3K4 distinguishes active enhancers, whereas Noncoding RNAs are small, functional RNA
trimethylation of H3K4 is highly enriched at molecules that are not translated into protein but
promoters and around transcriptional start sites are implicated in transcriptional and posttran-
(Heintzman etal., 2007). Binding of H3K4-specific scriptional regulation of gene expression. They
methyltransferases protects promoters of devel- include microRNAs (miRNAs; He and Hannon,
opmental genes from DNA methylation (Smith 2004), long noncoding RNAs (Mercer et al.,
and Meissner, 2013), further underlining its role 2009), and PIWI-interacting RNAs (P-element
in gene activation. Intriguingly, developmen- induced wimpy testis in Drosophilas; Luteijn and
tally critical genes frequently contain a bivalent Ketting, 2013), all characterized by individual
domain within their promoters, where function- biogenesis, mode of target recognition, regula-
ally opposing H3K4- and H3K27-trimethylation tory properties, and transgenerational epigenetic
marks are present at the same time. This pattern states. Here we focus on miRNAs, a group of
allows lineage-specific genes to be either silenced highly conserved small regulatory RNAs playing
or activated as differentiation proceeds (Bernstein essential roles in CNS development and neuronal
etal.,2006). function, including proliferation of neural stem
Epigenetics 191

and progenitor cells, neuronal differentiation of mRNA translation. There is also evidence for
and maturation (De Pietri Tonelli et al., 2008; transcriptional and translational activation by
Sun etal., 2013a), neurite outgrowth, and synap- miRNAs, introducing a new level of complexity
togenesis (Jovicic etal., 2013; Olde Loohuis etal., (Figure 12.3; Vasudevan etal.,2007).
2012; Yoo etal., 2011). Despite their critical role Studies on region-specific genetic ablation of
in neurodevelopment, miRNAs are by definition Dicer and the subsequent blocking of miR bio-
not epigenetic players per se but can regulate genesis at different developing stages are com-
other epigenetic layers (Zhou et al., 2010) or be patible with global effects of miRNAs on CNS
epigenetically regulated themselves (Lopez-Serra development (Davis et al., 2008; Kawase-Koga
and Esteller,2012). et al., 2009). Cortex-specific Dicer conditional
Biogenesis of miRNAs initially starts with knockout mice displayed a reduced neural pro-
transcription from noncoding regions within genitor pool and abnormal neuronal differen-
the genome followed by sequential cleavage tiation (Kawase-Koga et al., 2009). Similarly,
of the primary stem loop structure transcripts knockout of specific miR genes (e.g. miR-9,
(pri-microRNA) to form mature miRNAs (Kim miR-124) proved their implication in embry-
et al., 2009). Processing of miRNA precursors onic and adult neurogenesis (Sun et al., 2013a).
is mediated sequentially by RNase-III enzymes Despite the presented global relevance of miR
Drosha and Dicer. Finally, mature miRNAs signaling in brain development, activity of miR-
guide the RNA-induced silencing complex to 3 NAs may also be locally restricted to specific cell
untranslated regions of complementary target types or even subcellular compartments, where
mRNAs and repress gene expression by either they, for example, modulate synaptic activity
destabilization of mRNA transcripts (i.e., decap- and neuronal connectivity (Hengst etal., 2006;
ping, deadenylation, degradation) or repression Schratt etal.,2006).

CYTOPLASM

Dicer Exportin
double stranded
mature microRNA

microRNA Drosha
RISC precursor
primary microRNA
ORF AAAAAAA transcript

NUCLEUS

epigenetic chromatin
mRNA degradation translational inhibition modifications
(perfect match) (imperfect match)

FIGURE 12.3: MicroRNA biogenesis and function. Simplified schematic on microRNA biogenesis and
RNA-induced gene silencing. Transcription of primary microRNA from miRNA genes is followed by cleavage
to precursor microRNA by the Drosha nuclear RNase III. The microRNA precursor is then exported to the cyto-
plasm by exportin via nuclear pore. In the cytoplasm, the precursor microRNA is further processed by RNase
activity of Dicer to the mature microRNA duplex. The duplex loads onto the RNA-induced silencing complex
(RISC) complex, but only one of the mature microRNA strands (red strand) mediates RNA silencing by degrading
the target mRNA or interfering with translation, while the second microRNA strand (grey strand) is degraded
itself by the RNase activity of RISC. The outcome of RISC formation varies with the degree of complementarity
of the seed sequence of microRNA with the 3 untranslated region of the target mRNA. MicroRNAs can regulate
different epigenetic layers including DNA methylation, histone modification, and chromatin remodeling or be
epigenetically regulated themselves (miR expression, miR function).
Modified from Cuellar and McManus,2005
192 Part II:Homeostatic Control

Chromatin Remodelers Hox gene expression is spatially controlled


Alterations in gene expression are closely related by Polycomb group (PcG) and trithorax group
to changes in higher-order and/or locus-specific (trxG) proteins, which comprise protein com-
chromatin structure, and chromatin-remodeling plexes with epigenetic function implicated in
enzymes play key roles in differentiation and devel- lineage commitment and cellular memory. The
opment. ATP-dependent chromatin-remodeling specific relevance of the PcG system in neuro-
enzymes can be divided into three subfamilies genesis and CNS development is exemplified by
according to structure and function:the SWItch/ neuronal defects in various PcG mouse mutants
Sucrose Nonfermentable (SWI/SNF), chromodo- (Fasano etal., 2009). In mice, genetic ablation of
main and helicase-like domain (CHD), and imi- components of the polycomb repressor complex
tation SWI families (Flaus and Owen-Hughes, 1 (e.g., Bmi1 polycomb ring finger oncogene,
2011). Each subfamily harbors a unique domain Bmi1; Ring finger protein 2, Rnf2) interferes
(Bromo, Chromo, SANT) that helps to recruit with cerebral neural stem cell renewal, result-
remodeling complexes to regions of specifically ing in progressive postnatal growth retardation
modified chromatin. Importantly, remodeling and neurological abnormalities manifested, for
enzymes use energy provided by ATP hydrolysis example, by an ataxic gait and sporadic seizures
to locally disrupt histone-DNA associations and (Molofsky etal., 2003; van der Lugt etal.,1994).
shift nucleosomes to alternate positions. They are In the course of neocortical development,
further organized into protein complexes and neural progenitors produce neurons first and
may be regulated by subunit composition as sug- astrocytes later. Evidence that the PcG system
gested by the isolation of SWI/SNF complexes also regulates the temporal regulation of neural
containing a neuron-specific subunit from the progenitor cell fate comes from studies show-
brain of mice (Kim etal., 2001). There is evidence ing that ablation of several Polycomb-repressive
that chromatin remodeling complexes are key complex 2 components (e.g., enhancer of zeste
regulators of neural development (Potts et al., homolog 2, Ezh2; embryonic ectoderm devel-
2011; Ronan et al., 2013; Seo et al., 2005), with opment, Eed) prolonged the neurogenic and
some of their components being abundantly, delayed the gliogenic phase in vivo (Hirabayashi
sometimes exclusively, expressed within the etal., 2009; Pereira etal., 2010), thereby severely
mammalian brain, spinal cord, or retina. affecting timing of cortical development. Histone
modification dynamics, also independent from
EPIGENETICS the PcG/trxG system, are relevant for proper
INNEUROGENESIS AND regulation of neuronal differentiation, matura-
BR AIN DEVELOPMENT tion, and migration, as well as maintenance of
Neurogenesis is defined as the generation of new neuronal function as deduced from the specific
functional neurons and can be divided into two spatiotemporal expression of (e.g., SET domain,
phases: embryonic/developmental neurogenesis, bifurcated 1, Setdb1, a H3K9 specific methyl-
which basically forms the CNS, and adult neuro- transferase). It is highly expressed at early stages
genesis, which continues at low levels in postna- of mouse brain development and is particularly
tal and adult brains (Jobe etal., 2012). Epigenetic located in the ventricular zone, where neural
mechanisms carry out diverse roles in regulating progenitor cells reside. Setdb1 expression could
specific aspects of embryonic and adult neuro- be associated with cell fate specification. Not
genesis, including stem cell renewal, neuronal surprisingly, Setdb1 knockout in vivo resulted in
fate specification, as well as maturation and inte- severe brain defects and early postnatal lethality
gration (Ma etal.,2010). (Tan etal., 2012). Chromatin remodelers are also
During development, multipotent neural implicated in early neurogenesis and CNS devel-
stem cells give rise to a wide range of cell types opment. Mice depleted of major components of
within the CNS along the body axis. Homeobox the SWI/SNF chromatin remodeling complex die
(Hox) genes are key regulators of neural stem cell at peri-implantation stages. Furthermore, many
renewal and lineage commitment and determine heterozygotes display neural tube abnormali-
not only the identity of individual cells within ties (Bultman et al., 2000) and are predisposed
the anterior-posterior and dorso-ventral axes to tumors of neural origin (Bultman etal., 2000;
of the brain, but also direct cellular migration, Kim etal.,2001).
axonal direction and the process of somatogene- Prenatal and early postnatal periods in mam-
sis during embryogenesis and development (Park malian development are characterized by rapid
etal.,2007). changes in neuronal organization, providing a
Epigenetics 193

critical time window during which environmen- S Y N A P T I C P L A S T I C I T Y,


tal factors (i.e., intrinsic and extrinsic stimuli) LEARNING, ANDMEMORY
can lead to long-term influences on brain devel- Epigenetic modifications on chromosomal DNA
opment and adapting brain function. Epigenetic do not only comprise the molecular memory of
mechanisms seem to play a critical role in a single cell but assist in learning and memory
mediating the effects of (early) sensory experi- as higher order brain functions. Histone acety-
ence on gene expression in the brain. Prenatal lation is regarded as a promoter of memory for-
stress is associated with an increased vulner- mation, whereas lack of this modification has
ability to neurodevelopmental and psychiat- been causally related to cognitive impairment in
ric disorders, including autism, schizophrenia, neurodevelopmental disorders, neurodegenera-
and depression (Champagne and Curley, 2009). tion, and aging (Graff and Tsai, 2013; Peleg etal.,
Experimental studies in humans and mice have 2010). Specific forms of learning in rodents have
demonstrated that prenatal exposure to mater- been correlated with increased HAT activity and
nal stress as well as early life stress in the infant global histone acetylation, and more recently
induces localized and global changes in DNA the role of specific histone modifications (e.g.,
methylation associated with stable changes in H4K12 and H4K5 acetylation) has been investi-
gene expression, which emerge in infancy and gated (Fischer, 2014; Park etal., 2013; Peleg etal.,
are sustained into adulthood, proving a lasting 2010). Given that neurodegenerative diseases are
effect on infant development (Champagne and frequently associated with impaired learning
Curley, 2009; Fagiolini et al., 2009; Suderman and memory, the question arises regarding how
et al., 2014). Nutrition is another environmen- learning ability and access to long-term memo-
tal factor, which exerts a strong impact on CNS ries can be improved. Studies in mice provided
development and function. One critical meta- evidence that both environmental enrichment
bolic pathway is the methionine cycle, in which as well as pharmacological inhibition of histone
folate and B12 convert homocysteine to methio- deacetylases (HDACs) re-established histone-tail
nine, which is in turn converted to SAM the acetylation and correlated well with increased
methyl group donor in all transmethylation reac- sprouting of dendrites, increased number of
tions (Kobow and Blumcke, 2012). Intriguingly, synapses, as well as reinstated learning behav-
the folate-dependent methionine cycle plays a ior and access to long-term memories (Fischer
key role in neuronal homeostasis. Dietary folate et al., 2007; Peleg et al., 2010). Interestingly,
deficiency or genetic defects in folate metabolism Hdac2 as well as Hdac3 knockout mice showed
inhibit the methionine cycle, thereby increas- enhanced learning behavior, whereas mice
ing developmental abnormalities of the fetal and lacking Hdac4 in the adult forebrain exhibited
neonatal nervous system (Chen etal., 2001), and impaired hippocampus-dependent memory for-
have been associated with autism and epilepsy in mation. Likewise, haploinsufficiency of HDAC4
youth, as well as depression, schizophrenia, and in humans is linked to mental retardation
neurodegeneration in the adult (Goyette et al., (Fischer,2014).
1995; Iskandar et al., 2010; Popp et al., 2009; Rubinstein-Taybi syndrome, a congenital
Shea and Rogers, 2014; Tonetti etal., 2000). SAM anomaly syndrome characterized by intellectual
is required for maintaining DNA methylation disability, postnatal growth deficiency, micro-
levels during critical periods of high neuronal cephaly, and dysmorphic facial features, is caused
proliferation. Furthermore, homocysteine is a by heterozygous mutations in the gene encoding
neurotoxic agent that, if accumulating in the the transcriptional co-activator CREB-binding
methionine cycle, can induce aberrant activation protein (CREBBP; Petrij et al., 1995). CREBBP
of NMDA receptors, calcium influx, and oxida- is known to possess intrinsic HAT activity and
tive damage (Shea and Rogers,2014). plays a critical role in embryonic development,
Taken together, there is converging evidence growth control, and neuronal homeostasis by
for a role of multiple epigenetic mechanisms dur- coupling chromatin remodeling to transcrip-
ing all stages of brain development and neuronal tion factor recognition (Roelfsema et al., 2005).
homeostasis. Perturbations in epigenetic gene Although CREBBP is the most thoroughly stud-
regulation, whether driven by genetic or environ- ied HAT with regard to learning and memory,
mental factors, may have lasting effects on syn- recent studies have shown a role for E1A-binding
aptic function, neuronal network activity, and protein (P300) and P300/CREBBP-associated
proliferation and can be deleterious to the nerv- factor (PCAF) in memory processes (Barrett and
ous system (Ramocki and Zoghbi,2008). Wood, 2008). Genetic mouse models targeting
194 Part II:Homeostatic Control

either one of the three factors display impaired system and play a significant role in learning and
memory consolidation, although mechanisms memory. However, these phenomena are mecha-
underlying this effect seem to be poorly under- nistically not yet well understood.
stood. So far CREBBP function has been linked
to increased calcium (Ca2+) signaling and tran- NEUROLOGIC DISEASE
scriptional activation as well as synaptic plas- Epigenetic changes are fundamental in normal
ticity. Beside its ability to acetylate histone development, and disruption of the epigenetic
and nonhistone proteins, CREBBP selectively program can unchain a variety of pathogenic
recruits transcriptional co-activators during the processes. Cancer was the first disease described
formation of the transcription preinitiation com- for altered epigenetic marks. Epigenetic silencing
plex and has therefore been suggested to act as of tumor suppressor genes by promoter methyla-
scaffold to stabilize protein interactions with the tion is a common event also during brain tumor
transcription machinery. development (Hegi et al., 2009). Epigenetic
Histone methylation is another key regula- alterations in tumors frequently target essen-
tor for hippocampal and cortical gene activation tial components of DNA repair pathways (e.g.
and silencing during memory consolidation. O6-methylguanine-DNA methyltransferase
Conditional knockout of histone methylating (MGMT). Most interestingly, these alterations
and demethylating enzymes (e.g., euchromatic sensitize affected tumors to treatment with cer-
histone-lysine n-methyltransferase 2, Ehmt2; tain classes of anticancer agents (e.g., alkylat-
lysine-specific methyltransferase 2a and 2b, ing agents). Consequently, MGMT promoter
Kmt2a/2b) in adult mice forebrain or hippocam- methylation in glioblastoma patients has been
pus resulted in altered histone methylation and associated with overall longer survival and
corresponding gene expression changes, as well better response to chemotherapy (Hegi et al.,
as complex behavioral abnormalities and learn- 2005). More recently, recurrent somatic muta-
ing impairment. Brain morphology remained tions in isocitrate dehydrogenase (IDH) genes
unaffected, however (Gupta-Agarwal et al., have been associated with glioblastoma but also
2012; Gupta etal., 2010; Kerimoglu etal., 2013). low-grade gliomas (Sun et al., 2013b; Yan et al.,
Evidence for DNA methylation in learning and 2009). Cytoplasmic IDH1 and mitochondrial
memory comes from studies showing that Dnmt IDH2 isoforms normally catalyse the intercon-
inhibition blocked hippocampal memory forma- version of isocitrate and -ketoglutarate but also
tion associated with deregulated expression of play a role in lipid synthesis, glucose sensing, or
genes known to contribute to synaptic plasticity energy metabolism. Moreover, IDH enzymes
(Feng etal., 2010; Miller and Sweatt, 2007). Fear regulate redox homeostasis and protect the cell
conditioning in mice increased the expression against oxidative stress by generating the reduc-
of Dnmt3a and Dnmt3b and, moreover, induced ing equivalent NAPDH. NADPH is essential for
localized changes in DNA methylation patterns the regeneration of glutathione that neutralizes
and concomitant changes in gene expression free radicals and reactive oxygen species and
(Miller and Sweatt, 2007). Ten-eleven transloca- for the activity of the thioredoxin system.
tion family enzymes, which promote active DNA Although the pentose phosphate pathway is the
demethylation in the nervous system, have also main source of NADPH, IDH1/2 are similarly
been shown to regulate the expression of CNS important (Mellai et al., 2013). Redox homeo-
activity-dependent genes and memory formation stasis is linked to transmethylation and recy-
(Kaas etal., 2013). Genetic deletion of Gadd45b, cling of SAM as universal methyl group donor.
another regulator of active DNA demethyla- Under oxidative conditions, SAM synthetase
tion, enhances long-term memory and synaptic can become inactivated, thus stopping the flow
plasticity (Sultan et al., 2012). Finally, noncod- of metabolites through the methionine cycle and
ing RNAs are also implicated in cognitive brain inhibiting, for example, DNA or histone meth-
function (e.g., PIWI-interacting RNAs were ylation (Hitchler and Domann, 2012). In addi-
recently identified to be abundantly expressed tion, -ketoglutarate is an essential cofactor of
in the CNS and mediate activity-induced CpG ten-eleven translocation family enzymes and of
methylation and transcriptional silencing of key Jumonji C domain containing histone demethy-
synaptic plasticity-related genes). lases. Mutations targeting IDH1/2 as much as
In conclusion, epigenetic mechanisms includ- changes in the expression or activity of IDH1/2
ing histone modifications and DNA methylation enzymes may therefore globally alter 5-mC and
are dynamically regulated in the adult nervous 5-hmC levels (Chia etal.,2011).
Epigenetics 195

Other than brain tumors, many other neu- can induce epigenetic alterations, resulting in
rological diseases primarily or secondarily sustained changes of gene expression, thereby
associate with epigenetic alterations, including promoting and enhancing the epileptogenic
autism, bipolar disorder, schizophrenia, neu- condition (Kobow and Blumcke, 2011, 2012).
rodegeneration, stroke, and epilepsy (Kobow In genetically determined human epilepsies,
and Blumcke, 2012; Kobow etal., 2013; Mehler, it can further be noted that all different types
2008; Moon et al., 2013; Qureshi and Mehler, of epigenetic players have been identified to
2010). As mentioned, epigenetic mechanisms be affected by mutations including chromatin
affect cell fate determination and maturation, remodelers (e.g., chromodomain helicase DNA
migration, and lamination. More recent stud- binding protein 2, CHD2; SWI/SNF-related,
ies suggest neuronal membrane excitation, matrix associated, actin dependent regulator of
memory formation, and cognition are linked chromatin, subfamily A, member 4, SMARCA4;
to epigenetic chromatin modifications, thereby Alpha-thalassemia/mental retardation,
adapting gene expression levels to functional ATRX), histone modifiers (e.g. euchromatic
network activity (Dulac, 2010; Fischer et al., histone-lysine N-methyltransferase 1, EHMT1;
2007; Peleg et al., 2010; Qureshi and Mehler, lysine-specific demethylase 5C, KDM5C; his-
2010). In human and experimental epilepsy, tone deacetylase 4, HDAC4) as well as readers
localized and global changes in DNA methyla- of DNA methylation (e.g. methyl CpG binding
tion have been identified and correlated with protein 2, MeCP2; Kobow and Blumcke,2012).
gene expression changes. Prominent examples Taken together, a two-tailed interdepend-
of genes sensitive to DNA methylation-based ence between epigenetics and brain function
silencing in experimental and human epi- can be assumed that allows alterations in epi-
lepsy include Reelin (Reln), a master regulator genetics to be a cause as well as a consequence
of neuronal migration (Kobow et al., 2009), of brain malfunction and disease. In complex
and carboxypeptidase A6 (CPA6), an enzyme diseases, knowledge about epigenetic altera-
involved in the selective biosynthesis of neu- tions can probably elucidate the origin of
roendocrine peptides (Belhedi etal., 2014; Sapio some non-Mendelian inheritance and etiology
etal., 2012). Genome-wide methylation analysis beyond genetic mutations. Phenotypic vari-
in rodent models of epilepsy identified meth- ation and disease susceptibility, response to
ylation signatures distinguishing animals that treatment, and the complex outcome of com-
experienced status epilepticus as an initial pre- mon and rare diseases may be compounded by
cipitating injury from chronic epileptic animals both genetic and epigenetic anomalies as well
and/or healthy controls (Kobow et al., 2013; as the interaction between them (Huidobro
Miller-Delaney et al., 2012). Increased DNA etal.,2013).
methylation in human and experimental epi-
lepsy could be associated with either increased CONCLUSIONS
DNA methyltransferase expression or increased Epigenetic mechanisms comprise a large spec-
enzymatic activity (Williams-Karnesky et al., trum of DNA-modifying molecular mecha-
2013; Zhu etal.,2011). nisms and play a major role in adapting brain
Ca2+ is a major second messenger that helps function to environmental stimuli during
to transmit depolarization status and synap- development and adolescence but also aging.
tic activity to the biochemical machinery of a Epigenetic alterations have been consistently
given neuron. Neuronal hyperactivity, as dur- identified in many neurological diseases, open-
ing seizures, is intricately linked to altered ing new avenues to drug treatment, as the key
Ca2+ and redox homeostasis, neuronal energy molecular players represent promising targets
metabolism, and second messenger signal- for pharmacological intervention. Currently,
ing to the nucleus. All of these processes feed global effects of epigenetic inhibitors impede
each other and impact epigenetic processes, normal cellular functions, which is a clear limi-
and some examples have been more closely dis- tation, and severe side effects must be envis-
cussed in this chapter. It has previously been aged for unselective systemic epigenetic drug
suggested that initial precipitating injuries treatment (Frisch et al., 2009; Harden et al.,
(e.g., traumatic brain injury, inflammation, 2009). Therefore, future studies need to address
prolonged febrile seizures, or status epilepti- how the enzymology can be specifically tar-
cus), which are frequently observed in acquired geted in the affected brain region or even at
epilepsies, as much as seizures by themselves selectedgenes.
196 Part II:Homeostatic Control
References aortic lipid deposition. Hum Mol Genet 10,
Barres, R., Osler, M.E., Yan, J., Rune, A., Fritz, T., 433443.
Caidahl, K., Krook, A., and Zierath, J.R. (2009). Chia, N., Wang, L., Lu, X., Senut, M.C., Brenner, C., and
Non-CpG methylation of the PGC-1alpha pro- Ruden, D.M. (2011). Hypothesis:Environmental
moter through DNMT3B controls mitochon- regulation of 5-hydroxymethylcytosine by oxi-
drial density. Cell Metab 10, 189198. dative stress. Epigenetics 6, 853856.
Barrett, R.M., and Wood, M.A. (2008). Beyond tran- Clark, S.J., Harrison, J., and Frommer, M. (1995).
scription factors:The role of chromatin modify- CpNpG methylation in mammalian cells. Nat
ing enzymes in regulating transcription required Genet 10,2027.
for memory. Lern Mem 15, 460467. Cuellar, T.L., and McManus, M.T. (2005). MicroRNAs
Barski,A.,Cuddapah,S.,Cui,K.,Roh,T.Y.,Schones,D.E., and endocrine biology. J Endocrinol 187, 327332.
Wang, Z., Wei, G., Chepelev, I., and Zhao, K. Davie, J.R., and Murphy, L.C. (1990). Level of ubiq-
(2007). High-resolution profiling of histone meth- uitinated histone H2B in chromatin is coupled
ylations in the human genome. Cell 129, 823837. to ongoing transcription. Biochemistry 29,
Belhedi, N., Perroud, N., Karege, F., Vessaz, M., 47524757.
Malafosse, A., and Salzmann, A. (2014). Increased Davis, T.H., Cuellar, T.L., Koch, S.M., Barker, A.J.,
CPA6 promoter methylation in focal epilepsy and Harfe, B.D., McManus, M.T., and Ullian, E.M.
in febrile seizures. Epilepsy Res 108, 144148. (2008). Conditional loss of Dicer disrupts cellu-
Bergman, Y., and Cedar, H. (2013). DNA methyla- lar and tissue morphogenesis in the cortex and
tion dynamics in health and disease. Nat Struct hippocampus. J Neurosci 28, 43224330.
Mol Biol 20, 274281. de Napoles, M., Mermoud, J.E., Wakao, R., Tang,
Bernstein, B.E., Mikkelsen, T.S., Xie, X., Kamal, M., Y.A., Endoh, M., Appanah, R., Nesterova, T.B.,
Huebert, D.J., Cuff, J., Fry, B., Meissner, A., Silva, J., Otte, A.P., Vidal, M., et al. (2004).
Wernig, M., Plath, K., et al. (2006). A bivalent Polycomb group proteins ring1A/B link ubiqui-
chromatin structure marks key developmental tylation of histone H2A to heritable gene silenc-
genes in embryonic stem cells. Cell 125, 315326. ing and X inactivation. Dev Cell 7, 663676.
Bestor, T.H. (2000). The DNA methyltransferases of De Pietri Tonelli, D., Pulvers, J.N., Haffner, C.,
mammals. Hum Mol Genet 9, 23952402. Murchison, E.P., Hannon, G.J., and Huttner,
Bird, A. (2002). DNA methylation patterns and epi- W.B. (2008). miRNAs are essential for survival
genetic memory. Genes Dev 16,621. and differentiation of newborn neurons but not
Brinkman, A.B., Gu, H., Bartels, S.J., Zhang, Y., for expansion of neural progenitors during early
Matarese, F., Simmer, F., Marks, H., Bock, C., neurogenesis in the mouse embryonic neocortex.
Gnirke, A., Meissner, A., etal. (2012). Sequential Development 135, 39113921.
ChIP-bisulfite sequencing enables direct Deaton, A.M., and Bird, A. (2011). CpG islands and
genome-scale investigation of chromatin and the regulation of transcription. Genes Dev 25,
DNA methylation cross-talk. Genome Res 22, 10101022.
11281138. Di Lorenzo, A., and Bedford, M.T. (2011). Histone
Buck-Koehntop, B.A., and Defossez, P.A. (2013). On arginine methylation. FEBS Lett 585, 20242031.
how mammalian transcription factors recognize Dulac, C. (2010). Brain function and chromatin plas-
methylated DNA. Epigenetics 8, 131137. ticity. Nature 465, 728735.
Bultman, S., Gebuhr, T., Yee, D., La Mantia, C., Eckhardt, F., Lewin, J., Cortese, R., Rakyan, V.K.,
Nicholson, J., Gilliam, A., Randazzo, F., Metzger, D., Attwood, J., Burger, M., Burton, J., Cox, T.V.,
Chambon, P., Crabtree, G., etal. (2000). A Brg1 Davies, R., Down, T.A., etal. (2006). DNA meth-
null mutation in the mouse reveals functional ylation profiling of human chromosomes 6, 20
differences among mammalian SWI/SNF com- and 22. Nat Genet 38, 13781385.
plexes. Mol Cell 6, 12871295. Edwards, C.A., and Ferguson-Smith, A.C. (2007).
Champagne, F.A., and Curley, J.P. (2009). Epigenetic Mechanisms regulating imprinted genes in clus-
mechanisms mediating the long-term effects ters. Curr Opin Cell Biol 19, 281289.
of maternal care on development. Neurosci Edwards, J.R., ODonnell, A.H., Rollins, R.A.,
Biobehav Rev 33, 593600. Peckham, H.E., Lee, C., Milekic, M.H., Chanrion,
Chen, Z., Karaplis, A.C., Ackerman, S.L., Pogribny, I.P., B., Fu, Y., Su, T., Hibshoosh, H., et al. (2010).
Melnyk, S., Lussier-Cacan, S., Chen, M.F., Pai, A., Chromatin and sequence features that define the
John, S.W., Smith, R.S., et al. (2001). Mice defi- fine and gross structure of genomic methylation
cient in methylenetetrahydrofolate reductase patterns. Genome Res 20, 972980.
exhibit hyperhomocysteinemia and decreased Engel, N., Tront, J.S., Erinle, T., Nguyen, N., Latham,
methylation capacity, with neuropathology and K.E., Sapienza, C., Hoffman, B., and Liebermann,
Epigenetics 197

D.A. (2009). Conserved DNA methylation in B., Ford, E., etal. (2011a). Neuronal activity mod-
Gadd45a(/) mice. Epigenetics 4,9899. ifies the DNA methylation landscape in the adult
Fagiolini, M., Jensen, C.L., and Champagne, F.A. brain. Nat Neurosci 14, 13451351.
(2009). Epigenetic influences on brain develop- Guo, J.U., Su, Y., Zhong, C., Ming, G.L., and Song,
ment and plasticity. Curr Opin Neurobiol 19, H. (2011b). Hydroxylation of 5-methylcytosine
207212. by TET1 promotes active DNA demethylation in
Fasano, C.A., Phoenix, T.N., Kokovay, E., Lowry, N., the adult brain. Cell 145, 423434.
Elkabetz, Y., Dimos, J.T., Lemischka, I.R., Studer, L., Gupta-Agarwal, S., Franklin, A.V., DeRamus, T.,
and Temple, S. (2009). Bmi-1 cooperates with Wheelock, M., Davis, R.L., McMahon, L.L.,
Foxg1 to maintain neural stem cell self-renewal and Lubin, F.D. (2012). G9a/GLP histone lysine
in the forebrain. Genes Dev 23, 561574. dimethyltransferase complex activity in the hip-
Feng, J., Zhou, Y., Campbell, S.L., Le, T., Li, E., Sweatt, pocampus and the entorhinal cortex is required
J.D., Silva, A.J., and Fan, G. (2010). Dnmt1 and for gene activation and silencing during memory
Dnmt3a maintain DNA methylation and regu- consolidation. J Neurosci 32, 54405453.
late synaptic function in adult forebrain neurons. Gupta, S., Kim, S.Y., Artis, S., Molfese, D.L.,
Nat Neurosci 13, 423430. Schumacher, A., Sweatt, J.D., Paylor, R.E., and
Fischer, A. (2014). Epigenetic memory: The Lubin, F.D. (2010). Histone Methylation regulates
Lamarckian brain. EMBO J 33, 945967. memory formation. J Neurosci 30, 35893599.
Fischer, A., Sananbenesi, F., Wang, X., Dobbin, M., Hahn, M.A., Qiu, R., Wu, X., Li, A.X., Zhang, H.,
and Tsai, L.H. (2007). Recovery of learning and Wang, J., Jui, J., Jin, S.G., Jiang, Y., Pfeifer, G.P., eta l.
memory is associated with chromatin remodel- (2013). Dynamics of 5-hydroxymethylcytosine
ling. Nature 447, 178182. and chromatin marks in Mammalian neurogen-
Fischle, W. (2008). Talk is cheapcross-talk in estab- esis. Cell Rep 3, 291300.
lishment, maintenance, and readout of chroma- Harden, C.L., Meador, K.J., Pennell, P.B., Hauser,
tin modifications. Genes Dev 22, 33753382. W.A., Gronseth, G.S., French, J.A., Wiebe, S.,
Flaus, A., and Owen-Hughes, T. (2011). Mechanisms Thurman, D., Koppel, B.S., Kaplan, P.W., et al.
for ATP-dependent chromatin remodelling:The (2009). Practice parameter update:Management
means to the end. FEBS J 278, 35793595. issues for women with epilepsyfocus on preg-
Fournier, A., Sasai, N., Nakao, M., and Defossez, P.A. nancy (an evidence-based review):Teratogenesis
(2012). The role of methyl-binding proteins in and perinatal outcomes: Report of the Quality
chromatin organization and epigenome mainte- Standards Subcommittee and Therapeutics and
nance. Brief Funct Genomics 11, 251264. Technology Assessment Subcommittee of the
Frisch, C., Husch, K., Angenstein, F., Kudin, A., American Academy of Neurology and American
Kunz, W., Elger, C.E., and Helmstaedter, C. Epilepsy Society. Neurology 73, 133141.
(2009). Dose-dependent memory effects and He, L., and Hannon, G.J. (2004). MicroRNAs:Small
cerebral volume changes after in utero exposure RNAs with a big role in gene regulation. Nat Rev
to valproate in the rat. Epilepsia 50, 14321441. Genet 5, 522531.
Gong, Z., and Zhu, J.K. (2011). Active DNA demeth- Hegi, M.E., Diserens, A.C., Gorlia, T., Hamou, M.F.,
ylation by oxidation and repair. Cell Res 21, de Tribolet, N., Weller, M., Kros, J.M., Hainfellner,
16491651. J.A., Mason, W., Mariani, L., etal. (2005). MGMT
Goyette, P., Frosst, P., Rosenblatt, D.S., and Rozen, R. gene silencing and benefit from temozolomide in
(1995). Seven novel mutations in the methylene- glioblastoma. N Engl J Med 352, 9971003.
tetrahydrofolate reductase gene and genotype/ Hegi, M.E., Sciuscio, D., Murat, A., Levivier, M., and
phenotype correlations in severe methylenetet- Stupp, R. (2009). Epigenetic deregulation of DNA
rahydrofolate reductase deficiency. Am J Hum repair and its potential for therapy. Clin Cancer
Genet 56, 10521059. Res 15, 50265031.
Graff, J., and Tsai, L.H. (2013). Histone acetyla- Heintzman, N.D., Stuart, R.K., Hon, G., Fu, Y.,
tion: Molecular mnemonics on the chromatin. Ching, C.W., Hawkins, R.D., Barrera, L.O., Van
Nat Rev Neurosci 14, 97111. Calcar, S., Qu, C., Ching, K.A., et al. (2007).
Grandjean, V., Yaman, R., Cuzin, F., and Rassoulzadegan, Distinct and predictive chromatin signatures of
M. (2007). Inheritance of an epigenetic mark:The transcriptional promoters and enhancers in the
CpG DNA methyltransferase 1 is required for human genome. Nat Genet 39, 311318.
de novo establishment of a complex pattern of Hengst, U., Cox, L.J., Macosko, E.Z., and Jaffrey, S.R.
non-CpG methylation. PLoS One 2,e1136. (2006). Functional and selective RNA interfer-
Guo, J.U., Ma, D.K., Mo, H., Ball, M.P., Jang, M.H., ence in developing axons and growth cones. J
Bonaguidi, M.A., Balazer, J.A., Eaves, H.L., Xie, Neurosci 26, 57275732.
198 Part II:Homeostatic Control

Hirabayashi, Y., Suzki, N., Tsuboi, M., Endo, T.A., H., and Sweatt, J.D. (2013). TET1 controls CNS
Toyoda, T., Shinga, J., Koseki, H., Vidal, M., and 5-methylcytosine hydroxylation, active DNA
Gotoh, Y. (2009). Polycomb limits the neurogenic demethylation, gene transcription, and memory
competence of neural precursor cells to promote formation. Neuron 79, 10861093.
astrogenic fate transition. Neuron 63, 600613. Kawase-Koga, Y., Otaegi, G., and Sun, T. (2009).
Hitchler, M.J., and Domann, F.E. (2012). Redox Different timings of Dicer deletion affect neu-
regulation of the epigenetic landscape in cancer: rogenesis and gliogenesis in the developing
A role for metabolic reprogramming in remod- mouse central nervous system. Dev Dyn 238,
eling the epigenome. Free Radic Biol Med 53, 28002812.
21782187. Kerimoglu, C., Agis-Balboa, R.C., Kranz, A.,
Huidobro, C., Fernandez, A.F., and Fraga, M.F. Stilling, R., Bahari-Javan, S., Benito-Garagorri,
(2013). The role of genetics in the establishment E., Halder, R., Burkhardt, S., Stewart, A.F., and
and maintenance of the epigenome. Cell Mol Life Fischer, A. (2013). Histone-methyltransferase
Sci 70(9), 15431573. MLL2 (KMT2B) is required for memory forma-
Ichiyanagi, T., Ichiyanagi, K., Miyake, M., and tion in mice. J Neurosci 33, 34523464.
Sasaki, H. (2013). Accumulation and loss of Khorasanizadeh, S. (2004). The nucleosome: From
asymmetric non-CpG methylation during male genomic organization to genomic regulation.
germ-cell development. Nucleic Acids Res 41, Cell 116, 259272.
738745. Kim, J.K., Huh, S.-O., Choi, H., Lee, K.-S., Shin, D.,
Ikura, T., Tashiro, S., Kakino, A., Shima, H., Jacob, Lee, C., Nam, J.-S., Kim, H., Chung, H., Lee,
N., Amunugama, R., Yoder, K., Izumi, S., H.W., et al. (2001). Srg3, a mouse homolog of
Kuraoka, I., Tanaka, K., et al. (2007). DNA yeast SWI3, is essential for early embryogenesis
damage-dependent acetylation and ubiquitina- and involved in brain development. Mol Cell Biol
tion of H2AX enhances chromatin. Dynamics. 21, 77877795.
Mol Cell Biol 27, 70287040. Kim, V.N., Han, J., and Siomi, M.C. (2009). Biogenesis
Illingworth, R.S., Gruenewald-Schneider, U., Webb, S., of small RNAs in animals. Nat Rev Mol Cell Biol
Kerr, A.R., James, K.D., Turner, D.J., Smith, C., 10, 126139.
Harrison, D.J., Andrews, R., and Bird, A.P. Kobow, K., and Blumcke, I. (2011). The methylation
(2010). Orphan CpG islands identify numer- hypothesis: Do epigenetic chromatin modifica-
ous conserved promoters in the mammalian tions play a role in epileptogenesis? Epilepsia 52
genome. PLoS Genet 6, e1001134. Suppl 4,1519.
Iskandar, B.J., Rizk, E., Meier, B., Hariharan, N., Kobow, K., and Blumcke, I. (2012). The emerging role
Bottiglieri, T., Finnell, R.H., Jarrard, D.F., of DNA methylation in epileptogenesis. Epilepsia
Banerjee, R.V., Skene, J.H., Nelson, A., et al. 53 Suppl 9,1120.
(2010). Folate regulation of axonal regeneration Kobow, K., Jeske, I., Hildebrandt, M., Hauke, J.,
in the rodent central nervous system through Hahnen, E., Buslei, R., Buchfelder, M., Weigel,
DNA methylation. J Clin Invest 120, 16031616. D., Stefan, H., Kasper, B., etal. (2009). Increased
Jaenisch, R., and Bird, A. (2003). Epigenetic regula- reelin promoter methylation is associated with
tion of gene expression: How the genome inte- granule cell dispersion in human temporal lobe
grates intrinsic and environmental signals. Nat epilepsy. J Neuropathol Exp Neurol 68, 356364.
Genet 33 Suppl, 245254. Kobow, K., Kaspi, A., Harikrishnan, K.N., Kiese, K.,
Jobe, E.M., McQuate, A.L., and Zhao, X. (2012). Ziemann, M., Khurana, I., Fritzsche, I., Hauke,
Crosstalk among epigenetic pathways regulates J., Hahnen, E., Coras, R., et al. (2013). Deep
neurogenesis. Front Neurosci6,59. sequencing reveals increased DNA methylation
Jrgensen, S., Schotta, G., and Srensen, C.S. (2013). in chronic rat epilepsy. Acta Neuropathol 126(5),
Histone H4 Lysine 20 methylation: Key player 741756.
in epigenetic regulation of genomic integrity. Kouzarides, T. (2007). Chromatin modifications and
Nucleic Acids Res 41, 27972806. their function. Cell 128, 693705.
Jovicic, A., Roshan, R., Moisoi, N., Pradervand, S., Lachner, M., and Jenuwein, T. (2002). The many faces
Moser, R., Pillai, B., and Luthi-Carter, R. (2013). of histone lysine methylation. Curr Opin Cell
Comprehensive expression analyses of neural Biol 14, 286298.
cell-type-specific miRNAs identify new deter- Lee, J.S., Shukla, A., Schneider, J., Swanson, S.K.,
minants of the specification and maintenance of Washburn, M.P., Florens, L., Bhaumik, S.R., and
neuronal phenotypes. J Neurosci 33, 51275137. Shilatifard, A. (2007). Histone crosstalk between
Kaas, G.A., Zhong, C., Eason, D.E., Ross, D.L., H2B monoubiquitination and H3 methylation
Vachhani, R.V., Ming, G.L., King, J.R., Song, mediated by COMPASS. Cell 131, 10841096.
Epigenetics 199

Lehnertz, B., Ueda, Y., Derijck, A.A., Braunschweig, U., mutations in gliomas (Rijeka, Croatia: Intech
Perez-Burgos, L., Kubicek, S., Chen, T., Li, E., Open Access).
Jenuwein, T., and Peters, A.H. (2003). Suv39h- Mercer, T.R., Dinger, M.E., and Mattick, J.S. (2009).
mediated histone H3 lysine 9 methylation directs Long non-coding RNAs:Insights into functions.
DNA methylation to major satellite repeats Nat Rev Genet 10, 155159.
at pericentric heterochromatin. Curr Biol 13, Miller-Delaney, S.F., Das, S., Sano, T.,
11921200. Jimenez-Mateos, E.M., Bryan, K., Buckley,
Levenson, J.M., Roth, T.L., Lubin, F.D., Miller, C.A., P.G., Stallings, R.L., and Henshall, D.C. (2012).
Huang, I.C., Desai, P., Malone, L.M., and Sweatt, Differential DNA methylation patterns define
J.D. (2006). Evidence that DNA (cytosine-5) status epilepticus and epileptic tolerance. J
methyltransferase regulates synaptic plasticity in Neurosci 32, 15771588.
the hippocampus. J Biol Chem 281, 1576315773. Miller, C.A., and Sweatt, J.D. (2007). Covalent modi-
Lister, R., Mukamel, E.A., Nery, J.R., Urich, M., fication of DNA regulates memory formation.
Puddifoot, C.A., Johnson, N.D., Lucero, J., Neuron 53, 857869.
Huang, Y., Dwork, A.J., Schultz, M.D., et al. Minkovsky, A., Patel, S., and Plath, K. (2012).
(2013). Global epigenomic reconfiguration dur- Concise review:Pluripotency and the transcrip-
ing mammalian brain development. Science 341, tional inactivation of the female Mammalian X
1237905. chromosome. Stem Cells 30,4854.
Lopez-Serra, P., and Esteller, M. (2012). DNA Molofsky, A.V., Pardal, R., Iwashita, T., Park, I.K.,
methylation-associated silencing of tumor- Clarke, M.F., and Morrison, S.J. (2003). Bmi-1
suppressor microRNAs in cancer. Oncogene 31, dependence distinguishes neural stem cell
16091622. self-renewal from progenitor proliferation.
Lunyak, V.V., Burgess, R., Prefontaine, G.G., Nelson, C., Nature 425, 962967.
Sze, S.H., Chenoweth, J., Schwartz, P., Pevzner, P.A., Moon, J.M., Xu, L., and Giffard, R.G. (2013).
Glass, C., Mandel, G., etal. (2002). Corepressor- Inhibition of microRNA-181 reduces forebrain
dependent silencing of chromosomal regions ischemia-induced neuronal loss. J Cereb Blood
encoding neuronal genes. Science 298, Flow Metab 33, 19761982.
17471752. Nelson, E.D., Kavalali, E.T., and Monteggia, L.M.
Luteijn, M.J., and Ketting, R.F. (2013). PIWI- (2008). Activity-dependent suppression of min-
interacting RNAs: From generation to transgen- iature neurotransmission through the regulation
erational epigenetics. Nat Rev Genet 14, 523534. of DNA methylation. J Neurosci 28, 395406.
Ma, D.K., Marchetto, M.C., Guo, J.U., Ming, G.L., Numata, S., Ye, T., Hyde, T.M., Guitart-Navarro,
Gage, F.H., and Song, H. (2010). Epigenetic cho- X., Tao, R., Wininger, M., Colantuoni, C.,
reographers of neurogenesis in the adult mam- Weinberger, D.R., Kleinman, J.E., and Lipska,
malian brain. Nat Neurosci 13, 13381344. B.K. (2012). DNA methylation signatures in
Madrigano, J., Baccarelli, A., Mittleman, M.A., development and aging of the human prefrontal
Sparrow, D., Vokonas, P.S., Tarantini, L., cortex. Am J Hum Genet 90, 260272.
and Schwartz, J. (2012). Aging and epi- Olde Loohuis, N.F., Kos, A., Martens, G.J., Van
genetics: Longitudinal changes in gene-specific Bokhoven, H., Nadif Kasri, N., and Aschrafi,
DNA methylation. Epigenetics 7,6370. A. (2012). MicroRNA networks direct neuronal
Martin, C., and Zhang, Y. (2005). The diverse func- development and plasticity. Cell Mol Life Sci 69,
tions of histone lysine methylation. Nat Rev Mol 89102.
Cell Biol 6, 838849. Ooi, S.K., and Bestor, T.H. (2008). The colorful his-
Martinez-Garcia, E., and Licht, J.D. (2010). tory of active DNA demethylation. Cell 133,
Deregulation of H3K27 methylation in cancer. 11451148.
Nat Genet 42, 100101. Park, C.S., Rehrauer, H., and Mansuy, I.M. (2013).
Maunakea, A.K., Nagarajan, R.P., Bilenky, M., Genome-wide analysis of H4K5 acetylation
Ballinger, T.J., DSouza, C., Fouse, S.D., Johnson, associated with fear memory in mice. BMC
B.E., Hong, C., Nielsen, C., Zhao, Y., etal. (2010). Genomics 14,539.
Conserved role of intragenic DNA methylation Park, S.Y., Kim, J.B., and Han, Y.-M. (2007). REST
in regulating alternative promoters. Nature 466, is a key regulator in brain-specific homeobox
253257. gene expression during neuronal differentiation.
Mehler, M.F. (2008). Epigenetics and the nervous J Neurochem 103, 25652574.
system. Ann Neurol 64, 602617. Peleg, S., Sananbenesi, F., Zovoilis, A., Burkhardt,
Mellai, M., Caldera, V., Annovazzi, L., and Schiffer, D. S., Bahari-Javan, S., Agis-Balboa, R.C., Cota,
(2013). The distribution and significance of IDH P., Wittnam, J.L., Gogol-Doering, A., Opitz, L.,
200 Part II:Homeostatic Control

etal. (2010). Altered histone acetylation is associ- Sapio, M.R., Salzmann, A., Vessaz, M., Crespel,
ated with age-dependent memory impairment in A., Lyons, P.J., Malafosse, A., and Fricker, L.D.
mice. Science 328, 753756. (2012). Naturally occurring carboxypeptidase
Pereira, J.D., Sansom, S.N., Smith, J., Dobenecker, A6 mutations: Effect on enzyme function and
M.W., Tarakhovsky, A., and Livesey, F.J. (2010). association with epilepsy. J Biol Chem 287,
Ezh2, the histone methyltransferase of PRC2, 4290042909.
regulates the balance between self-renewal and Sarcinella, E., Zuzarte, P.C., Lau, P.N., Draker, R.,
differentiation in the cerebral cortex. Proc Natl and Cheung, P. (2007). Monoubiquitylation of
Acad Sci USA 107, 1595715962. H2A.Z distinguishes its association with euchro-
Petrij, F., Giles, R.H., Dauwerse, H.G., Saris, J.J., matin or facultative heterochromatin. Mol Cell
Hennekam, R.C., Masuno, M., Tommerup, N., Biol 27, 64576468.
van Ommen, G.J., Goodman, R.H., Peters, D.J., Sati, S., Tanwar, V.S., Kumar, K.A., Patowary, A.,
etal. (1995). Rubinstein-Taybi syndrome caused Jain, V., Ghosh, S., Ahmad, S., Singh, M., Reddy,
by mutations in the transcriptional co-activator S.U., Chandak, G.R., et al. (2012). High resolu-
CBP. Nature 376, 348351. tion methylome map of rat indicates role of
Popp, J., Lewczuk, P., Linnebank, M., Cvetanovska, intragenic DNA methylation in identification of
G., Smulders, Y., Kolsch, H., Frommann, I., coding region. PLoS One 7, e31621.
Kornhuber, J., Maier, W., and Jessen, F. (2009). Schafer, A. (2013). Gadd45 proteins: Key players of
Homocysteine metabolism and cerebrospi- repair-mediated DNA demethylation. Adv Exp
nal fluid markers for Alzheimers disease. J Med Biol 793,3550.
Alzheimers Dis 18, 819828. Schratt, G.M., Tuebing, F., Nigh, E.A., Kane, C.G.,
Potts, R.C., Zhang, P., Wurster, A.L., Precht, P., Sabatini, M.E., Kiebler, M., and Greenberg,
Mughal, M.R., Wood, W.H., 3rd, Zhang, Y., M.E. (2006). A brain-specific microRNA regu-
Becker, K.G., Mattson, M.P., and Pazin, M.J. lates dendritic spine development. Nature 439,
(2011). CHD5, a brain-specific paralog of Mi2 283289.
chromatin remodeling enzymes, regulates Seo, S., Richardson, G.A., and Kroll, K.L. (2005).
expression of neuronal genes. PLoS One 6, e24515. The SWI/SNF chromatin remodeling protein
Qureshi, I.A., and Mehler, M.F. (2010). Epigenetic Brg1 is required for vertebrate neurogenesis and
mechanisms underlying human epileptic dis- mediates transactivation of Ngn and NeuroD.
orders and the process of epileptogenesis. Development 132, 105115.
Neurobiol Dis 39,5360. Shea, T.B., and Rogers, E. (2014). Lifetime require-
Ramocki, M.B., and Zoghbi, H.Y. (2008). Failure of ment of the methionine cycle for neuronal devel-
neuronal homeostasis results in common neuro- opment and maintenance. Curr Opin Psychiatry
psychiatric phenotypes. Nature 455, 912918. 27, 138142.
Rando, O.J. (2012). Combinatorial complexity in Smith, Z.D., and Meissner, A. (2013). DNA methyla-
chromatin structure and function:Revisiting the tion:Roles in mammalian development. Nat Rev
histone code. Curr Opin Genet Dev 22, 148155. Genet 14, 204220.
Reik, W. (2007). Stability and flexibility of epigenetic Suderman, M., Borghol, N., Pappas, J., Pinto Pereira,
gene regulation in mammalian development. S., Pembrey, M., Hertzman, C., Power, C., and
Nature 447, 425432. Szyf, M. (2014). Childhood abuse is associated
Reik, W., Dean, W., and Walter, J. (2001). Epigenetic with methylation of multiple loci in adult DNA.
reprogramming in mammalian development. BMC Med Genomics7,13.
Science 293, 10891093. Sultan, F.A., Wang, J., Tront, J., Liebermann, D.A.,
Reik, W., and Walter, J. (2001). Genomic imprint- and Sweatt, J.D. (2012). Genetic deletion of
ing:Parental influence on the genome. Nat Rev Gadd45b, a regulator of active DNA demethyl-
Genet 2,2132. ation, enhances long-term memory and synaptic
Roelfsema, J.H., White, S.J., Ariyurek, Y., Bartholdi, D., plasticity. J Neurosci 32, 1705917066.
Niedrist, D., Papadia, F., Bacino, C.A., den Sun, A.X., Crabtree, G.R., and Yoo, A.S. (2013a).
Dunnen, J.T., van Ommen, G.J., Breuning, MicroRNAs: Regulators of neuronal fate. Curr
M.H., et al. (2005). Genetic heterogeneity in Opin Cell Biol 25, 215221.
Rubinstein-Taybi syndrome: Mutations in both Sun, H., Yin, L., Li, S., Han, S., Song, G., Liu, N.,
the CBP and EP300 genes cause disease. Am J and Yan, C. (2013b). Prognostic significance
Hum Genet 76, 572580. of IDH mutation in adult low-grade glio-
Ronan, J.L., Wu, W., and Crabtree, G.R. (2013). From mas:Ameta-analysis. J Neurooncol 113, 277284.
neural development to cognition: Unexpected Szulwach, K.E., Li, X., Li, Y., Song, C.X., Wu, H., Dai,
roles for chromatin. Nat Rev Genet 14, 347359. Q., Irier, H., Upadhyay, A.K., Gearing, M., Levey,
Epigenetics 201

A.I., et al. (2011). 5-hmC-mediated epigenetic W., Zhang, M.Q., et al. (2008). Combinatorial
dynamics during postnatal neurodevelopment patterns of histone acetylations and methylations
and aging. Nat Neurosci 14, 16071616. in the human genome. Nat Genet 40, 897903.
Tan, S.L., Nishi, M., Ohtsuka, T., Matsui, T., Weber, M., Hellmann, I., Stadler, M.B., Ramos, L.,
Takemoto, K., Kamio-Miura, A., Aburatani, H., Paabo, S., Rebhan, M., and Schubeler, D. (2007).
Shinkai, Y., and Kageyama, R. (2012). Essential Distribution, silencing potential and evolution-
roles of the histone methyltransferase ESET in the ary impact of promoter DNA methylation in the
epigenetic control of neural progenitor cells dur- human genome. Nat Genet 39, 457466.
ing development. Development 139, 38063816. Williams-Karnesky, R.L., Sandau, U.S., Lusardi, T.A.,
Tardat, M., Murr, R., Herceg, Z., Sardet, C., and Lytle, N.K., Farrell, J.M., Pritchard, E.M.,
Julien, E. (2007). PR-Set7dependent lysine Kaplan, D.L., and Boison, D. (2013). Epigenetic
methylation ensures genome replication and sta- changes induced by adenosine augmentation
bility through S phase. J Cell Biol 179, 14131426. therapy prevent epileptogenesis. J Clin Invest
Tonetti, C., Burtscher, A., Bories, D., Tulliez, M., and 123, 35523563.
Zittoun, J. (2000). Methylenetetrahydrofolate Wu, H., Min, J., Lunin, V.V., Antoshenko, T.,
reductase deficiency in four siblings:Aclinical, Dombrovski, L., Zeng, H., Allali-Hassani, A.,
biochemical, and molecular study of the family. Campagna-Slater, V., Vedadi, M., Arrowsmith,
Am J Med Genet 91, 363367. C.H., etal. (2010). Structural biology of human
van der Lugt, N.M., Domen, J., Linders, K., van H3K9 methyltransferases. PLoS One 5,e8570.
Roon, M., Robanus-Maandag, E., te Riele, H., Wu, S.C., and Zhang, Y. (2010). Active DNA demeth-
van der Valk, M., Deschamps, J., Sofroniew, M., ylation:Many roads lead to Rome. Nat Rev Mol
van Lohuizen, M., etal. (1994). Posterior trans- Cell Biol 11, 607620.
formation, neurological abnormalities, and Wu, X., Gong, Y., Yue, J., Qiang, B., Yuan, J., and
severe hematopoietic defects in mice with a Peng, X. (2008). Cooperation between EZH2,
targeted deletion of the bmi-1 proto-oncogene. NSPc1-mediated histone H2A ubiquitination
Genes Dev 8, 757769. and Dnmt1 in HOX gene silencing. Nucleic
Varley, K.E., Gertz, J., Bowling, K.M., Parker, Acids Res 36, 35903599.
S.L., Reddy, T.E., Pauli-Behn, F., Cross, M.K., Yan, H., Parsons, D.W., Jin, G., McLendon, R., Rasheed,
Williams, B.A., Stamatoyannopoulos, J.A., B.A., Yuan, W., Kos, I., Batinic-Haberle, I., Jones,
Crawford, G.E., et al. (2013). Dynamic DNA S., Riggins, G.J., et al. (2009). IDH1 and IDH2
methylation across diverse human cell lines and mutations in gliomas. N Engl J Med 360, 765773.
tissues. Genome Res 23, 555567. Yoo, A.S., Sun, A.X., Li, L., Shcheglovitov, A.,
Vasudevan, S., Tong, Y., and Steitz, J.A. (2007). Portmann, T., Li, Y., Lee-Messer, C., Dolmetsch,
Switching from repression to activa- R.E., Tsien, R.W., and Crabtree, G.R. (2011).
tion: MicroRNAs can up-regulate translation. MicroRNA-mediated conversion of human
Science 318, 19311934. fibroblasts to neurons. Nature 476, 228231.
Vire, E., Brenner, C., Deplus, R., Blanchon, L., Zhang, Y. (2003). Transcriptional regulation by his-
Fraga, M., Didelot, C., Morey, L., Van Eynde, A., tone ubiquitination and deubiquitination. Genes
Bernard, D., Vanderwinden, J.M., et al. (2006). Dev 17, 27332740.
The Polycomb group protein EZH2 directly con- Zhou, H., Hu, H., and Lai, M. (2010). Non-coding
trols DNA methylation. Nature 439, 871874. RNAs and their epigenetic regulatory mecha-
Wang, H., Wang, L., Erdjument-Bromage, H., Vidal, nisms. Biol Cell 102, 645655.
M., Tempst, P., Jones, R.S., and Zhang, Y. (2004). Zhu, Q., Wang, L., Zhang, Y., Zhao, F.H., Luo, J.,
Role of histone H2A ubiquitination in Polycomb Xiao, Z., Chen, G.J., and Wang, X.F. (2011).
silencing. Nature 431, 873878. Increased expression of DNA methyltransferase
Wang, T., Pan, Q., Lin, L., Szulwach, K.E., Song, C.X., 1 and 3a in human temporal lobe epilepsy. J Mol
He, C., Wu, H., Warren, S.T., Jin, P., Duan, R., Neurosci 46, 420426.
etal. (2012). Genome-wide DNA hydroxymeth- Ziller, M.J., Muller, F., Liao, J., Zhang, Y., Gu, H.,
ylation changes are associated with neurode- Bock, C., Boyle, P., Epstein, C.B., Bernstein, B.E.,
velopmental genes in the developing human Lengauer, T., etal. (2011). Genomic distribution
cerebellum. Hum Mol Genet 21, 55005510. and inter-sample variation of non-CpG meth-
Wang, Z., Zang, C., Rosenfeld, J.A., Schones, D.E., ylation across human cell types. PLoS Genet 7,
Barski, A., Cuddapah, S., Cui, K., Roh, T.-Y., Peng, e1002389.
13
Adult Neural Stem Cells
and Brain Homeostasis
ASHOK K.SHET T Y

INTRODUCTION the persistence of NSCs and the occurrence of


One of the foremost dogmas in early neurosci- neurogenesis all through life in two regions of
ence research was that neurons in the mamma- the adult mammalian brain: the subventricu-
lian brain are generated in their entirety during lar zone (SVZ) lining the lateral ventricles of
the early developmental periods and hence are the forebrain and the subgranular zone (SGZ)
not reinstated when they die due to either neuro- of the DG in the hippocampal formation (Ihrie
degenerative diseases or brain injury. Although and Alvarez-Buylla, 2011; Ming and Song, 2011;
the concept of adult neurogenesis (i.e., the forma- Yao et al., 2012; Figure 13.1). Typically, prolif-
tion of new neurons in the adult brain) emerged eration of NSCs in the SVZ produces a large
in the 1960s through a finding that new cells are pool of immature neurons (neuroblasts), which
generated in the olfactory system and dentate migrate into the olfactory bulb through a path-
gyrus (DG) of the adult brain by Altman and Das way called the rostral migratory stream (Ihrie
(1965), lack of definitive markers to detect neu- and Alvarez-Buylla, 2011, Figure 13.1). Once
rons among tritiated-thymidine labeled newly they reach the olfactory bulb, they differentiate
born cells made this discovery go mostly unno- into different types of olfactory interneurons.
ticed for nearly two decades. Findings in the Proliferation of NSCs in the SGZ, on the other
1980s that neurogenesis occurs in 3-month-old hand, results in production of new neurons and
rat visual cortex (Kaplan, 1981)and adult canar- glia. Newly born neurons then migrate into the
ies learn new songs through the addition of new granule cell layer (GCL) of the DG, where they
neurons (Goldman and Notteboum, 1983) reig- mature, emanate dendrites into the molecular
nited the perception of neurogenesis in the adult layer to establish connectivity with axons com-
brain but did not attract many researchers to ing from the entorhinal cortex, and send axons
work in this area. However, a seminal finding into the stratum lucidum of the CA3 to make
in the early 1990s that both neurons and astro- synaptic contacts with primary dendrites of CA3
cytes can be generated in culture from cells iso- pyramidal neurons (Ming and Song, 2011; Yao
lated from the adult mammalian brain (Reynolds etal., 2012, Figure13.1).
and Weiss, 1992)triggered a massive interest in Furthermore, many studies support the idea
examining neural stem/progenitor cells (NSCs) that some amount of neurogenesis also occurs
and neurogenesis in the adultbrain. in the so-called nonneurogenic regions of the
Cells that produce new neurons in different brain (Gould, 2007). This is initially revealed
regions of the adult brain are broadly classified through in vitro observations that proliferat-
as NSCs (Ihrie and Alvarez-Buylla, 2011; Ming ing multipotent NSC-like cells can be isolated
and Song, 2011; Yao et al., 2012). Neural stem from many nonneurogenic regions of the adult
cells have the ability of extensive proliferation, brain, which include the cerebral cortex, stria-
self-renewal, and production of all three major tum, spinal cord, retina, ventral mesencephalon,
phenotypes (neurons, astrocytes, oligodendro- and substantia nigra (Palmer et al., 1999; Lie
cytes) in the central nervous system. Progenitor et al., 2002; Bonfanti and Peretto, 2011). Many
cells, on the other hand, seem to have limited in vivo studies also suggest limited neurogenesis
proliferation potential and lack self-renewal in several regions of the adult brain such as the
ability. Multiple studies have now confirmed hippocampus CA1 subfield (Rietze etal., 2000),
Adult Neural Stem Cells and Brain Homeostasis 203

A
OB
CBM
CTX

DG
SVZ
RMS
C GFAP
B &
Sox-2

ML
D GFAP & Sox-2

E Sox-2 & Ki-67


Mature
Granule Cell
GCL NeuN, Prox1
SGZ

F DCX & Ki-67


Type 3 Cells
(Neuroblasts)
Type 2 Cells
DCX
(TAPs)
sox-2, Mash1
Type 1 Cells (NSCs)
GFAP, Sox-2, Nestin

FIGURE13.1: Neurogenic regions and steps of neurogenesis. Figure Adepicts the two active neurogenic
regions in the adult brain, the dentate gyrus (DG) of the hippocampus and the subventricular zone (SVZ) lining
the lateral ventricle (red zones). RMS=rostral migratory stream; OB=olfactory bulb; CTX=cortex; CBM=cer-
ebellum. Figure B illustrates the various steps involved in DG neurogenesis within the hippocampus. Type 1 cells
(both radial and horizontal cells shown in green) express markers such as the glial fibrillary acidic protein, Sox-2,
and nestin. These cells occasionally divide to self-renew and to produce type 2 cells. Type 2 cells (or transit ampli-
fying cells [TAPs], shown in blue]) express Sox-2 and mammalian achaete-scute homolog 1 (Mash1) and divide
more actively to produce type 3 cells (neuroblasts). Type 3 cells express doublecortin (DCX) and exhibit limited
proliferation before maturing into dentate granule cells expressing markers such as NeuN and Prox-1. Figures C
and D show examples of radial (C)and horizontal (D)type 1 cells expressing glial fibrillary acidic protein (GFAP)
and Sox-2 in the subgranular zone of the DG. Figures E and F show proliferation of type 2 cells (i.e., cells express-
ing Sox-2 and Ki-67 in E) and type 3 cells (i.e., cells expressing DCX and Ki- in F) in the subgranular zone of the
DG. GCL=granule cell layer; ML=molecular layer; SGZ=subgranularzone.

substantia nigra of the mesencephalon (Lie etal., existence of multipotent NSC-like cells in these
2002; Zhao et al., 2003), neocortex (Cameron adult brain regions displaying the ability for pro-
and Dayer, 2008), striatum (Bedard etal., 2006), liferation in the presence of apt neurotrophic
amygdala (Fowler et al., 2008), piriform cortex factors is promising. This is because, with the
(Shapiro et al., 2007), and hypothalamus (Yuan advent of new neurogenic compounds and drugs
and Arias-Carrin et al., 2011). While there is (e.g., aminopropyl carbazole; Pieper etal., 2010;
still some skepticism pertaining to the functional Walker etal., 2014), these cells may be amenable
integration of new neurons born in nonneuro- for extensive proliferation to produce large num-
genic regions in normal conditions (Koketsu bers of site-specific new neurons in neurodegen-
et al., 2003; Bonfanti and Peretto, 2011), the erative disease conditions.
204 Part II:Homeostatic Control

Thus, both adult NSC and neurogenesis stud- vast majority of proliferating cells in the SGZ
ies in multiple species during the past two decades of the hippocampus differentiate into neurons
have clearly refuted the longstanding doctrine (Cameron etal., 1993; Rao etal., 2005; Rao etal,
that adult mammalian brain does not generate 2006), though astrocytes, oligodendrocytes, and
new neurons. Furthermore, Eriksson and col- endothelial cells also emerge from these prolif-
leagues (1998) offered the first proof of neurogen- erating cells (Kornack and Rakic, 1999). It is
esis occurring in the hippocampus of the adult widely believed that a subset of slowly dividing
human brain. Since then, there has been immense cells expressing the glial fibrillary acidic pro-
attention paid to ascertaining the behavior of tein (GFAP) and exhibiting a single radial pro-
NSCs and the pattern and extent of neurogenesis cess that extends through the GCL are NSCs or
in different regions of the adult brain in normal radial glial cells (type 1 cells) in this region (Seri
and disease conditions. Nonetheless, because etal., 2001; Kempermann etal., 2004; Ihrie and
neurogenesis was observed at lower levels in Alvarez-Buylla, 2008, Figure 13.1). These cells
nonhuman primates than rodents, skepticism also express other markers of NSCs such as the
prevailed regarding the extent of neurogenesis in transcription factor sex determining region Y
the adult human brain such as the hippocampus [SRY]-box 2 (Sox-2; Hattiangady and Shetty,
and whether a small number of newly generated 2008a), the primitive neurofilament protein nes-
neurons can impact human behavior (Kheirbek tin (Filippov etal., 2003; Lagace etal., 2007), the
and Hen, 2013). A recent study, by providing radial glial marker vimentin (Hattiangady and
strong evidence for widespread neurogenesis Shetty, 2008a), and the RNA binding protein
in the DG of humans throughout adulthood, musashi-1 (Okano etal., 2005). However, another
has elegantly answered the query regarding the class of cells having short, horizontal processes
amount of neurogenesis that occurs in the adult has also been suggested as type 1 cells in the SGZ
human hippocampus (Spalding etal., 2013). An (Lugert etal., 2010, Figure13.1). Both radial and
exciting finding of this study is that neurogen- nonradial type 1 cells seem to shuttle between
esis does not considerably decline with aging active and quiescent states (Rolando and Taylor,
in the human hippocampus, in contrast to a 2014). The quiescent nature of NSCs between
dramatic age-related decline in neurogenesis their proliferative states is widely believed to not
observed in rodents (Kuhn etal., 1996; Rao etal., only prevent their exhaustion in normal con-
2005; Rao et al., 2006; Hattiangady and Shetty, ditions but also to reduce chromosomal aber-
2008a). Although the functional significance of rations resulting from frequent cell division
adult-born neurons in the human hippocampus (Rolando and Taylor, 2014). In normal condi-
remains to be elucidated, studies in animal mod- tions, slow or infrequent proliferation of these
els have provided substantial insights regard- NSCs produces a pool of transit amplifying cells
ing their role in cognitive and mood function (TAPs, or type 2 cells; Figure 13.1). However, the
(Kheirbek and Hen, 2013). Therefore, this chap- response of radial and nonradial type 1 NSCs in
ter is focused on the present knowledge concern- the adult DG to pathophysiological signals seems
ing the activity of NSCs and neurogenesis in the to be dependent on the type of cues. For instance,
adult mammalian hippocampus under standard a study has shown that physical exercise induces
and certain disease environments and on how proliferation of radial NSCs whereas seizures
aberrant or declined NSC activity and neurogen- activate only nonradial NSCs (Lugert etal., 2010).
esis in neurological diseases can weaken brain Type 2 cells display smaller size, show short tan-
homeostasis principally with reference to cogni- gential processes, and typically occur in clusters
tive and mood function. The way increased hip- (Hsieh, 2012, Figure13.1). These cells expressing
pocampal neurogenesis in depressive disorders Sox-2 and mammalian achaete-scute homolog 1
or improved forebrain neurogenesis after stroke proliferate rapidly and give rise to doublecortin
may aid in functional recovery is also discussed. expressing neuroblasts (type 3 cells; Figure 13.1)
and glia (Encinas etal., 2011). Neuroblasts later
NSC NICHE INTHE evolve into full-fledged granule cells express-
HIPPOCAMPUS ing the mature neuronal marker neuron specific
Neurogenesis in the DG of hippocampus occurs nuclear antigen (NeuN) and the granule cell spe-
in a wide variety of species (Kuhn et al., 1996; cific transcription factor prospero-related home-
Gould et al., 1997, 1999; Kornack and Rakic, obox gene (Prox-1; Figure 13.1). These mature
1999; Eriksson et al., 1998). Interestingly, a granule cells also establish afferent connectivity
Adult Neural Stem Cells and Brain Homeostasis 205

with the perforant path fibers coming from the Taylor, 2014). The role of several other transcrip-
entorhinal cortex and efferent connectivity with tion factors in the maintenance of NSCs have
the CA3 subfield pyramidal neurons, which takes also been noted, which include paired domain
about 6 to 8 weeks in the rodent hippocampus and homeodomain-containing transcription fac-
(Zhao etal., 2006; Hsieh,2012). tor, Ascl1 (another bHLH transcription factor),
Activity of NSCs is determined by multiple forkhead domain transcription factor, repres-
signals arising from the milieu. First, a variety sor element1 silencing transcription factor, and
of cellular components existing in and around nuclear hormone transcription factor (Jessberger
NSC niches such as astrocytes, oligodendrocyte etal., 2008; Renault etal., 2009; Gao etal., 2011;
progenitors, endothelial cells, pericytes, micro- Hsieh,2012).
glia, mature granule cells, and GABA-ergic The formation of type 2 cells (TAPs) and
interneurons can influence NSC activity type 3 cells (neuroblasts) from types 1 and 2 cells
(Palmer etal., 2000; Goldman and Chen, 2011; involves a sequential upregulation of transcrip-
Gemma and Bachstetter, 2013). Second, noncel- tion factors Neurogenin 2 (a bHLH transcription
lular components such as secreted molecules factor involved in neural fate-choice decision)
and the extracellular matrix proteins play roles and Tbr2 (T-brain gene 2, involved in conversion
(Palmer etal., 2000; Morrens etal., 2012; Hsieh, of radial glia into intermediate progenitors during
2012). Third, studies have shown that both neu- cortex development). Differentiation of neuro-
rons and astrocytes in NSC niches can influence blasts into mature neurons involves the expres-
self-renewal or differentiation of NSCs (Song sion of neuronal differentiation 1 (Gao et al.,
et al., 2002; Tozuka et al., 2005). For example, 2009) regulated through Wnt/-catenin signal-
astrocytes have been shown to be a source of ing (Kuwabara etal., 2009; Seib etal., 2013)and
several important paracrine factors having vital expression of Sox-3, Sox-11, and Forkhead box
roles in neurogenesis, which include Notch, G1, a winged helix transcriptional repressor
Sonic hedgehog, bone morphogenetic proteins, (Shen et al., 2006; Wang et al., 2006; Haslinger
and Wnt proteins (Ahn and Joyner, 2005; Lie et al., 2009). Inhibition of Wnt activity results
etal., 2005; Ables etal., 2010; Hsieh, 2012). Thus in reduced neurogenesis whereas its activation
adult neurogenesis is orchestrated through a through Wnt3a protein expression enhances
multicellular niche, where NSCs respond to neurogenesis (Lie etal., 2005). Furthermore, dele-
instructive signals from other cell types, result- tion of a negative regulator of the Wnt/-catenin
ing in their proliferation, self-renewal, and/ pathway such as Dickkopf1 enhances neurogen-
or differentiation of their progeny into mature esis (Seib etal., 2013). The survival and matura-
neurons orglia. tion of newly generated neurons is influenced
by Prox-1 (Jessberger etal., 2008; Karalay etal.,
R E G U L AT O R S O F N S C 2011)and cyclic AMP response element-binding
ACTIVIT Y AND NEUROGENESIS protein (CREB; Jagasia etal.,2009).
I N T H E A D U LT H I P P O C A M P U S Additionally, several neurotransmitters can
Maintenance of NSCs and cell fate specification influence the extent of adult hippocampal neu-
of NSC-derived cells in the SGZ are mediated rogenesis. These include the inhibitory neuro-
through several transcription factors and sign- transmitter gamma-amino butyric acid (GABA),
aling pathways. Multiple studies have shown serotonin (5-hyroxytryptamine [5-HT]), and
that preservation of type 1 cells with expres- acetylcholine. The role of GABA has received
sion of transcription factors Sox-2 and hairy and the most attention because the GABA released
enhancer of split 5 (which encodes a member of by a subpopulation of interneurons expressing
a family of basic helixloophelix [bHLH] tran- the calcium binding protein parvalbumin (PV)
scription repressors) occurs through Notch sign- in the DG has been found to regulate both stem
aling (Ables et al., 2010; Imayoshi et al., 2010; cell quiescence and neuronal cell fate decision
Lugert etal., 2010; Hsieh, 2012). Both radial and by newly born cells. Specifically, PV+ interneu-
nonradial type 1 cells have been shown to exhibit rons repress the activation of quiescent NSCs by
canonical Notch activity (Lugert et al., 2010; releasing nonsynaptic GABA under normal con-
Lugert et al., 2012). It has been suggested that ditions (Song etal., 2012). Such regulation medi-
Notch signaling blocks neuron generation from ated through 2-containing GABAA Rs expressed
NSCs through suppression of the expression of on NSCs is thought to be important, as it averts
proneural transcription factors (Rolando and the exhaustion of primary NSCs through rapid
206 Part II:Homeostatic Control

proliferation (Song et al., 2013). On the other First, positive environmental conditions (physi-
hand, specific activation of PV+ interneurons pro- cal exercise or exposure to enriched environ-
motes the survival of proliferating DCX+ imma- ments) that enhanced hippocampus-dependent
ture neurons through establishment of immature memory function also increased DG neurogen-
synapses between axons of PV+ interneurons esis (Kempermann etal., 1997; Van Praag etal.,
and newly born neurons. Pertaining to 5-HT, 1999). Second, aged rats with unimpaired spatial
multiple studies using pharmacological enhance- memory retrieval ability displayed greater levels
ment of 5-HT levels, reduction of serotonergic of neurogenesis than aged rats exhibiting mem-
neurons, or stimulation of 5-HT receptors have ory impairment (Drapeau et al., 2003). Third,
shown that 5-HT can enhance the production of impaired hippocampus-dependent memory
new neurons in the DG (Doze and Perez, 2012). function in animals that underwent stress was
Likewise, acetylcholine levels also seem to influ- associated with greatly reduced DG neurogenesis
ence neurogenesis because ablation of cholinergic (Gould et al., 1999). Fourth, neurogenesis abla-
neurons in the medial septum decreased neuro- tion studies performed using approaches such
genesis and administration of cholinergic ago- as whole brain radiation or administration of
nists enhanced neurogenesis in the hippocampus drugs that target dividing NSCs and their prog-
(Mohapel etal.,2005). eny also supported a role for DG neurogenesis
in certain forms of hippocampus-dependent
FUNCTIONAL SIGNIFICANCE memory (Shors etal., 2001; Snyder etal., 2005).
OFNSC ACTIVIT Y AND Nevertheless, conflicting results in other studies
NEUROGENESIS INTHE and questions regarding the efficiency of tech-
A D U LT B R A I N niques used to selectively ablate DG neurogen-
Newly generated neurons (granule cells) in the esis in its entirety without altering the existing
DG integrate into the existing hippocampal cir- hippocampal cytoarchitecture or circuitry raised
cuitry and become functional (van Praag et al., some doubts on the purported role of DG neu-
2002). They display special electrophysiological rogenesis in spatial learning and memory func-
features such as an increased amenability for tion for some time (Deng etal., 2010; Braun and
long-term potentiation (a substrate underlying Jessberger,2014).
learning and memory) for about a month after Follow-up studies, however, provided
their birth, after which they exhibit similar prop- stronger evidence that spatial learning astutely
erties as their older counterparts (Schmidt-Heber inserts or eliminates newly born granule cells
et al., 2004; Wang et al., 2000; Ming and Song, depending on their stage of the development
2011). Thus, continuous production of new den- and the functional importance, and that popula-
tate granule cells would serve to maintain a pool tions of new granule cells rescued are likely those
of neurons in the DG with special properties. that are successfully integrated into the learning
Studies suggest that such continuous turnover of and memory circuitry (Tronel etal., 2010, Deng
DG granule cells is highly efficient for meeting et al., 2010). Furthermore, a memory-retrieval
some of the unique computational needs of the task after the water maze training preferentially
hippocampus (Appleby etal., 2011). It is believed activated new granule cells that were ~4 to 6
that the newly born granule cells in the adult DG weeks old at the time of learning the task and ~10
not only promote plasticity but also provide sta- weeks old at the time of memory-retrieval test in
bility through their integration into the circuitry mice (Kee etal., 2007). Moreover, computational
for longer periods (Kempermann,2013). models suggested a role for newly born granule
At the behavioral level, adult DG neuro- cells in the encoding of temporal information as
genesis has been suggested to be important for well as in the maintenance of old memories dur-
hippocampus-dependent learning and memory ing the encoding of new information (Aimone
(Braun and Jessberger, 2014). Initially, a mul- etal., 2010; Deng etal., 2010; Aimone etal., 2011).
titude of studies correlated the extent of DG Studies utilizing sophisticated genetic ablation
neurogenesis with spatial learning and memory techniques also showed spatial learning defi-
performance using tasks such as water maze test. cits with the loss of hippocampal neurogenesis
Many of these investigations have suggested a link (Dupret etal., 2008; Zhang etal., 2008; Jessberger
between spatial memory function and the num- etal., 2009). Another study, by utilizing a combi-
ber of newly generated neurons, which has been nation of retroviral and optogenetic approaches
substantiated through multiple observations. to birth date and reversibly controlling a group
Adult Neural Stem Cells and Brain Homeostasis 207

of adult-born neurons in adult mice, showed that to die over a period of time (Gould etal., 1999;
silencing a cohort of ~4-week-old newly born Dayer et al., 2003; Gould et al., 2007). Thus,
granule cells after water-maze learning substan- despite continuous neurogenesis, the total num-
tially disrupts retrieval of spatial memory (Gu ber of granule cells in the DG remains nearly
etal.,2012). static throughout life under normal conditions.
Moreover, several recent studies through While newly born granule cells have been shown
selective testing of DG circuitry point out that to replace some granule cells that were born
newly generated neurons are critical for pattern during early development, they mostly seem
separation function (Clelland etal., 2009; Sahay to replace granule cells that were born during
et al., 2011a,b; Kheirbek et al., 2012; Nakashiba adulthood (Dayer etal., 2003). While the precise
etal., 2012). Pattern separation refers to the abil- lifespan of adult-born granule cells is unknown,
ity to discriminate similar experiences through it is believed to be dependent on their ability to
the storage of similar representations in a nono- become integrated into the hippocampus cir-
verlapping manner (Leutgeb et al., 2007; Yassa cuitry, as learning and complex experience can
and Stark, 2011). The precise mechanisms by promote the survival of newly born granule cells
which newly born neurons promote pattern (Kempermann, 2012). Moreover, in their matu-
separation function are still being investigated. ration period, newly born granule cells display
Computational, anatomical, and electrophysio- enhanced synaptic plasticity, which is evidenced
logical studies imply that sparseness of activation through a lower threshold for eliciting long-term
in the granule cell layer of DG (resulting in sparse potentiation in these cells (Schmidt-Hieber
functional connectivity between DG and CA3) is et al., 2004; Snyder et al., 2001). Furthermore,
conducive for pattern separation by enhancing newly born granule cells form a major compo-
global remapping, a process by which similar rep- nent of granule cells that contribute to long-term
resentations are encoded by nonoverlapping neu- potentiation in the DG because the preexisting
ronal ensembles (OReilly and McClelland, 1994). network is inhibited from being activated (Saxe
Consistent with this idea, a recent study suggests etal., 2006; Garthe etal., 2009). Taken together,
that modulation of DG excitability by adult-born it emerges that newly born granule cells are more
neurons (i.e., new granule cells) enhances sparse plastic than older granule cells and are more
coding in the granule cell layer to influence pat- likely to generate action potentials in response to
tern separation (Ikrar et al., 2013). Addition of an incoming stimulus. Therefore, young granule
increased numbers of newly born granule cells cells respond to a broad variety of synaptic input
into the DG altered the excitability of mature or in comparison to old (preexisting) granule cells
existing dentate granule cells, which appeared to that seem to respond to a more specific input.
be mediated through local circuit mechanisms This differential response between young and
comprising increased excitatory drive from old granule cells has been suggested to be benefi-
newly generated granule cells onto GABA-ergic cial because young granule cells being tuned to a
interneurons in the dentatehilus. broad variety of inputs can be good integrators,
Nonetheless, one of the issues being dis- whereas older cells displaying high input speci-
cussed in the field is whether the extent of neu- ficity can be better separators (Marn-Burgin
rogenesis occurring in adulthood is adequate for et al., 2012; Kempermann, 2012). These attrib-
influencing hippocampus function. The amount utes are considered to be critical for the DG
of neurogenesis occurring after the initial devel- to perform its functional roles, which include
opment of the DG is not trivial, because ~9,000 facilitating pattern separation and preventing
new neurons (granule cells) are generated every catastrophic interference, providing contextual
day in the DG of a young adult rat, and the num- and affective annotations, and contributing to
ber of new granule cells generated each month cognitive flexibility in situations where novel
amounts to roughly 6% of the total size of the information has to be integrated into previously
granule cell population (Cameron and McKay, formed representations (Marn-Burgin et al.,
2001). In the human brain, that figure is ~700 2012; Kempermann, 2012). Therefore, continu-
new neurons per day for an annual turnover ous production of new granule cells seem to be
rate of 1.75% (Kempermann, 2013). However, necessary for maintaining normal hippocampus
the volume of DG or the hippocampus does not function, as it provides a pool of highly plastic
increase as a function of age because a substan- immature cells to facilitate complex computa-
tial fraction of newly generated neurons appear tional needs (Kempermann,2012).
208 Part II:Homeostatic Control

Reduced DG neurogenesis has also been during the above-ground nuclear bomb tests
implicated in psychiatric disorders (Bergmann between 1945 and 1963 (Kempermann, 2013).
and Frisen, 2013). It has been suggested that This approach relies on the principle that 14C in
fewer newly born neurons impairing pattern the atmosphere becomes integrated into the DNA
separation function may impair the ability to of dividing human cells because of eating plants
distinguish threats from similar, but safe, situ- that have absorbed 14C from the atmosphere or
ations and contribute to a generalized percep- eating animals that have consumed plants absorb-
tion of fear and anxiety in posttraumatic stress ing atmospheric 14C (Welberg, 2013). Because 14C
disorder (Kheirbek et al., 2012; Bergmann and is incorporated in the DNA during cell division,
Frisen, 2013). Furthermore, most antidepres- the 14C content of a cell is thought to reflect 14C
sant treatments enhance DG neurogenesis, and levels in the atmosphere at the time of the birth
some of the effects of antidepressants in animal of the cell. As atomic bomb testing in the 1950s
models are dependent on increased neurogenesis and 1960s caused a spike in atmospheric 14C lev-
(Santarelli etal., 2003). Thus DG neurogenesis in els and levels declined after 1963 (because of the
the adult hippocampus is important for main- limited test ban treaty), the level of 14C in cellu-
taining homeostasis in the hippocampus, partic- lar DNA facilitated determination of a cells birth
ularly its ability for promoting normal memory date (Welberg, 2013). Spalding and colleagues,
and mood function. using hippocampus from postmortem brains of
individuals who were born in different years in
N E U R O G E N E S I S I N T H E A D U LT the 20th century, sorted neurons from glial cells,
HUMAN HIPPOCAMPUS purified neuronal DNA, and measured 14C levels.
Coherent with the results in other mammalian As the amount of incorporated 14C correlates with
species, the DG of the human hippocampus has the atmospheric 14C at the time of cell division,
been validated as a neurogenic region. Eriksson this method allowed determination of the age of
and colleagues (1998) specifically examined cells. This study provided three major novel find-
putative NSCs and newly born neurons within ings. First, adult neurogenesis in the human brain
the DG of the adult human hippocampus using is restricted to the hippocampus. Second, based
postmortem hippocampal tissues from five can- on computer modeling, it is estimated that nearly
cer patients receiving one intravenous infusion 80% of DG granule cells undergo renewal in adult-
of 5-bromodeoxyuridine (BrdU) for diagnostic hood and ~700 new neurons are added per day for
purposes (Eriksson et al., 1998). Examination an annual turnover rate of 1.75% (Kempermann,
of BrdU+ cells in the DG using BrdU-NeuN and 2013). Third, the turnover rate was quite similar
BrdU-GFAP dual immunofluorescence and con- between men and women and decreased only
focal microscopy revealed ~22% neurons and modestly with aging, unlike in rodents where a
~18% astrocytes among newly born cells. The dramatic age-related decline in neurogenesis is
remaining BrdU+ cells were smaller in size and observed (Kuhn etal., 1996; Rao etal., 2005; Rao
appeared to be quiescent undifferentiated NSCs. etal., 2006; Hattiangady and Shetty, 2008a). Thus
Cells that double-labeled for BrdU and calbindin the occurrence of NSCs and neurogenesis in the
were also observed, which provided further sup- adult human hippocampus has now been proved
port for the occurrence of newly born granule beyonddoubt.
cells in the adult human DG. While this study
provided the first evidence for the occurrence NSC ACTIVIT Y AND
of NSCs and neurogenesis in the adult human NEUROGENESIS IN
hippocampus, skepticism prevailed in the field NEUROLOGICAL DISORDERS
because some studies have suggested that neuro-
genesis is less abundant in nonhuman primates Altered Homeostasis of NSCs
in comparison to rodents (Amrein et al., 2011; and Neurogenesis in Temporal
Kheirbek and Hen,2013). Lobe Epilepsy
However, a recent study provides strong evi- Epilepsy, a medical condition typified by sei-
dence for widespread neurogenesis in the DG of zures and interfering with a variety of mental
humans throughout adulthood (Spalding et al., functions, affects over 2million Americans and
2013). The authors employed a unique birth-dating ~65 million people worldwide. Temporal lobe
strategy based on the peak of carbon isotope epilepsy (TLE) is among the most common
14 (14C) that was released into the atmosphere types of drug-resistant epilepsy (Engel, 2001;
Adult Neural Stem Cells and Brain Homeostasis 209

Strine etal., 2005). While the etiology of TLE is vascular endothelial growth factor, and sonic
unidentified in most cases, an initial precipitat- hedgehog, all of which are known to rise after
ing injury such as status epilepticus (SE), head acute seizures and to positively influence NSC
trauma, brain infections, or childhood febrile activity in the hippocampus (Lowenstein et al.,
seizures leads to TLE in others (Pitkanen and 1993; Bugra etal., 1994; Shetty etal., 2003; Croll
Sutula, 2002; Lewis, 2005). Classically, a dor- et al., 2004; Shetty et al., 2004; Banerjee et al.,
mant period of several years is seen between an 2005). Furthermore, increased levels of GABA
initial precipitating injury and the appearance of and neuropeptide Y (NPY) in the DG during the
chronic TLE exemplified by spontaneous recur- early postseizure period likely play roles, as both
rent seizures (SRS) commencing from temporal GABA and NPY have been shown to promote
lobe foci, learning and memory difficulties, and DG neurogenesis (Scharfman and Gray, 2006; Ge
depression (Devinsky, 2004; Butler and Zeman, etal., 2007; Masiulis etal., 2011). Moreover, there
2008; Bell etal., 2011). In addition, TLE is often is some indirect evidence that repressor element
associated with hippocampal sclerosis owing 1-silencing transcription factor (a transcriptional
to a considerable loss of neurons in the dentate repressor that functions to limit transcription
hilus and CA1 and CA3 subfields. Over the past of target genes) plays a role in seizure-induced
decade, altered activity of NSCs and abnormal increased neurogenesis (Jessberger et al., 2007).
hippocampal neurogenesis has emerged as one Besides these, augmented neuronal activity
of the most conspicuous pathological altera- during and after seizures may be promoting an
tions in TLE (Parent etal., 1997; Scharfman and increased proliferation of NSCs, as increased
Gray, 2007; Kuruba and Shetty, 2009; Hester and excitatory stimuli can act directly on NSCs
Danzer,2014). and influence the production of new neurons
(Deisseroth etal.,2004).
Homeostasis ofNSCs and Abnormal migration of a significant frac-
Neurogenesis Is Distraught inthe tion of newly born granule cells into the hilus
Early Phase ofTLE via Increased and the molecular layer is one of the major
Proliferation ofNSCs and Aberrant features of increased DG neurogenesis in the
Connectivity ofNewly Born Neurons early phase of TLE (Figure 13.2; Bengzon etal.,
Studies in animal models highlight considerably 1997; Parent etal., 1997; Scharfman etal., 2000;
increased NSC proliferation and DG neurogen- Hattiangady et al., 2004; Scharfman and Gray,
esis in the early phase of TLE (for 23 weeks fol- 2007; Shetty and Hattiangady, 2007; Hester and
lowing acute seizures or SE; Bengzon etal., 1997; Danzer, 2014). Loss or reduced levels of reelin
Parent et al., 1997; Hattiangady et al., 2004). (a migration guidance cue released from a sub-
Interestingly, acute seizures do not alter the pro- class of GABA-ergic interneurons in the dentate
liferation of type 1 NSCs in the SGZ but instead hilus) after acute seizures is believed to be one
promote the proliferation of TAPs and immature of the causes of aberrant migration of newly
neurons (Jessberger et al., 2005). In agreement born dentate granule cells (Gong et al., 2007).
with these results, the hippocampus from young Additional analyses has revealed that these
TLE patients (<24years old) displayed increased ectopic newly born granule cells likely become
proliferation of NSC-like cells (Blumcke et al., seizure-inducing hub cells as they (i)incorpo-
2001), signifying that the onset of TLE in pedi- rate aberrantly into the CA3 network, promote
atric patients is likely also accompanied by the development of proepileptogenic circuitry,
increased DG neurogenesis. Even though the spe- undergo activation when epileptic rats are expe-
cific mechanisms triggering the seizure-induced riencing SRS, and react with a longer latency
surge in DG neurogenesis are still unclear, vari- to onset of evoked responses following per-
ous possibilities have been advocated. Multiple forant path stimulation (Scharfman etal., 2002;
neurotrophic factors, believed to be released from Scharfman etal., 2003a, 2003b); (ii) exhibit basal
dying neurons, deafferented neurons, and reac- dendrites and establish afferent connectivity
tive glia after seizures, are thought to increase the with mossy fiber terminals (axons of other gran-
proliferation of types 2 and 3 cells as well as pro- ule cells; Pierce etal., 2005; Shapiro etal., 2005;
mote the neuronal differentiation of their prog- Walter etal., 2007); (iii) sprout axons (mossy fib-
eny. The neurotrophic factors include the nerve ers) abnormally into the dentate inner molecu-
growth factor, brain-derived neurotrophic fac- lar layer (Hester and Danzer, 2014); and (iv)
tor (BDNF), fibroblast growth factor 2 (FGF-2), exhibit spontaneous bursts of action potentials
210 Part II:Homeostatic Control

Newly Born Neurons in the DG of a Naive Rat


A1 A2

SGZ
GCL
DH

GCL

SGZ

Newly Born Neurons in the DG of a Rat that


Underwent Status Epilepticus
B1 B2 Survival of Newly
GCL
Born Neurons in the
Epileptic Hippocampus
(~5 months after birth)
DH

D1 ML

GCL
GCL

SGZ SGZ
Aberrant Migration of Newly Born Neurons into the
DH in a Rat that Underwent Status Epilepticus DH
C1
GCL
D2
SGZ
GCL

SGZ
DH

DH

FIGURE 13.2: Changes in hippocampal neurogenesis following kainic acid induced status epilepticus. Newly
born neurons in the dentate gyrus (DG) of a nave adult rat (A1) and an adult rat that underwent status epilepticus
12days prior to euthanasia (B1) were visualized with immunostaining for doublecortin (a marker of newly born
neurons). A2 and B2 show magnified views of regions of dentate gyrus from A1 and B1, respectively. Note that, in
comparison to the dentate gyrus of a control rat (A1, A2), a rat that underwent status epilepticus (B1, B2) exhibits
considerably increased density of doublecortin+ new neurons and abnormal migration of newly born neurons
into the dentate hilus (indicated by arrowheads in B1). C1 is magnified view of a region from B1 showing aber-
rantly migrated newly born neurons in the dentate hilus. Figures D1 and D2 show that neurons born in the first
two weeks after status epilepticus survive for prolonged periods (~5months) and incorporate into both granule
cell layer (arrows in D1) and the dentate hilus (arrows in D2) of the epileptic hippocampus. DH=dentate hilus;
GCL=granule cell layer; ML=molecular layer; SGZ=subgranularzone.
Figure reproduced with modifications from Shetty, A.K., and Hattiangady, B.[2007], Prospects of stem cell therapy for temporal
lobe epilepsy. Stem Cells 25, 21042117

and contribute to SRS in chronically epilep- TLE also comprise similarly displaced granule
tic animals (Scharfman etal., 2000; Jung etal., cells (Houser etal.,1990).
2004; McCloskey etal., 2006). Interestingly, hip- Thus altered homeostasis of NSCs and DG
pocampal tissues obtained from patients with neurogenesis after acute seizures clearly facilitates
Adult Neural Stem Cells and Brain Homeostasis 211

aberrant circuitry development. The role of this exhibiting neuronal differentiation in the intact
aberrant circuitry in the development of chronic hippocampus (Hattiangady and Shetty, 2010).
TLE has been a subject of intense scrutiny in a vari- Furthermore, differentiation of newly born cells
ety of TLE models. While studies in some models of into S-100 + mature astrocytes or NG2+ oligo-
TLE have raised doubts (Raedt etal., 2007; Pekcec dendrocyte progenitors enhanced to ~79% in the
et al., 2007; Pekcec et al., 2011), findings in other chronically epileptic hippocampus from ~25%
models of TLE are supportive of the involvement of seen in the intact hippocampus. Thus a dramatic
this circuitry as one of the pathophysiology under- decline in the neuronal fate-choice decision of
lying SRS. These include findings that reduction of newly born cells underlies decreased neurogen-
seizure-induced neurogenesis using an antimitotic esis in chronic epilepsy. Moreover, chronic TLE
agent can decrease the frequency and duration maintains abnormal features of neurogenesis
of SRS (Jung etal., 2004)and animals with large seen in the early phase, which include an aber-
numbers of abnormal cells display greater numbers rant migration of some newly born neurons
of seizures than animals with only a few abnor- into the hilus and occurrences of basal den-
mal cells (McCloskey etal., 2006). Additionally, a drites from newly born neurons that incorporate
recent study demonstrates that seizure frequency into the GCL (Hattiangady and Shetty, 2008b).
in chronic TLE is determined by multiple factors, The DG from children exhibiting frequent SRS
which include the severity of mossy fiber sprout- and adult TLE patients also showed decreased
ing and frequencies of hilar ectopic granule cells density of newly born neurons positive for the
and basal dendrites (Hester and Danzer, 2013), all polysialylated neuronal cell adhesion molecule
of which result from altered homeostasis of neuro- (PSA-NCAM; Mathern etal., 2002; Pirttila etal.,
genesis after seizures. Suppression of this aberrant 2005; Crespel et al., 2005; Fahrner et al., 2007).
circuitry may also minimize comorbidities of TLE The potential reasons underlying the waned neu-
such as cognitive and mood dysfunction because rogenesis in chronic epilepsy include an altered
animal studies have shown that blockage of abnor- milieu of NSC niches in the SGZ, including
mal DG neurogenesis after acute seizures can decreased concentrations of neurotrophic factors
prevent seizure-induced cognitive impairments (such as BDNF, FGF-2, and IGF-1) important
(Jessberger etal., 2007; Pekcec etal., 2008). From for NSC activity and neuronal differentiation of
these perspectives, it appears that development their progeny (Shetty et al., 2003; Hattiangady
of clinically applicable therapeutic strategies that etal., 2004), depletion of multipotent NSCs that
are capable of stabilizing NSC and neurogenesis can generate neurons (Hattiangady and Shetty,
homeostasis after acute seizures will be beneficial 2008b), or modifications in signaling pathways
for preventing chronic epilepsy and behavioral causing reduced neuronal differentiation of the
abnormalities resulting from SE or hippocampus progeny of NSCs such as increased concentra-
injury. tion of Dickkopf-1 (an inhibitor of neurogenesis
promoting protein Wnt; Busceti etal., 2007; Lie
Homeostasis ofNeurogenesis Is etal.,2005).
Altered inthe Chronic Phase ofTLE Thus studies on hippocampi from both
Through Altered Potential ofNSCs animal models and TLE patients authenti-
and Decreased Generation ofNew cate that chronic epilepsy is associated with
GranuleCells greatly declined hippocampal neurogenesis.
Studies in animal models clearly demonstrate Considering the functions of hippocampal neu-
greatly weakened neurogenesis in the chronic rogenesis discussed earlier, it is likely that altered
phase of TLE (Figure 13.3; Hattiangady et al., homeostasis of hippocampal neurogenesis is one
2004, Hattiangady and Shetty, 2010). Moreover, of the major causes of memory dysfunction and
an inverse relationship was seen between the depression seen in TLE. These links are sup-
frequency of SRS and the extent of neurogen- ported by observations that (i)the overall granule
esis (Hattiangady et al., 2004). Interestingly, a cell density is a significant predictor accounting
study in the kainate model demonstrated that for the total memory capacity in an individual
chronic epilepsy did not affect the overall addi- TLE patient (Pauli etal., 2006; Siebzehnrubl and
tion or long-term survival of newly born cells Blumcke, 2008), and (ii) increased production
in the GCL. However, only 45% of newly born of new neurons in the hippocampus is essential
cells (i.e., BrdU+ cells) differentiated into neu- for effective action of antidepressants (Santarelli
rons, in comparison to 7380% of such cells et al., 2003; Sahay and Hen, 2007; Perera et al.,
DCX-Positive Newly Born Neurons in an Age-Matched Control Rat
A1 A2

SGZ GCL ML

DH

GCL

SGZ

DCX-Positive Newly Born Neurons at months after ICV KA administration


B1 B2 ML
GCL
SGZ

DH GCL

SGZ

DCX-Positive Newly Born Neurons at 5 months after IP KA-induced SE

C1 GCL C2
GCL
SGZ
SGZ

DH

DH

ML

D
p < 0.001
15000
Number per Dentate Gyrus

= Age-Matched Control Hippocampus


10000
= Hippocampus at 5-months post-ICV KA
= Hippocampus at 5-months post-SE
5000 p < 0.01

0
Doublecortin Positive Neurons

FIGURE 13.3: Status of dentate neurogenesis in chronic epilepsy as revealed by doublecortin (DCX) immu-
nostaining. A1 through C1 illustrate the distribution of DCX positive newly born neurons in the dentate gyrus
of an age-matched intact hippocampus (A1), a chronically injured hippocampus at 5months after an intracere-
broventricular (ICV) kainic acid (KA) administration (B1), and a hippocampus displaying robust chronic epilepsy
at 5months after the KA-induced status epilepticus (SE) (C1). Note that, in comparison to the age-matched intact
Adult Neural Stem Cells and Brain Homeostasis 213

2007). From these viewpoints, it appears that because selective serotonin reuptake inhibitors
strategies capable of normalizing neurogenesis such as fluoxetine enhance neurogenesis through
levels in chronic epilepsy (such as administra- activation of the 5HT1A receptors (Santarelli
tion of distinct neurotrophic factors, a physical etal., 2003), stress can increase cortisol levels and
exercise regimen, exposure to an enriched envi- both together can alter NSC activity and decrease
ronment, antidepressant therapy, or grafting of neurogenesis (Gould etal., 1997; Sapolsky, 2004),
fresh NSCs) would be efficacious for easing these and reduced levels of p-CREB and BDNF can
impairments (Shetty, 2011; Barkas et al., 2012). reduce neurogenesis (Lee etal., 2002; Nakagawa
Positive outcomes of normalizing DG neuro- etal.,2002).
genesis levels in chronic epilepsy will, however, While precise mechanisms by which neuro-
largely depend on whether or not newly born genesis influences mood are unknown, multiple
granule cells establish normal connectivity with studies in animal models provide support for
the surviving target CA3 pyramidal neurons. an association between mood function and hip-
pocampal neurogenesis. For example, factors
ROLE OF NSC AND that induce depressive-like behavior (e.g., mater-
N E U R O G E N E S I S H O M E O S TA S I S nal separation or social hierarchy related stress)
F O R T R E AT I N G D E P R E S S I V E also diminish neurogenesis (Mirescu etal., 2004;
DISORDERS Lajud etal., 2012). Moreover, animals exposed to
Depression affects nearly 15% of the population antidepressant activities (such as physical exer-
and a major depressive disorder (MDD) is asso- cise and exposure to enriched environments)
ciated with considerable morbidity and disabil- or treated with antidepressant drugs display
ity (Pandya etal., 2012). One of the hallmarks of increased levels of neurogenesis with improved
MDD is a reduction in the size of hippocampus, cognitive and mood function (van Praag et al.,
which may be related to altered homeostasis of 1999; Santarelli etal., 2003; Sahay and Hen, 2007;
NSCs and declined DG neurogenesis for pro- Lee etal.,2013).
longed periods (Masi and Brovedani, 2011; Lee Although doubts have been raised that an
etal., 2013). However, other factors such as neu- increased level of neurogenesis after antidepres-
ron loss, impairments of synaptic plasticity in sant treatments may be just an epiphenomenon,
the form of reduced axon branching, dendritic some clinical and animal model studies support
regression, and reduced numbers of synapses the notion that improved DG neurogenesis is a
may also contribute to the reduced size of the hip- prerequisite for the beneficial effects of antide-
pocampus (Sapolsky, 2001; Masi and Brovedani, pressants. First, beneficial effects from antide-
2011; Duman and Aghajanian, 2012). Altered pressants happen only after a chronic treatment
homeostasis of NSCs and neurogenesis in depres- regimen, which rules out an acute effect on sero-
sion likely emerges from multiple changes in the tonergic neurotransmission and instead points
hippocampus. This may include decreased sero- to alternative mechanisms such as the require-
tonergic neurotransmission, increased levels of ment for antidepressant-induced improvements
stress and cortisol, and reduced levels of p-CREB in neurogenesis, which would take several
and BDNF (DSa and Duman, 2002). This is weeks before drug-induced neurons become

FIGURE13.3: Continued
hippocampus, chronically epileptic hippocampi exhibit dramatically reduced density of DCX positive newly gen-
erated neurons. Arrowheads point to regions in the subgranular zone (SGZ) where neurogenesis is active. The
arrow in C1 denotes a neuron that has migrated into the dentate hilus. A2, B2, and C2 are magnified views of
regions from A1, B1, and C1 demonstrating the morphology of newly generated neurons in three groups. In the
dentate gyrus of the age-matched intact rat (A2), DCX positive newly born neurons exhibit long apical dendrites
that extend into the molecular layer (ML) through the granule cell layer (GCL). Contrastingly, in the hippocampi
from epileptic animals (B2, C2), a vast majority of DCX positive neurons display basal dendrites (indicated by
short arrows). Figure D shows that the extent of neurogenesis declines greatly in the epileptic hippocampus in
comparison to the age-matched intact hippocampus. DH=dentatehilus.
Figure reproduced from Hattiangady, B., and Shetty, A.K. (2008), Implications of decreased hippocampal neurogenesis in chronic
temporal lobe epilepsy. Epilepsia, 49 Suppl 5,2641
214 Part II:Homeostatic Control

functionally integrated (Braun and Jessberger, symptoms. First, the accumulation of a protein
2014). Second, the beneficial effect of antidepres- called amyloid-beta (A) in the extracellular
sant treatment correlates with the time of onset space leads to the formation of amyloid plaques.
of increased neurogenesis, as ablation of prolif- Amyloid-beta proteins are liberated from a larger
erating NSCs in the hippocampus blocked the integral membrane protein called the amyloid
antidepressant actions of drugs fluoxetine and precursor protein (APP) through sequential
imipramine (Santarelli etal., 2003). Third, norep- cleavage by - and -secretases (Mu and Gage,
inephrine and antidepressants that block its reup- 2011). Second, the accumulation of another pro-
take enhance neurogenesis through increased tein, tau, inside neurons leads to the formation of
proliferation of quiescent NSCs in the hippocam- neurofibrillary tangles (Reitz and Mayeux, 2014).
pus (Jhaveri etal., 2010). Fourth, an animal study As these changes progress, other alterations
has demonstrated that newly generated neurons become evident, which include impaired transfer
serve as a buffer for stress response through their of information at synapses, loss of synapses, and
effects on the hypothalamicpituitaryadrenal neurodegeneration (Selkoe,2001).
axis (Snyder etal., 2011). However, several other While pathological changes affect multiple
studies contradict the positive link between brain areas in AD, the hippocampus is con-
neurogenesis and antidepressants. For example, sidered one of the earliest brain regions to be
not all drugs with antidepressant effects require affected (Scheff and Price, 2006; Ohm, 2007; Mu
adequate neurogenesis to ease depression (David and Gage, 2011). It is generally believed that an
et al., 2009). Furthermore, an increased level of alteration in hippocampal neurogenesis may
neurogenesis obtained through genetic altera- underlie cognitive and mood dysfunction seen in
tions in normal animals enhances pattern separa- aging and the early stages of AD (Lazarov etal.,
tion function but has no effect on mood function 2010). Characterization of senile AD brains using
(Sahay etal., 2011a). Thus altered homeostasis of markers of newly born neurons such as DCX.
NSCs and neurogenesis is one of the features of PSA-NCAM surprisingly suggested increased
MDD, but it remains to be examined whether hippocampal neurogenesis in AD, and these
it is one of the causes or consequences of MDD. increases were correlated with the severity of the
Nonetheless, drug therapies that enhance NSC disease (Jin et al., 2004a). There was also some
activity and neurogenesis seem to ease cognitive indication of increased proliferative activity
and mood impairments seen in MDD (Sahay and of NSC-like cells in senile AD brains, based on
Hen,2007). increased levels of proliferating cell nuclear anti-
gen (PCNA) positive cells in the hippocampus
A LT E R E D N S C A C T I V I T Y A N D (Jin etal., 2004a). In contrast, a study examining
N E U R O G E N E S I S H O M E O S TA S I S younger (pre-senile) AD brains reported no alter-
INALZHEIMERS DISEASE ations in the DG neurogenesis, based on analyses
Alzheimers disease (AD), an age-related neuro- of cells expressing the proliferative marker Ki-67
degenerative disease typified by a progressive loss (Boekhoorn etal., 2006). Another study reported
of memory, affects ~5 million Americans and reduced markers of neurogenesis in association
~45million people worldwide. Individuals with with increased bone morphogenetic protein-6
AD display difficulties in learning, reasoning, levels in the hippocampus of AD patients (Crews
judgment, and communication; depression; and etal.,2010).
personality changes (Goedert and Spillantini, Studies in animal models of AD have
2006; Mu and Gage, 2011). Although the risk also provided divergent findings regarding
for developing AD doubles every five years after AD-induced changes in NSC activity and neuro-
age 65, younger people may also get AD. While genesis. Astudy using a transgenic mouse model
the precise causes of AD are still unknown, it is of AD with the Swedish and Indiana mutations
widely believed that AD occurs as a result of mul- under the platelet-derived growth factor pro-
tiple factors, which include genetics (e.g., pres- moter reported increased neurogenesis in the
ence of a risk gene apolipoprotein E 4 [APOE 4]) hippocampus (Jin etal., 2004b; Lopez-Toledano
and environmental and lifestyle factors such as and Shelanski, 2007), consistent with the obser-
heart disease, stroke, high blood pressure, dia- vations in senile AD brains discussed earlier
betes, and obesity (Reitz and Mayeux, 2014). (Jin et al., 2004a). However, studies in multiple
There are two alterations in the brain that are other models of AD mostly revealed decreases in
believed to contribute to the evolution of AD hippocampal neurogenesis. These comprise the
Adult Neural Stem Cells and Brain Homeostasis 215

presinilin1 (PS1) conditional knockout mouse applied at the early stage of disease. Animal stud-
(Feng et al., 2001), the mouse overexpressing ies performed so far are mostly supportive of
a mutant form of APP (the Swedish mutation this possibility. Multiple strategies that enhance
APP695; Haughey et al., 2002), the mutant PS1 neurogenesis have been shown to improve cog-
knock-in mouse (Wang etal., 2004a), the mutant nitive performance in several transgenic mouse
P117L transgenic mouse (Wen et al., 2004), the models of AD. The strategies include physical
homozygous PDAPP mouse (Donovan et al., exercise or exposure to enriched environments
2006), the double knock-in mouse carrying (Costa et al., 2007; Nichol et al., 2007, 2009),
targeted mutations in both APP and PS-1 (Zhang inhibition of microglial activation (Biscaro
et al., 2007), the mouse overexpressing the etal., 2012), choline supplementation (Velazquez
Swedish variant of APP and the exon 9-deleted et al., 2013), neural precursor cell grafting (Ben
variant of human PS1 (Verret et al., 2007), the Menachem-Zidon, et al et al., 2014), curcumin
triple transgenic mouse 3xTg-AD that harbors treatment (Tiwari et al., 2014), and electroacu-
three mutant genes (APPswe, PS1M146V, and puncture (Li et al., 2014). Thus normalizing the
tauP301L; Rodriguez etal., 2008), and the Tg2576 homeostasis of NSC activity and neurogenesis in
transgenic mouse overexpressing the human APP the early stages of AD may considerably delay the
isoform harboring the Swedish double mutation severity of the disease and promote better quality
(Krezymon etal.,2013). of life to AD patients.
When taken together, most studies imply Furthermore, because impaired neurogen-
changes in the homeostasis of hippocampal NSCs esis likely precedes the deposition of A plaques
and DG neurogenesis in AD, although there is (Chuang, 2010) and normal aging is not associ-
disagreement regarding upregulation versus ated with significant decreases in neurogenesis
downregulation of neurogenesis in AD brains. in humans (Spalding et al., 2013), examining
Increased neurogenesis observed in some senile neurogenesis in patients with mild cognitive
AD brains could be a compensatory response impairment may aid in early diagnosis of AD.
to deafferentation and neuronal loss. This may Although some techniques that can indirectly
also be due to the effects of AD drug therapy, as measure neurogenesis in living human brains,
administration of AD drugs can enhance DG such as the measurement of blood flow in the DG
neurogenesis in mouse models (Jin etal., 2006a). or the detection of a spectroscopic biomarker by
Thus discrepancies in findings between studies magnetic resonance spectroscopy, are available
likely reflect differences in the stage of the disease (Pereira etal., 2007; Manganas etal., 2007), more
at the time of characterization, drug treatment advanced techniques need to be developed for
provided to patients, and methods employed for accurately quantifying neurogenesis in vivo in
analyzing proliferating cells. This necessitates healthy and disease conditions (Ho etal.,2013).
systematic investigation of larger cohorts of AD
brains at different stages of the disease. It will SIGNIFICANCE OF
be also necessary to consider other issues such NSC ACTIVIT Y AND
as the age of patient at the time of AD onset, the NEUROGENESIS FOR BR AIN
type and doses of drugs taken, and the duration R E PA I R A F T E R S T R O K E
of drug therapy in different AD patient popula- Stroke affects nearly 15 million people world-
tions. Decreased neurogenesis seen in several wide each year. Of these, 5 million die due to
mouse models of AD may reflect the advanced stroke-related complications. Among patients
stage of the disease at the time of analyses. This is who survive stroke, ~90% endure neurological
because in some mouse models of AD, cell prolif- deficits, requiring rehabilitation therapy and/or
eration was significantly enhanced at early stages prolonged clinical care (Jorgensen et al., 1997;
of neurodegeneration, whereas the survival of Wang et al., 2012). Currently there is no effec-
newly generated neurons was impaired at later tive drug or alternative therapy that adequately
stages, implying increased adverse changes in the reverses neurological deficits caused by stroke,
microenvironment with time after the onset of and hence nearly 50% of stroke survivors expe-
disease (Chen etal., 2008; Perry etal.,2012). rience lifelong disability. However, the observa-
Nevertheless, it remains to be determined tion in animal models that NSCs in the SVZ of
whether therapies that normalize the homeosta- the forebrain and the SGZ of the hippocampus
sis of NSCs and neurogenesis would slow down respond to stroke or ischemia with increased
the progression of AD if such interventions were proliferation and enhanced production of new
216 Part II:Homeostatic Control

neurons has provided a glimpse of self-repair Importantly, NSC and neurogenesis abla-
capability of the adult brain after insults. This tion approaches after ischemia have demon-
has resulted in a new direction of stroke research strated an exacerbation of ischemia-induced
focused on restoration of function through mobi- deficits, implying that targeted addition of new
lization of forebrain NSCs (Wang etal.,2012). neurons by NSCs after stroke/ischemia indeed
Multiple animal studies have shown that focal contributes to functional recovery (Zhang etal.,
cerebral ischemia stimulates NSC proliferation 2004; Wang et al., 2012). Hence, therapies hav-
in the SVZ, resulting in the production of a large ing promise to increase the activity of NSCs after
number of neuroblasts (Jin etal., 2001), which take stroke have attracted considerable attention.
a detour (from their normal path of entering the Studies in animal models have uncovered multi-
rostral migratory stream and reaching olfactory ple strategies to increase forebrain neurogenesis
bulb) and arrive in areas of stroke-induced brain as well as functional recovery through stimula-
damage (Parent etal., 2002; Ohab etal., 2006). In tion of NSCs after stroke. These include findings
these regions, neuroblasts undergo maturation that (i) intrastriatal infusion of glial cell-line
and express markers, mature neurons such as derived neurotrophic factor in the postischemic
the microtubule associated protein-2 and NeuN. period promotes several steps of striatal neuro-
An elegant study by Arvidsson and colleagues genesis after stroke (Kobayashi et al., 2006); (ii)
(2002) has further shown that stroke-generated the vascular endothelial growth factor improves
new neurons, following their migration into the stroke-induced striatal neurogenesis (Sun et al.,
severely damaged area of the striatum, differen- 2003; Wang etal., 2007); (iii) intrathecal admin-
tiate into mature, striatal medium-sized spiny istration of sonic hedgehog stimulates NSC pro-
neurons, implying that stroke induces differen- liferation and improves neurological recovery
tiation of NSC-derived new neurons into specific after stroke in rats (Bambakidis etal., 2012); (iv)
phenotypes that are destroyed by the ischemic overexpression of Wnt signaling enhances neuro-
lesion. Studies have suggested that up-regulation genesis and improves neurological function after
of erythropoitin after stroke promotes NSC pro- focal ischemic injury in mice (Shruster et al.,
liferation in the SVZ, the migration of newly 2012); (v)grafting of NSCs augments endogenous
born neuroblasts into the peri-infarct cortex, and neurogenesis and improves behavioral recov-
angiogenesis in the damaged brain area (Wang ery after stroke in rats (Mine et al., 2013); (vi)
et al., 2004b; Tsai et al., 2006). Poststroke neu- physical exercise boosts neurogenesis and eases
roblast migration has also been attributed to the functional deficits after stroke in rats (Zhang
vascular production of stromal-derived factor-1 et al., 2013); (vii) intravenous administration of
and angiopoietin-1 (Ohab etal., 2006). Follow-up bone marrow derived mesenchymal stem cells or
studies have also demonstrated the formation umbilical cord derived mesenchymal stem cells
of synapses between stroke-generated new neu- improves neurogenesis and promotes functional
rons and the host striatal neurons (Yamashita recovery in neonatal and adult rats undergo-
etal., 2006), as well as electrophysiological prop- ing ischemia (Yang et al., 2013; Iskander et al.,
erties of synaptic integration (Lai et al., 2008). 2013; Tsai etal., 2014; Tsuji etal., 2014); and (viii)
Investigations employing the global cerebral improved social environment (housing a mouse
ischemia models have also revealed the ability having stroke with a healthy cage mate) enhances
of NSCs for functional neuronal replacement both BDNF levels and neurogenesis (Venna
after injury (Nakatomi etal., 2002). In this pro- etal., 2014). Despite these findings, causeeffect
totype, activation of NSCs in the SVZ bordering relationship has not been established between
the hippocampus led to a massive regeneration of increased neurogenesis and functional recov-
hippocampal pyramidal neurons after ischemic ery after stroke. As neurogenesis enhanced by
brain injury. Neural progenitor cells generated strategies described here is often coupled with
from NSCs appeared to spontaneously migrate angiogenesis, it is plausible that the improved
into the hippocampus to regenerate new neu- neurological function observed may be a result
rons following ischemia in the CA1 subfield. of a combination of effects, including angiogen-
Interestingly, infusion of growth factors into the esis, neurogenesis, and axonal as well as dendritic
lateral ventricle enhanced this regeneration and plasticity (Zhang etal.,2014).
promoted integration of newly added neurons Ischemic injury also enhances NSC pro-
into the existing brain circuitry and ameliorated liferation and neurogenesis in the SGZ of the
neurological deficits (Nakatomi etal.,2002). hippocampus (Liu et al., 1998; Jin et al., 2001).
Adult Neural Stem Cells and Brain Homeostasis 217

Furthermore, poststroke-generated neurons are the ischemic region. However, it remains to be


recruited into the hippocampal networks (Geibig evaluated whether newly born neurons exhibit
etal, 2012). Studies imply that stroke stimulates long-term survival or contribute to brain repair
DG neurogenesis through activation of the cAMP after stroke in humans.
response element binding protein (Zhu et al.,
2004), Ca 2+ influx through L-type voltage-gated OVER ALL CONCLUSIONS
Ca2+ channels (Luo et al., 2005), reduced neu- It is now validated that NSCs persist in numer-
ronal nitric oxide synthase and upregulation ous regions of the adult mammalian brain in a
inducible nitric oxide synthase expression (Luo state of quiescence but become activated in con-
etal., 2007). The role of hippocampal neurogen- ditions such as disease or injury. Channeling
esis in functional recovery following stroke is these NSCs by facilitating their proliferation
unclear, however. Some studies imply that it helps and the neuronal differentiation of their prog-
in the recovery from spatial memory deficits eny would likely enhance neural repair after
(Luo etal., 2007), but direct evidence is lacking. injury or disease and help in maintaining brain
In contrast, a recent study shows that stimula- homeostasis. Neural stem cells in the adult hip-
tion of DG neurogenesis after stroke coincides pocampus in particular have received the most
with an increased rate of aberrantly integrated attention because their presence has been vali-
neurons into the hippocampus circuitry. This dated in the human brain at all ages and multi-
abnormal circuitry development may contribute ple animal studies signify that they are vital for
to functional impairments, cognitive deficits, or functions such as learning, memory, and mood.
epilepsy often seen in stroke patients (Niv etal., The extent of NSC proliferation and neurogenesis
2012). From this perspective, it may be neces- in the adult brain is regulated by multiple physi-
sary to target therapies to specifically increase ological stimuli and signaling pathways. Neural
SVZ neurogenesis to avoid an increased aberrant stem cells and hippocampal neurogenesis are
recruitment of newly born dentate granulecells. also highly sensitive to pathological conditions,
Studies on poststroke neurogenesis in and their altered homeostasis likely contributes
humans are rare. Jin and colleagues (2006b) pro- to functional deficits in several neurological
vided evidence for stroke-induced neurogenesis disorders. First, in the early phase of TLE devel-
in the human brain. The status of neurogenesis opment after SE or hippocampus injury, homeo-
was analyzed in brain sections obtained from stasis of NSCs and hippocampal neurogenesis is
human brain biopsies performed for the diagno- severely disrupted in the form of increased pro-
sis of cerebral lesions that proved to be ischemic liferation of NSCs and aberrant connectivity of
strokes. Control specimens obtained from the neurons recruited from their progeny. Clinically
cerebral cortex of autopsied patients who died applicable therapeutic strategies that can stabi-
without brain pathology exhibited either negligi- lize NSC and neurogenesis homeostasis after SE
ble or no proliferating cells. However, in speci- or hippocampus injury are therefore believed to
mens from stroke patients, the cortical region restrain the development of chronic spontane-
adjacent to the infarct core (ischemic penumbra) ous seizures as well as cognitive and mood dys-
contained considerable numbers of proliferating function. Second, in the chronic phase of TLE,
Ki-67+ cells, a fraction of which co-expressed the homeostasis of hippocampal neurogenesis is
immature neuronal marker DCX. Examination altered in the form of modified potential of NSCs
of MCM2 and PCNA expression in Ki-67+ cells and greatly decreased generation of new neurons
confirmed that Ki-67 labeling reflected recent (dentate granule cells). Application of approaches
cell proliferation. Furthermore, the ischemic capable of normalizing the potential of NSCs and
penumbra exhibited considerable numbers of increasing the extent of normal pattern of neuro-
cells positive for neuron-specific markers such as genesis is hence considered important for easing
DCX and Tuj-1. Some of the DCX+ or TuJ-1+ cells cognitive and mood impairments seen in chronic
also exhibited Ki-67 expression, suggesting that TLE. Third, impaired homeostasis of NSCs and
these are newly born neurons. Although analy- neurogenesis in the hippocampus is one of the
ses of biopsy specimens in this study precluded features of depressive disorders, and drug thera-
precise identification of the source of newly born pies that improve NSC activity and neurogenesis
neurons, their perivascular location suggested seem to ease cognitive and mood impairments
that newly born neurons likely originated in seen in depression. Fourth, altered NSC activ-
the SVZ and migrated along blood vessels into ity and hippocampal neurogenesis homeostasis
218 Part II:Homeostatic Control

has also been observed in AD though the extent Appleby, P.A., Kempermann, G., and Wiskott, L.
of changes (increase, no change, or decrease) (2011). The role of additive neurogenesis and syn-
seemed to depend on the stage of the disease. aptic plasticity in a hippocampal memory model
Hence, therapies that maintain or increase the with grid-cell like input. PLoS Comput. Biol. 7,
activity of NSCs and neurogenesis may slow e1001063.
down the progression of AD, if such therapies are Arvidsson, A., Collin, T., Kirik, D., Kokaia, Z., and
applied early after the disease onset. Fifth, stroke Lindvall, O. (2002). Neuronal replacement from
increases neurogenesis not only in the hippocam- endogenous precursors in the adult brain after
pus but also from the SVZ of the forebrain, which stroke. Nat. Med. 8, 963970.
has prompted the development of several strate- Bambakidis, N.C., Petrullis, M., Kui, X., Rothstein, B.,
gies to increase the activity of NSCs and neuro- Karampelas, I., Kuang, Y., Selman, W.R.,
genesis after stroke. Such approaches have shown LaManna, J.C., and Miller, R.H. (2012).
better functional recovery after stroke in animal Improvement of neurological recovery and stim-
models and hence have promise for ameliorating ulation of neural progenitor cell proliferation by
intrathecal administration of Sonic hedgehog. J.
functional deficits in stroke patients.
Neurosurg. 116, 11141120.
Banerjee, S.B., Rajendran, R., Dias, B.G., Ladiwala, U.,
ACK NOWLEDGMENTS
Tole, S., and Vaidya, V.A. (2005). Recruitment of
This work was supported by grants from the
the Sonic hedgehog signalling cascade in electro-
Texas A&M Health Science Center (Emerging
convulsive seizure-mediated regulation of adult
Technology Funds from the State of Texas),
rat hippocampal neurogenesis. Eur. J.Neurosci.
National Institute of Neurological Disorders and 22, 15701580.
Stroke (R01 NS054780 & R01 NS043507), and Barkas, L., Redhead, E., Taylor, M., Shtaya, A.,
the Department of Veterans Affairs (VA Merit Hamilton, D.A., and Gray, W.P. (2012). Fluoxetine
Review Award and Research Career Scientist restores spatial learning but not accelerated for-
Award). The author thanks Drs. B.Hattiangady, getting in mesial temporal lobe epilepsy. Brain
M.S. Rao, R.Kuruba, B.Shuai, and B.Waldau for 135, 23582374.
their excellent contributions to Shetty Lab neural Bedard, A., Gravel, C., and Parent, A. (2006).
stem cell and neurogenesis studies discussed in Chemical characterization of newly generated
this chapter. neurons in the striatum of adult primates. Exp.
Brain Res. 170, 501512.
References Bell, B., Lin, J.J., Seidenberg, M., and Hermann, B.
Ables, J.L., Decarolis, N.A., Johnson, M.A., Rivera, (2011). The neurobiology of cognitive disorders
P.D., Gao, Z., Cooper, D.C., Radtke, F., Hsieh, J., in temporal lobe epilepsy. Nat. Rev. Neurol. 7,
and Eisch, A.J. (2010). Notch1 is required for 154164.
maintenance of the reservoir of adult hippocam- Ben Menachem-Zidon, O., Menahem, Y.B., Hur,
pal stem cells. J. Neurosci. 30, 1048410492. T.B., and Yirmiya, R. (2014). Intra-hippocampal
Ahn, S., and Joyner, A.L. (2005). In vivo analysis of transplantation of neural precursor cells with
quiescent adult neural stem cells responding to transgenic over-expression of IL-1 receptor
Sonic hedgehog. Nature 437, 894897. antagonist rescues memory and neurogenesis
Aimone, J.B., Deng, W., and Gage, F.H. (2010). Adult impairments in an Alzheimers disease model.
neurogenesis: Integrating theories and separat- Neuropsychopharmacology 39, 401414.
ing functions. Trends Cogn. Sci. 14, 325337. Bengzon, J., Kokaia, Z., Elmer, E., Nanobashvili, A.,
Aimone, J.B., Deng, W., and Gage, F.H. (2011). Kokaia, M., and Lindvall, O. (1997). Apoptosis
Resolving new memories: A critical look at the and proliferation of dentate gyrus neurons after
dentate gyrus, adult neurogenesis, and pattern single and intermittent limbic seizures. Proc.
separation. Neuron 70, 589596. Natl. Acad. Sci. U.S.A. 94, 1043210437.
Altman, J., and Das, G.D. (1965). Autoradiographic Bergmann, O., and Frisen, J. (2013). Neuroscience:
and histological evidence of postnatal hippo- Why adults need new brain cells. Science 340,
campal neurogenesis in rats. J. Comp. Neurol. 695696.
124, 319335. Biscaro, B., Lindvall, O., Tesco, G., Ekdahl, C.T., and
Amrein, I., Isler, K., and Lipp, H.P. (2011). Comparing Nitsch, R.M. (2012). Inhibition of microglial acti-
adult hippocampal neurogenesis in mammalian vation protects hippocampal neurogenesis and
species and orders: Influence of chronological improves cognitive deficits in a transgenic mouse
age and life history stage. Eur. J. Neurosci. 34, model for Alzheimers disease. Neurodegener.
978987. Dis. 9, 187198.
Adult Neural Stem Cells and Brain Homeostasis 219

Blumcke, I., Schewe, J.C., Normann, S., Brustle, O., Saksida, L.M., Barker, R.A., Gage, F.H., and
Schramm, J., Elger, C.E., and Wiestler, O.D. Bussey, T.J. (2009). A functional role for adult
(2001). Increase of nestin-immunoreactive neu- hippocampal neurogenesis in spatial pattern
ral precursor cells in the dentate gyrus of pedi- separation. Science 325, 210213.
atric patients with early-onset temporal lobe Costa, D.A., Cracchiolo, J.R., Bachstetter, A.D.,
epilepsy. Hippocampus 11, 311321. Hughes, T.F., Bales, K.R., Paul, S.M., Mervis,
Boekhoorn, K., Joels, M., and Lucassen, P.J. (2006). R.F., Arendash, G.W., and Potter, H. (2007).
Increased proliferation reflects glial and Enrichment improves cognition in AD mice
vascular-associated changes, but not neurogen- by amyloid-related and unrelated mechanisms.
esis in the presenile Alzheimer hippocampus. Neurobiol. Aging 28, 831844.
Neurobiol. Dis. 24,114. Crespel, A., Rigau, V., Coubes, P., Rousset, M.C.,
Bonfanti, L., and Peretto, P. (2011). Adult neurogene- de Bock, F., Okano, H., Baldy-Moulinier, M.,
sis in mammalsa theme with many variations. Bockaert, J., and Lerner-Natoli, M. (2005).
Eur. J.Neurosci. 34, 930950. Increased number of neural progenitors in
Braun, S.M., and Jessberger, S. (2014). Review:Adult human temporal lobe epilepsy. Neurobiol. Dis.
neurogenesis and its role in neuropsychiatric 19, 436450.
disease, brain repair and normal brain function. Crews, L., Adame, A., Patrick, C., Delaney, A., Pham,
Neuropathol. Appl. Neurobiol. 40,312. E., Rockenstein, E., Hansen, L., and Masliah, E.
Bugra, K., Pollard, H., Charton, G., Moreau, J., (2010). Increased BMP6 levels in the brains of
Ben-Ari, Y., and Khrestchatisky, M. (1994). Alzheimers disease patients and APP transgenic
aFGF, bFGF and flg mRNAs show distinct pat- mice are accompanied by impaired neurogen-
terns of induction in the hippocampus follow- esis. J. Neurosci. 30, 1225212262.
ing kainate-induced seizures. Eur. J. Neurosci. Croll, S.D., Goodman, J.H., and Scharfman, H.E.
6,5866. (2004). Vascular endothelial growth factor
Busceti, C.L., Biagioni, F., Aronica, E., Riozzi, B., (VEGF) in seizures:Adouble-edged sword. Adv.
Storto, M., Battaglia, G., Giorgi, F.S., Gradini, R., Exp. Med. Biol. 548,5768.
Fornai, F., Caricasole, A., Nicoletti, F., and David, D.J., Samuels, B.A., Rainer, Q., Wang, J.W.,
Bruno, V. (2007). Induction of the Wnt inhibitor, Marsteller, D., Mendez, I., Drew, M., Craig,
Dickkopf-1, is associated with neurodegenera- D.A., Guiard, B.P., Guilloux, J.P., et al. (2009).
tion related to temporal lobe epilepsy. Epilepsia Neurogenesis-dependent and -independent
48, 694705. effects of fluoxetine in an animal model of anxi-
Butler, C.R., and Zeman, A.Z. (2008). Recent ety/depression. Neuron 62, 479493.
insights into the impairment of memory in Dayer, A.G., Ford, A.A., Cleaver, K.M., Yassaee,
epilepsy: Transient epileptic amnesia, acceler- M., and Cameron, H.A. (2003). Short-term and
ated long-term forgetting and remote memory long-term survival of new neurons in the rat den-
impairment. Brain 131, 22432263. tate gyrus. J. Comp. Neurol. 460, 563572.
Cameron, H.A., and Dayer, A.G. (2008). New inter- Deisseroth, K., Singla, S., Toda, H., Monje, M.,
neurons in the adult neocortex: Small, sparse, Palmer, T.D., and Malenka, R.C. (2004).
but significant? Biol. Psychiatry 63, 650655. Excitation-neurogenesis coupling in adult neural
Cameron, H. A., and McKay, R.D.G. (2001) Adult stem/progenitor cells. Neuron 42, 535552.
neurogenesis produces a large pool of new gran- Deng, W., Aimone, J.B., and Gage, F.H. (2010). New
ule cells in the dentate gyrus. J. Comp. Neurol. neurons and new memories: How does adult
435, 406417. hippocampal neurogenesis affect learning and
Cameron, H.A., Woolley, C.S., McEwen, B.S., and memory? Nat. Rev. Neurosci. 11, 339350.
Gould, E. (1993). Differentiation of newly born Devinsky, O. (2004). Therapy for neurobehavioral
neurons and glia in the dentate gyrus of the adult disorders in epilepsy. Epilepsia 45 Suppl 2,3440.
rat. Neurosci. 56, 337344. Donovan, M.H., Yazdani, U., Norris, R.D., Games,
Chen, Q., Nakajima, A., Choi, S.H., Xiong, X., D., German, D.C., and Eisch, A.J. (2006).
Sisodia, S.S., and Tang, Y.P. (2008). Adult neu- Decreased adult hippocampal neurogenesis in
rogenesis is functionally associated with AD-like the PDAPP mouse model of Alzheimers disease.
neurodegeneration. Neurobiol. Dis. 29, 316326. J. Comp. Neurol. 495,7083.
Chuang, T.T. (2010). Neurogenesis in mouse models Doze, V.A., Perez, D.M. (2012). G-protein-coupled
of Alzheimers disease. Biochim. Biophys. Acta receptors in adult neurogenesis. Pharmacol. Rev.
1802, 872880. 64, 645675.
Clelland, C.D., Choi, M., Romberg, C., Clemenson, Drapeau, E., Mayo, W., Aurousseau, C., Le Moal,
G.D. Jr., Fragniere, A., Tyers, P., Jessberger, S., M., Piazza, P.V., and Abrous, D.N. (2003). Spatial
220 Part II:Homeostatic Control

memory performances of aged rats in the water flexible use of spatially precise learning strate-
maze predict levels of hippocampal neurogenesis. gies. PLoS One 4, e45464.
Proc. Natl. Acad. Sci. U.S.A. 100, 1438514390. Ge, S., Pradhan, D.A., Ming, G.L., and Song, H. (2007).
DSa, C., and Duman, R.S. (2002). Antidepressants GABA sets the tempo for activity-dependent
and neuroplasticity. Bipolar Disord. 4, 183194. adult neurogenesis. Trends Neurosci. 30,18.
Duman, R.S., and Aghajanian, G.K. (2012). Synaptic Geibig, C.S., Keiner, S., and Redecker, C. (2012).
dysfunction in depression:Potential therapeutic Functional recruitment of newborn hippo-
targets. Science 338,6872. campal neurons after experimental stroke.
Dupret, D., Revest, J.M., Koehl, M., Ichas, F., De Neurobiol. Dis. 46, 431439.
Giorgi, F., Costet, P., Abrous, D.N., and Piazza, Gemma, C., and Bachstetter, A.D. (2013). The role of
P.V. (2008). Spatial relational memory requires microglia in adult hippocampal neurogenesis.
hippocampal adult neurogenesis. PLoS One Front. Cell. Neurosci. 7,229.
3,e1959. Goedert, M., and Spillantini, M.G. (2006). A century
Encinas, J.M., Michurina, T.V., Peunova, N., of Alzheimers disease. Science 314, 777781.
Park, J.H., Tordo, J., Peterson, D.A., Fishell, Goldman, S.A., and Chen, Z. (2011). Perivascular
G., Koulakov, A., and Enikolopov, G. (2011). instruction of cell genesis and fate in the adult
Division-coupled astrocytic differentiation and brain. Nat. Neurosci. 14, 13821389.
age-related depletion of neural stem cells in the Goldman, S.A., and Nottebohm, F. (1983). Neuronal
adult hippocampus. Cell. Stem Cell. 8, 566579. production, migration, and differentiation
Engel, J. Jr. (2001). Mesial temporal lobe epi- in a vocal control nucleus of the adult female
lepsy: What have we learned? Neuroscientist 7, canary brain. Proc. Natl. Acad. Sci. U.S.A. 80,
340352. 23902394.
Eriksson, P.S., Perfilieva, E., Bjork-Eriksson, T., Gong, C., Wang, T.W., Huang, H.S., and Parent, J.M.
Alborn, A.M., Nordborg, C., Peterson, D.A., (2007). Reelin regulates neuronal progenitor
and Gage, F.H. (1998). Neurogenesis in the adult migration in intact and epileptic hippocampus.
human hippocampus. Nat. Med. 4, 13131317. J. Neurosci. 27, 18031811.
Fahrner, A., Kann, G., Flubacher, A., Heinrich, C., Gould, E. (2007). How widespread is adult neurogen-
Freiman, T.M., Zentner, J., Frotscher, M., and esis in mammals? Nat. Rev. Neurosci. 8, 481488.
Haas, C.A. (2007). Granule cell dispersion is not Gould, E., McEwen, B.S., Tanapat, P., Galea, L.A.,
accompanied by enhanced neurogenesis in tem- and Fuchs, E. (1997). Neurogenesis in the dentate
poral lobe epilepsy patients. Exp. Neurol. 203, gyrus of the adult tree shrew is regulated by psy-
320332. chosocial stress and NMDA receptor activation.
Feng, R., Rampon, C., Tang, Y.P., Shrom, D., Jin, J., J. Neurosci. 17, 24922498.
Kyin, M., Sopher, B., Miller, M.W., Ware, C.B., Gould, E., Tanapat, P., Hastings, N.B., and Shors, T.J.
Martin, G.M., etal. (2001). Deficient neurogen- (1999). Neurogenesis in adulthood: A possible
esis in forebrain-specific presenilin-1 knockout role in learning. Trends Cogn. Sci. 3, 186192.
mice is associated with reduced clearance of hip- Gu, Y., Arruda-Carvalho, M., Wang, J., Janoschka,
pocampal memory traces. Neuron 32, 911926. S.R., Josselyn, S.A., Frankland, P.W., and
Filippov, V., Kronenberg, G., Pivneva, T., Reuter, K., Ge, S. (2012). Optical controlling reveals
Steiner, B., Wang, L.P., Yamaguchi, M., time-dependent roles for adult-born dentate
Kettenmann, H., and Kempermann, G. (2003). granule cells. Nat. Neurosci. 15, 17001706.
Subpopulation of nestin-expressing progenitor Haslinger, A., Schwarz, T.J., Covic, M., and Lie, D.C.
cells in the adult murine hippocampus shows elec- (2009). Expression of Sox11 in adult neurogenic
trophysiological and morphological characteris- niches suggests a stage-specific role in adult neu-
tics of astrocytes. Mol. Cell. Neurosci. 23, 373382. rogenesis. Eur. J.Neurosci. 29, 21032114.
Fowler, C.D., Liu, Y., and Wang, Z. (2008). Estrogen Hattiangady, B., Rao, M.S., and Shetty, A.K. (2004).
and adult neurogenesis in the amygdala and Chronic temporal lobe epilepsy is associated
hypothalamus. Brain Res. Rev. 57, 342351. with severely declined dentate neurogenesis
Gao, Z., Ure, K., Ding, P., Nashaat, M., Yuan, L., in the adult hippocampus. Neurobiol. Dis. 17,
Ma, J., Hammer, R.E., and Hsieh, J. (2011). The 473490.
master negative regulator REST/NRSF controls Hattiangady, B., and Shetty, A.K. (2008a). Aging does
adult neurogenesis by restraining the neurogenic not alter the number or phenotype of putative
program in quiescent stem cells. J. Neurosci. 31, stem/progenitor cells in the neurogenic region of
97729786. the hippocampus. Neurobiol. Aging 29, 129147.
Garthe, A., Behr, J., and Kempermann, G. (2009). Hattiangady, B., and Shetty, A.K. (2008b). Implications
Adult generated hippocampal neurons allow the of decreased hippocampal neurogenesis in
Adult Neural Stem Cells and Brain Homeostasis 221

chronic temporal lobe epilepsy. Epilepsia 49 Suppl and Lie, D.C. (2009). GABA-cAMP response
5, 2641. element-binding protein signaling regulates
Hattiangady, B., and Shetty, A.K. (2010). Decreased maturation and survival of newly generated neu-
neuronal differentiation of newly generated cells rons in the adult hippocampus. J. Neurosci. 29,
underlies reduced hippocampal neurogenesis in 79667977.
chronic temporal lobe epilepsy. Hippocampus Jessberger, S., Clark, R.E., Broadbent, N.J., Clemenson,
20, 97112. G.D. Jr., Consiglio, A., Lie, D.C., Squire, L.R., and
Haughey, N.J., Nath, A., Chan, S.L., Borchard, A.C., Gage, F.H. (2009). Dentate gyrus-specific knock-
Rao, M.S., and Mattson, M.P. (2002). Disruption down of adult neurogenesis impairs spatial and
of neurogenesis by amyloid beta-peptide, and object recognition memory in adult rats. Learn.
perturbed neural progenitor cell homeostasis, in Mem. 16, 147154.
models of Alzheimers disease. J. Neurochem. 83, Jessberger, S., Nakashima, K., Clemenson, G.D. Jr.,
15091524. Mejia, E., Mathews, E., Ure, K., Ogawa, S., Sinton,
Hester, M.S., and Danzer, S.C. (2014). Hippocampal C.M., Gage, F.H., and Hsieh, J. (2007). Epigenetic
granule cell pathology in epilepsya possible modulation of seizure-induced neurogenesis and
structural basis for comorbidities of epilepsy? cognitive decline. J. Neurosci. 27, 59675975.
Epilepsy Behav. 38, 105116. Jessberger, S., Romer, B., Babu, H., and Kempermann,
Hester, M.S., and Danzer, S.C. (2013). Accumulation G. (2005). Seizures induce proliferation and dis-
of abnormal adult-generated hippocampal gran- persion of doublecortin-positive hippocampal
ule cells predicts seizure frequency and severity. progenitor cells. Exp. Neurol. 196, 342351.
J. Neurosci. 33, 89268936. Jessberger, S., Toni, N., Clemenson, G.D. Jr., Ray, J.,
Ho, N.F., Hooker, J.M., Sahay, A., Holt, D.J., and and Gage, F.H. (2008). Directed differentiation of
Roffman, J.L. (2013). In vivo imaging of adult hippocampal stem/progenitor cells in the adult
human hippocampal neurogenesis:Progress, pit- brain. Nat. Neurosci. 11, 888893.
falls and promise. Mol. Psychiatry 18, 404416. Jhaveri, D.J., Mackay, E.W., Hamlin, A.S., Marathe,
Houser, C.R. (1990). Granule cell dispersion in the S.V., Nandam, L.S., Vaidya, V.A., and Bartlett, P.F.
dentate gyrus of humans with temporal lobe epi- (2010). Norepinephrine directly activates adult
lepsy. Brain Res. 535, 195204. hippocampal precursors via beta3-adrenergic
Hsieh, J. (2012). Orchestrating transcriptional receptors. J. Neurosci. 30, 27952806.
control of adult neurogenesis. Genes Dev. 26, Jin, K., Galvan, V., Xie, L., Mao, X.O., Gorostiza, O.F.,
10101021. Bredesen, D.E., and Greenberg, D.A. (2004b).
Ihrie, R.A., and Alvarez-Buylla, A. (2008). Cells in Enhanced neurogenesis in Alzheimers disease
the astroglial lineage are neural stem cells. Cell transgenic (PDGF-APPSw,Ind) mice. Proc. Natl.
Tissue Res. 331, 179191. Acad. Sci. U.S.A. 101, 1336313367.
Ihrie, R.A., and Alvarez-Buylla, A. (2011). Lake-front Jin, K., Minami, M., Lan, J.Q., Mao, X.O., Batteur,
property:Aunique germinal niche by the lateral S., Simon, R.P., and Greenberg, D.A. (2001).
ventricles of the adult brain. Neuron 70, 674686. Neurogenesis in dentate subgranular zone and
Ikrar, T., Guo, N., He, K., Besnard, A., Levinson, S., rostral subventricular zone after focal cerebral
Hill, A., Lee, H.K., Hen, R., Xu, X., and Sahay, A. ischemia in the rat. Proc. Natl. Acad. Sci. U.S.A.
(2013). Adult neurogenesis modifies excitability of 98, 47104715.
the dentate gyrus. Front. Neural Circuits 7,204. Jin, K., Peel, A.L., Mao, X.O., Xie, L., Cottrell,
Imayoshi, I., Sakamoto, M., Yamaguchi, M., Mori, K., B.A., Henshall, D.C., and Greenberg, D.A.
and Kageyama, R. (2010). Essential roles of (2004a). Increased hippocampal neurogenesis in
Notch signaling in maintenance of neural stem Alzheimers disease. Proc. Natl. Acad. Sci. U.S.A.
cells in developing and adult brains. J. Neurosci. 101, 343347.
30, 34893498. Jin, K., Wang, X., Xie, L., Mao, X.O., Zhu, W., Wang,
Iskander, A., Knight, R.A., Zhang, Z.G., Ewing, J.R., Y., Shen, J., Mao, Y., Banwait, S., and Greenberg,
Shankar, A., Varma, N.R., Bagher-Ebadian, H., D.A. (2006b). Evidence for stroke-induced neu-
Ali, M.M., Arbab, A.S., and Janic, B. (2013). rogenesis in the human brain. Proc. Natl. Acad.
Intravenous administration of human umbilical Sci. U.S.A. 103, 1319813202.
cord blood-derived AC133+ endothelial progeni- Jin, K., Xie, L., Mao, X.O., and Greenberg, D.A.
tor cells in rat stroke model reduces infarct vol- (2006a). Alzheimers disease drugs promote neu-
ume:Magnetic resonance imaging and histological rogenesis. Brain Res. 1085, 183188.
findings. Stem Cells Transl. Med. 2, 703714. Jorgensen, H.S., Nakayama, H., Raaschou, H.O.,
Jagasia, R., Steib, K., Englberger, E., Herold, S., and Olsen, T.S. (1997). Acute stroke care and
Faus-Kessler, T., Saxe, M., Gage, F.H., Song, H., rehabilitation: An analysis of the direct cost
222 Part II:Homeostatic Control

and its clinical and social determinants. The macaque monkey. Proc. Natl. Acad. Sci. U.S.A.
Copenhagen Stroke Study. Stroke 28, 11381141. 96, 57685773.
Jung, K.H., Chu, K., Kim, M., Jeong, S.W., Song, Krezymon, A., Richetin, K., Halley, H., Roybon,
Y.M., Lee, S.T., Kim, J.Y., Lee, S.K., and L., Lassalle, J.M., Frances, B., Verret, L., and
Roh, J.K. (2004). Continuous cytosine-b- Rampon, C. (2013). Modifications of hippocam-
D-arabinofuranoside infusion reduces ectopic pal circuits and early disruption of adult neuro-
granule cells in adult rat hippocampus with genesis in the tg2576 mouse model of Alzheimers
attenuation of spontaneous recurrent seizures disease. PLoS One 8, e76497.
following pilocarpine-induced status epilepti- Kuhn, H.G., Dickinson-Anson, H., and Gage, F.H.
cus. Eur. J.Neurosci. 19, 32193226. (1996). Neurogenesis in the dentate gyrus of the
Kaplan, M.S. (1981). Neurogenesis in the 3-month- adult rat:Age-related decrease of neuronal pro-
old rat visual cortex. J. Comp. Neurol. 195, genitor proliferation. J. Neurosci. 16, 20272033.
323338. Kuruba, R., Hattiangady, B., and Shetty, A.K. (2009).
Karalay, O., Doberauer, K., Vadodaria, K.C., Knobloch, Hippocampal neurogenesis and neural stem
M., Berti, L., Miquelajauregui, A., Schwark, M., cells in temporal lobe epilepsy. Epilepsy Behav.
Jagasia, R., Taketo, M.M., Tarabykin, V., Lie, D.C., 14 Suppl 1,6573.
and Jessberger, S. (2011). Prospero-related homeo- Kuwabara, T., Hsieh, J., Muotri, A., Yeo, G.,
box 1 gene (Prox1) is regulated by canonical Wnt Warashina, M., Lie, D.C., Moore, L., Nakashima,
signaling and has a stage-specific role in adult K., Asashima, M., and Gage, F.H. (2009).
hippocampal neurogenesis. Proc. Natl. Acad. Sci. Wnt-mediated activation of NeuroD1 and
U.S.A. 108, 58075812. retro-elements during adult neurogenesis. Nat.
Kee, N., Teixeira, C.M., Wang, A.H., and Frankland, Neurosci. 12, 10971105.
P.W. (2007). Preferential incorporation of adult- Lagace, D.C., Whitman, M.C., Noonan, M.A., Ables,
generated granule cells into spatial memory net- J.L., DeCarolis, N.A., Arguello, A.A., Donovan,
works in the dentate gyrus. Nat. Neurosci. 10, M.H., Fischer, S.J., Farnbauch, L.A., Beech,
355362. R.D., et al. (2007). Dynamic contribution of
Kempermann, G. (2012). Youth culture in the adult nestin-expressing stem cells to adult neurogen-
brain. Science 335, 11751176. esis. J. Neurosci. 27, 1262312629.
Kempermann, G. (2013). Neuroscience. What Lai, B., Mao, X.O., Xie, L., Jin, K., and Greenberg, D.A.
the bomb said about the brain. Science 340, (2008). Electrophysiological neurodifferentiation
11801181. of subventricular zone-derived precursor cells fol-
Kempermann, G., Jessberger, S., Steiner, B., and lowing stroke. Neurosci. Lett. 442, 305308.
Kronenberg, G. (2004). Milestones of neuronal Lajud, N., Roque, A., Cajero, M., Gutierrez-Ospina,
development in the adult hippocampus. Trends G., and Torner, L. (2012). Periodic maternal
Neurosci. 27, 447452. separation decreases hippocampal neurogen-
Kempermann, G., Kuhn, H.G., and Gage, F.H. (1997). esis without affecting basal corticosterone dur-
More hippocampal neurons in adult mice living in ing the stress hyporesponsive period, but alters
an enriched environment. Nature 386, 493495. HPA axis and coping behavior in adulthood.
Kheirbek, M.A., and Hen, R. (2013). (Radio)active Psychoneuroendocrinology 37, 410420.
neurogenesis in the human hippocampus. Cell Lazarov, O., Mattson, M.P., Peterson, D.A.,
153, 11831184. Pimplikar, S.W., and van Praag, H. (2010). When
Kheirbek, M.A., Klemenhagen, K.C., Sahay, A., and neurogenesis encounters aging and disease.
Hen, R. (2012). Neurogenesis and generaliza- Trends Neurosci. 33, 569579.
tion:Anew approach to stratify and treat anxiety Lee, J., Duan, W., and Mattson, M.P. (2002).
disorders. Nat. Neurosci. 15, 16131620. Evidence that brain-derived neurotrophic fac-
Kobayashi, T., Ahlenius, H., Thored, P., Kobayashi, R., tor is required for basal neurogenesis and medi-
Kokaia, Z., and Lindvall, O. (2006). Intracerebral ates, in part, the enhancement of neurogenesis by
infusion of glial cell line-derived neurotrophic dietary restriction in the hippocampus of adult
factor promotes striatal neurogenesis after stroke mice. J. Neurochem. 82, 13671375.
in adult rats. Stroke 37, 23612367. Lee, M.M., Reif, A., and Schmitt, A.G. (2013). Major
Koketsu, D., Mikami, A., Miyamoto, Y., and depression: A role for hippocampal neurogen-
Hisatsune, T. (2003). Nonrenewal of neurons in esis? Curr. Top. Behav. Neurosci. 14, 153179.
the cerebral neocortex of adult macaque mon- Leutgeb, J.K., Leutgeb, S., Moser, M.B., and Moser,
keys. J. Neurosci. 23, 937942. E.I. (2007). Pattern separation in the dentate
Kornack, D.R., and Rakic, P. (1999). Continuation gyrus and CA3 of the hippocampus. Science 315,
of neurogenesis in the hippocampus of the adult 961966.
Adult Neural Stem Cells and Brain Homeostasis 223

Lewis, D.V. (2005). Losing neurons:Selective vulner- by up-regulating inducible nitric oxide synthase
ability and mesial temporal sclerosis. Epilepsia expression. J. Neurochem. 103, 18721882.
46 Suppl 7,3944. Manganas, L.N., Zhang, X., Li, Y., Hazel, R.D.,
Li, X., Guo, F., Zhang, Q., Huo, T., Liu, L., Smith, S.D., Wagshul, M.E., Henn, F.,
Wei, H., Xiong, L., and Wang, Q. (2014). Benveniste, H., Djuric, P.M., Enikolopov, G., and
Electroacupuncture decreases cognitive impair- Maletic-Savatic, M. (2007). Magnetic resonance
ment and promotes neurogenesis in the APP/ spectroscopy identifies neural progenitor cells in
PS1 transgenic mice. BMC Complement. Altern. the live human brain. Science 318, 980985.
Med. 14, 37-6882-14-37. Marn-Burgin, A., Mongiat, L.A., Pardi, M.B., and
Lie, D.C., Colamarino, S.A., Song, H.J., Desire, L., Schinder A.F. (2012). Unique processing during
Mira, H., Consiglio, A., Lein, E.S., Jessberger, S., a period of high excitation/inhibition balance in
Lansford, H., Dearie, A.R., and Gage, F.H. (2005). adult born neurons. Science 335, 12381242.
Wnt signalling regulates adult hippocampal neu- Masi, G., and Brovedani, P. (2011). The hippocampus,
rogenesis. Nature 437, 13701375. neurotrophic factors and depression: Possible
Lie, D.C., Dziewczapolski, G., Willhoite, A.R., implications for the pharmacotherapy of depres-
Kaspar, B.K., Shults, C.W., and Gage, F.H. (2002). sion. CNS Drugs 25, 913931.
The adult substantia nigra contains progenitor Masiulis, I., Yun, S., and Eisch, A.J. (2011). The inter-
cells with neurogenic potential. J. Neurosci. 22, esting interplay between interneurons and adult
66396649. hippocampal neurogenesis. Mol. Neurobiol. 44,
Liu, J., Solway, K., Messing, R.O., and Sharp, F.R. 287302.
(1998). Increased neurogenesis in the dentate Mathern, G.W., Leiphart, J.L., De Vera, A., Adelson,
gyrus after transient global ischemia in gerbils. J. P.D., Seki, T., Neder, L., and Leite, J.P. (2002).
Neurosci. 18, 77687778. Seizures decrease postnatal neurogenesis and
Lopez-Toledano, M.A., and Shelanski, M.L. (2007). granule cell development in the human fascia
Increased neurogenesis in young transgenic dentata. Epilepsia 43 Suppl 5,6873.
mice overexpressing human APP(Sw, Ind). J. McCloskey, D.P., Hintz, T.M., Pierce, J.P., and
Alzheimers Dis. 12, 229240. Scharfman, H.E. (2006). Stereological methods
Lowenstein, D.H., Seren, M.S., and Longo, F.M. reveal the robust size and stability of ectopic
(1993). Prolonged increases in neurotrophic hilar granule cells after pilocarpine-induced sta-
activity associated with kainate-induced hippo- tus epilepticus in the adult rat. Eur. J.Neurosci.
campal synaptic reorganization. Neuroscience 24, 22032210.
56, 597604. Mine, Y., Tatarishvili, J., Oki, K., Monni, E., Kokaia,
Lugert, S., Basak, O., Knuckles, P., Haussler, U., Fabel, Z., and Lindvall, O. (2013). Grafted human neu-
K., Gotz, M., Haas, C.A., Kempermann, G., ral stem cells enhance several steps of endog-
Taylor, V., and Giachino, C. (2010). Quiescent enous neurogenesis and improve behavioral
and active hippocampal neural stem cells with recovery after middle cerebral artery occlusion
distinct morphologies respond selectively to in rats. Neurobiol. Dis. 52, 191203.
physiological and pathological stimuli and Ming, G.L., and Song, H. (2011). Adult neurogenesis
aging. Cell. Stem Cell. 6, 445456. in the mammalian brain: Significant answers
Lugert, S., Vogt, M., Tchorz, J.S., Muller, M., and significant questions. Neuron 70, 687702.
Giachino, C., and Taylor, V. (2012). Homeostatic Mirescu, C., Peters, J.D., and Gould, E. (2004). Early
neurogenesis in the adult hippocampus does not life experience alters response of adult neurogen-
involve amplification of Ascl1(high) intermedi- esis to stress. Nat. Neurosci. 7, 841846.
ate progenitors. Nat. Commun. 3,670. Mohapel, P., Leanza, G., Kokaia, M., Lindvall,
Luo, C.X., Zhu, X.J., Zhang, A.X., Wang, W., O. (2005). Forebrain acetylcholine regulates
Yang, X.M., Liu, S.H., Han, X., Sun, J., adult hippocampal neurogenesis and learning.
Zhang, S.G., Lu, Y., and Zhu, D.Y. (2005). Neurobiol. Aging 26, 93946.
Blockade of L-type voltage-gated Ca chan- Morrens, J., Van Den Broeck, W., and Kempermann,
nel inhibits ischemia-induced neurogenesis G. (2012). Glial cells in adult neurogenesis. Glia
by down-regulating iNOS expression in adult 60, 159174.
mouse. J. Neurochem. 94, 10771086. Mu, Y., and Gage, F.H. (2011). Adult hippocampal
Luo, C.X., Zhu, X.J., Zhou, Q.G., Wang, B., Wang, W., neurogenesis and its role in Alzheimers disease.
Cai, H.H., Sun, Y.J., Hu, M., Jiang, J., Hua, Y., Mol. Neurodegener6,85.
Han, X., and Zhu, D.Y. (2007). Reduced neu- Nakagawa, S., Kim, J.E., Lee, R., Chen, J., Fujioka,
ronal nitric oxide synthase is involved in T., Malberg, J., Tsuji, S., and Duman, R.S. (2002).
ischemia-induced hippocampal neurogenesis Localization of phosphorylated cAMP response
224 Part II:Homeostatic Control

element-binding protein in immature neurons of Parent, J.M., Yu, T.W., Leibowitz, R.T., Geschwind,
adult hippocampus. J. Neurosci. 22, 98689876. D.H., Sloviter, R.S., and Lowenstein, D.H. (1997).
Nakashiba, T., Cushman, J.D., Pelkey, K.A., Dentate granule cell neurogenesis is increased
Renaudineau, S., Buhl, D.L., McHugh, T.J., by seizures and contributes to aberrant network
Rodriguez Barrera, V., Chittajallu, R., Iwamoto, reorganization in the adult rat hippocampus. J.
K.S., McBain, C.J., Fanselow, M.S., and Neurosci. 17, 37273738.
Tonegawa, S. (2012). Young dentate granule cells Pekcec, A., Fuest, C., Muhlenhoff, M.,
mediate pattern separation, whereas old gran- Gerardy-Schahn, R., and Potschka, H. (2008).
ule cells facilitate pattern completion. Cell 149, Targeting epileptogenesis-associated induction
188201. of neurogenesis by enzymatic depolysialylation
Nakatomi, H., Kuriu, T., Okabe, S., Yamamoto, S., of NCAM counteracts spatial learning dysfunc-
Hatano, O., Kawahara, N., Tamura, A., Kirino, T., tion but fails to impact epilepsy development. J.
and Nakafuku, M. (2002). Regeneration of hip- Neurochem. 105, 389400.
pocampal pyramidal neurons after ischemic Pekcec, A., Lupke, M., Baumann, R., Seifert, H., and
brain injury by recruitment of endogenous neu- Potschka, H. (2011). Modulation of neurogen-
ral progenitors. Cell 110, 429441. esis by targeted hippocampal irradiation fails to
Nichol, K., Deeny, S.P., Seif, J., Camaclang, K., and affect kindling progression. Hippocampus 21,
Cotman, C.W. (2009). Exercise improves cogni- 866876.
tion and hippocampal plasticity in APOE epsi- Pekcec, A., Muhlenhoff, M., Gerardy-Schahn, R., and
lon4 mice. Alzheimers Dement. 5, 287294. Potschka, H. (2007). Impact of the PSA-NCAM
Nichol, K.E., Parachikova, A.I., and Cotman, C.W. system on pathophysiology in a chronic rodent
(2007). Three weeks of running wheel exposure model of temporal lobe epilepsy. Neurobiol. Dis.
improves cognitive performance in the aged 27,5466.
Tg2576 mouse. Behav. Brain Res. 184, 124132. Perera, T.D., Coplan, J.D., Lisanby, S.H., Lipira, C.M.,
Niv, F., Keiner, S., Krishna, Witte, O.W., Lie, D.C., Arif, M., Carpio, C., Spitzer, G., Santarelli, L.,
and Redecker, C. (2012). Aberrant neurogen- Scharf, B., Hen, R., et al. (2007). Antidepressant-
esis after stroke:Aretroviral cell labeling study. induced neurogenesis in the hippocampus
Stroke 43, 24682475. of adult nonhuman primates. J. Neurosci. 27,
Ohab, J.J., Fleming, S., Blesch, A., and Carmichael, S.T. 48944901.
(2006). A neurovascular niche for neurogenesis Pereira, A.C., Huddleston, D.E., Brickman, A.M.,
after stroke. J. Neurosci. 26, 1300713016. Sosunov, A.A., Hen, R., McKhann, G.M., Sloan, R.,
Ohm, T.G. (2007). The dentate gyrus in Alzheimers Gage, F.H., Brown, T.R., and Small, S.A. (2007).
disease. Prog. Brain Res. 163, 723740. An in vivo correlate of exercise-induced neuro-
Okano, H., Kawahara, H., Toriya, M., Nakao, K., genesis in the adult dentate gyrus. Proc. Natl.
Shibata, S., and Imai, T. (2005). Function of Acad. Sci. U.S.A. 104, 56385643.
RNA-binding protein Musashi-1 in stem cells. Perry, E.K., Johnson, M., Ekonomou, A., Perry, R.H.,
Exp. Cell Res. 306, 349356. Ballard, C., and Attems, J. (2012). Neurogenic
OReilly, R.C., and McClelland, J.L. (1994). abnormalities in Alzheimers disease differ
Hippocampal conjunctive encoding, storage, between stages of neurogenesis and are partly
and recall: Avoiding a trade-off. Hippocampus related to cholinergic pathology. Neurobiol. Dis.
4, 661682. 47, 155162.
Palmer, T.D., Markakis, E.A., Willhoite, A.R., Safar, F., Pieper, A.A., Xie, S., Capota, E., Estill, S.J., Zhong, J.,
and Gage, F.H. (1999). Fibroblast growth factor-2 Long, J.M., Becker, G.L., Huntington, P.,
activates a latent neurogenic program in neural Goldman, S.E., Shen, C.H., etal. (2010) Discovery
stem cells from diverse regions of the adult CNS. of proneurogenic, neuroprotective chemical.
J. Neurosci. 19, 84878497. Cell 142,3951.
Palmer, T.D., Willhoite, A.R., and Gage, F.H. (2000). Pierce, J.P., Melton, J., Punsoni, M., McCloskey, D.P.,
Vascular niche for adult hippocampal neurogen- and Scharfman, H.E. (2005). Mossy fibers
esis. J. Comp. Neurol. 425, 479494. are the primary source of afferent input
Pandya, M., Altinay, M., Malone, D.A. Jr., and to ectopic granule cells that are born after
Anand, A. (2012). Where in the brain is depres- pilocarpine-induced seizures. Exp. Neurol.
sion? Curr. Psychiatry Rep. 14, 634642. 196, 316331.
Parent, J.M., Vexler, Z.S., Gong, C., Derugin, N., and Pirttil, T.J., Manninen, A., Jutila, L., Nissinen, J.,
Ferriero, D.M. (2002). Rat forebrain neurogen- Klviinen, R., Vapalahti, M., Immonen, A.,
esis and striatal neuron replacement after focal Paljrvi, L., Karkola, K., Alafuzoff, I., Mervaala,
stroke. Ann. Neurol. 52, 802813. E., and Pitknen, A. (2005). Cystatin C expression
Adult Neural Stem Cells and Brain Homeostasis 225

is associated with granule cell dispersion in epi- in hippocampus and olfactory bulb. Neuron 70,
lepsy. Ann. Neurol. 58, 211223. 582588.
Raedt, R., Boon, P., Persson, A., Alborn, A.M., Santarelli, L., Saxe, M., Gross, C., Surget, A.,
Boterberg, T., Van Dycke, A., Linder, B., De Battaglia, F., Dulawa, S., Weisstaub, N., Lee, J.,
Smedt, T., Wadman, W.J., Ben-Menachem, E., Duman, R., Arancio, O., Belzung, C., and Hen, R.
and Eriksson, P.S. (2007). Radiation of the rat (2003). Requirement of hippocampal neurogen-
brain suppresses seizure-induced neurogenesis esis for the behavioral effects of antidepressants.
and transiently enhances excitability during kin- Science 301, 805809.
dling acquisition. Epilepsia 48, 19521963. Sapolsky, R.M. (2001). Depression, antidepressants,
Rao, M.S., Hattiangady, B., Abdel-Rahman, A., and the shrinking hippocampus. Proc. Natl.
Stanley, D.P., and Shetty, A.K. (2005). Newly Acad. Sci. U.S.A. 98, 1232012322.
born cells in the ageing dentate gyrus display Sapolsky, R.M. (2004). Is impaired neurogenesis rel-
normal migration, survival and neuronal fate evant to the affective symptoms of depression?
choice but endure retarded early maturation. Biol. Psychiatry 56, 137139.
Eur. J.Neurosci. 21, 464476. Saxe, M.D., Battaglia, F., Wang, J.W., Malleret,
Rao, M.S., Hattiangady, B., and Shetty, A.K. G., David, D.J., Monckton, J.E., Garcia, A.D.,
(2006). The window and mechanisms of major Sofroniew, M.V., Kandel, E.R., Santarelli, L.,
age-related decline in the production of new neu- et al. (2006). Ablation of hippocampal neuro-
rons within the dentate gyrus of the hippocam- genesis impairs contextual fear conditioning and
pus. Aging Cell. 5, 545558. synaptic plasticity in the dentate gyrus. Proc.
Reitz, C., and Mayeux, R. (2014). Alzheimer disease: Natl. Acad. Sci. U.S.A. 103, 1750117506.
Epidemiology, diagnostic criteria, risk factors and Scharfman, H.E., Goodman, J.H., and Sollas,
biomarkers. Biochem. Pharmacol. 88, 640651. A.L. (2000). Granule-like neurons at the
Renault, V.M., Rafalski, V.A., Morgan, A.A., Salih, D.A., hilar/CA3 border after status epilepticus and
Brett, J.O., Webb, A.E., Villeda, S.A., Thekkat, their synchrony with area CA3 pyramidal
P.U., Guillerey, C., Denko, N.C., et al. (2009). cells:Functional implications of seizure-induced
FoxO3 regulates neural stem cell homeostasis. neurogenesis. J. Neurosci. 20, 61446158.
Cell. Stem Cell. 5, 527539. Scharfman, H.E., and Gray, W.P. (2006). Plasticity
Reynolds, B.A., and Weiss, S. (1992). Generation of of neuropeptide Y in the dentate gyrus after sei-
neurons and astrocytes from isolated cells of zures, and its relevance to seizure-induced neu-
the adult mammalian central nervous system. rogenesis. EXS 95, 193211.
Science 255, 17071710. Scharfman, H.E., and Gray, W.P. (2007). Relevance
Rietze, R., Poulin, P., and Weiss, S. (2000). Mitotically of seizure-induced neurogenesis in animal mod-
active cells that generate neurons and astrocytes els of epilepsy to the etiology of temporal lobe
are present in multiple regions of the adult mouse epilepsy. Epilepsia 48 Suppl 2,3341.
hippocampus. J. Comp. Neurol. 424, 397408. Scharfman, H.E., Sollas, A.E., Berger, R.E., Goodman,
Rodriguez, J.J., Jones, V.C., Tabuchi, M., Allan, S.M., J.H., and Pierce, J.P. (2003a). Perforant path acti-
Knight, E.M., LaFerla, F.M., Oddo, S., and vation of ectopic granule cells that are born after
Verkhratsky, A. (2008). Impaired adult neuro- pilocarpine-induced seizures. Neuroscience 121,
genesis in the dentate gyrus of a triple transgenic 10171029.
mouse model of Alzheimers disease. PLoS One Scharfman, H.E., Sollas, A.L., Berger, R.E., and
3,e2935. Goodman, J.H. (2003b). Electrophysiological evi-
Rolando, C., and Taylor, V. (2014). Neural stem cell dence of monosynaptic excitatory transmission
of the hippocampus: Development, physiology between granule cells after seizure-induced mossy
regulation, and dysfunction in disease. Curr. fiber sprouting. J. Neurophysiol. 90, 25362547.
Top. Dev. Biol. 107, 183206. Scharfman, H.E., Sollas, A.L., and Goodman, J.H.
Sahay, A., and Hen, R. (2007). Adult hippocampal (2002). Spontaneous recurrent seizures after
neurogenesis in depression. Nat. Neurosci. 10, pilocarpine-induced status epilepticus activate
11101115. calbindin-immunoreactive hilar cells of the rat
Sahay, A., Scobie, K.N., Hill, A.S., OCarroll, C.M., dentate gyrus. Neuroscience 111,7181.
Kheirbek, M.A., Burghardt, N.S., Fenton, A.A., Scheff, S.W., and Price, D.A. (2006). Alzheimers
Dranovsky, A., and Hen, R. (2011a). Increasing disease-related alterations in synaptic den-
adult hippocampal neurogenesis is sufficient to sity:Neocortex and hippocampus. J. Alzheimers
improve pattern separation. Nature 472, 466470. Dis. 9, 101115.
Sahay, A., Wilson, D.A., and Hen, R. (2011b). Pattern Schmidt-Hieber, C., Jonas, P., and Bischofberger, J.
separation:Acommon function for new neurons (2004). Enhanced synaptic plasticity in newly
226 Part II:Homeostatic Control

generated granule cells of the adult hippocam- and improves neurological function after focal
pus. Nature 429, 184187. ischemic injury. PLoS One 7, e40843.
Seib, D.R., Corsini, N.S., Ellwanger, K., Plaas, C., Snyder, J.S., Hong, N.S., McDonald, R.J., and
Mateos, A., Pitzer, C., Niehrs, C., Celikel, T., Wojtowicz, J.M. (2005). A role for adult neu-
and Martin-Villalba, A. (2013). Loss of rogenesis in spatial long-term memory.
Dickkopf-1 restores neurogenesis in old age Neuroscience 130, 843852.
and counteracts cognitive decline. Cell. Stem Snyder, J.S., Kee, N., and Wojtowicz, J. M. (2001).
Cell. 12, 204214. Effects of adult neurogenesis on synaptic plastic-
Selkoe, D.J. (2001). Presenilin, Notch, and the genesis ity in the rat dentate gyrus. J. Neurophysiol. 85,
and treatment of Alzheimers disease. Proc. Natl. 24232431.
Acad. Sci. U.S.A. 98, 1103911041. Snyder, J.S., Soumier, A., Brewer, M., Pickel, J., and
Seri, B., Garcia-Verdugo, J.M., McEwen, B.S., and Cameron, H.A. (2011). Adult hippocampal neu-
Alvarez-Buylla, A. (2001). Astrocytes give rise rogenesis buffers stress responses and depressive
to new neurons in the adult mammalian hippo- behaviour. Nature 476, 458461.
campus. J. Neurosci. 21, 71537160. Song, H., Stevens, C.F., and Gage, F.H. (2002).
Shapiro, L.A., Korn, M.J., and Ribak, C.E. (2005). Astroglia induce neurogenesis from adult neural
Newly generated dentate granule cells from epi- stem cells. Nature 417,3944.
leptic rats exhibit elongated hilar basal dendrites Song, J., Sun, J., Moss, J., Wen, Z., Sun, G. J., Hsu,
that align along GFAP-immunolabeled pro- D., Zhong, C., Davoudi, H., Christian, K.M.,
cesses. Neuroscience 136, 823831. Toni, N., etal. (2013). Parvalbumin interneurons
Shapiro, L.A., Ng, K.L., Kinyamu, R., Whitaker- mediate neuronal circuitry-neurogenesis cou-
Azmitia, P., Geisert, E.E., Blurton-Jones, M., pling in the adult hippocampus. Nat. Neurosci.
Zhou, Q.Y., and Ribak, C.E. (2007). Origin, 16, 17281730.
migration and fate of newly generated neurons Song, J., Zhong, C., Bonaguidi, M. A., Sun, G. J.,
in the adult rodent piriform cortex. Brain Struct. Hsu, D., Gu, Y., Meletis, K., Huang, Z.J., Ge, S.,
Funct. 212, 133148. Enikolopov, G, et al. (2012). Neuronal circuitry
Shen, L., Nam, H.S., Song, P., Moore, H., and mechanism regulating adult quiescent neural
Anderson, S.A. (2006). FoxG1 haploinsufficiency stem-cell fate decision. Nature 489, 150154.
results in impaired neurogenesis in the postnatal Spalding, K.L., Bergmann, O., Alkass, K., Bernard,
hippocampus and contextual memory deficits. S., Salehpour, M., Huttner, H.B., Bostrom, E.,
Hippocampus 16, 875890. Westerlund, I., Vial, C., Buchholz, B.A., et al.
Shetty, A.K. (2011). Progress in cell grafting therapy (2013). Dynamics of hippocampal neurogenesis
for temporal lobe epilepsy. Neurotherapeutics 8, in adult humans. Cell 153, 12191227.
721735. Strine, T.W., Kobau, R., Chapman, D.P., Thurman,
Shetty, A.K., and Hattiangady, B. (2007). Concise D.J., Price, P., and Balluz, L.S. (2005).
review:Prospects of stem cell therapy for tempo- Psychological distress, comorbidities, and
ral lobe epilepsy. Stem Cells 25, 23962407. health behaviors among U.S. adults with sei-
Shetty, A.K., Rao, M.S., Hattiangady, B., Zaman, V., zures: Results from the 2002 National Health
and Shetty, G.A. (2004). Hippocampal neuro- Interview Survey. Epilepsia 46, 11331139.
trophin levels after injury: Relationship to the Sun, Y., Jin, K., Xie, L., Childs, J., Mao, X.O.,
age of the hippocampus at the time of injury. J. Logvinova, A., and Greenberg, D.A. (2003).
Neurosci. Res. 78, 520532. VEGF-induced neuroprotection, neurogenesis,
Shetty, A.K., Zaman, V., and Shetty, G.A. (2003). and angiogenesis after focal cerebral ischemia. J.
Hippocampal neurotrophin levels in a kainate Clin. Invest. 111, 18431851.
model of temporal lobe epilepsy: a lack of cor- Tiwari, S.K., Agarwal, S., Seth, B., Yadav, A., Nair, S.,
relation between brain-derived neurotrophic Bhatnagar, P., Karmakar, M., Kumari, M.,
factor content and progression of aberrant den- Chauhan, L.K., Patel, D.K., et al. (2014).
tate mossy fiber sprouting. J. Neurochem. 87, Curcumin-loaded nanoparticles potently induce
147159. adult neurogenesis and reverse cognitive defi-
Shors, T.J., Miesegaes, G., Beylin, A., Zhao, M., cits in Alzheimers disease model via canonical
Rydel, T., and Gould, E. (2001). Neurogenesis Wnt/-catenin pathway. ACS Nano 8, 76103.
in the adult is involved in the formation of trace Tozuka, Y., Fukuda, S., Namba, T., Seki, T., and
memories. Nature 410, 372376. Hisatsune, T. (2005). GABAergic excitation pro-
Shruster, A., Ben-Zur, T., Melamed, E., and Offen, D. motes neuronal differentiation in adult hippo-
(2012). Wnt signaling enhances neurogenesis campal progenitor cells. Neuron 47, 803815.
Adult Neural Stem Cells and Brain Homeostasis 227

Tronel, S., Fabre, A., Charrier, V., Oliet, S.H., Gage, F.H., Pilocarpine-induced seizures cause selective
and Abrous, D.N. (2010). Spatial learning sculpts time-dependent changes to adult-generated hip-
the dendritic arbor of adult-born hippocam- pocampal dentate granule cells. J. Neurosci. 27,
pal neurons. Proc. Natl. Acad. Sci. U.S.A. 107, 75417552.
79637968. Wang, L., Zhang, Z., Wang, Y., Zhang, R., and
Tsai, M.J., Tsai, S.K., Hu, B.R., Liou, D.Y., Huang, S.L., Chopp, M. (2004b). Treatment of stroke with
Huang, M.C., Huang, W.C., Cheng, H., and erythropoietin enhances neurogenesis and
Huang, S.S. (2014). Recovery of neurological angiogenesis and improves neurological func-
function of ischemic stroke by application of tion in rats. Stroke 35, 17321737.
conditioned medium of bone marrow mesenchy- Wang, R., Dineley, K.T., Sweatt, J.D., and Zheng, H.
mal stem cells derived from normal and cerebral (2004a). Presenilin 1 familial Alzheimers disease
ischemia rats. J. Biomed. Sci.21,5. mutation leads to defective associative learning
Tsai, P.T., Ohab, J.J., Kertesz, N., Groszer, M., Matter, C., and impaired adult neurogenesis. Neuroscience
Gao, J., Liu, X., Wu, H., and Carmichael, S.T. 126, 305312.
(2006). A critical role of erythropoietin recep- Wang, S., Scott, B.W., and Wojtowicz, J.M. (2000).
tor in neurogenesis and post-stroke recovery. J. Heterogeneous properties of dentate granule neu-
Neurosci. 26, 12691274. rons in the adult rat. J. Neurobiol. 42, 248257.
Tsuji, M., Taguchi, A., Ohshima, M., Kasahara, Y., Sato, Wang, X., Mao, X., Xie, L., Sun, F., Greenberg, D.A.,
Y., Tsuda, H., Otani, K., Yamahara, K., Ihara, M., and Jin, K. (2012). Conditional depletion of
Harada-Shiba, M., Ikeda, T., and Matsuyama, T. neurogenesis inhibits long-term recovery after
(2014). Effects of intravenous administration of experimental stroke in mice. PLoS One 7, e38932.
umbilical cord blood CD34 cells in a mouse model Wang, Y., Ristevski, S., and Harley, V.R. (2006).
of neonatal stroke. Neuroscience 263,148158. SOX13 exhibits a distinct spatial and temporal
van Praag, H., Christie, B.R., Sejnowski, T.J., and expression pattern during chondrogenesis, neu-
Gage, F.H. (1999). Running enhances neurogene- rogenesis, and limb development. J. Histochem.
sis, learning, and long-term potentiation in mice. Cytochem. 54, 13271333.
Proc. Natl. Acad. Sci. U.S.A. 96, 1342713431. Wang, Y.Q., Guo, X., Qiu, M.H., Feng, X.Y., and
van Praag, H., Schinder, A.F., Christie, B.R., Toni, N., Sun, F.Y. (2007). VEGF overexpression enhances
Palmer, T.D., and Gage, F.H. (2002). Functional striatal neurogenesis in brain of adult rat after
neurogenesis in the adult hippocampus. Nature a transient middle cerebral artery occlusion. J.
415, 10301034. Neurosci. Res. 85,7382.
Velazquez, R., Ash, J.A., Powers, B.E., Kelley, C.M., Welberg, L. (2013). Neurogenesis:Abombshell of a
Strawderman, M., Luscher, Z.I., Ginsberg, S.D., finding. Nat. Rev. Neurosci. 14,522.
Mufson, E.J., and Strupp, B.J. (2013). Maternal Wen, P.H., Hof, P.R., Chen, X., Gluck, K., Austin, G.,
choline supplementation improves spatial learn- Younkin, S.G., Younkin, L.H., DeGasperi, R.,
ing and adult hippocampal neurogenesis in Gama Sosa, M.A., Robakis, N.K., Haroutunian,
the Ts65Dn mouse model of Down syndrome. V., and Elder, G.A. (2004). The presenilin-1 famil-
Neurobiol. Dis. 58, 92101. ial Alzheimer disease mutant P117L impairs
Venna, V.R., Xu, Y., Doran, S.J., Patrizz, A., and neurogenesis in the hippocampus of adult mice.
McCullough, L.D. (2014). Social interaction plays Exp. Neurol. 188, 224237.
a critical role in neurogenesis and recovery after Yamashita, T., Ninomiya, M., Hernandez Acosta, P.,
stroke. Transl. Psychiatry. 4,e351. Garcia-Verdugo, J.M., Sunabori, T., Sakaguchi,
Verret, L., Jankowsky, J.L., Xu, G.M., Borchelt, D.R., M., Adachi, K., Kojima, T., Hirota, Y., Kawase, T.,
and Rampon, C. (2007). Alzheimers-type amy- et al. (2006). Subventricular zone-derived neu-
loidosis in transgenic mice impairs survival of roblasts migrate and differentiate into mature
newborn neurons derived from adult hippocam- neurons in the post-stroke adult striatum. J.
pal neurogenesis. J. Neurosci. 27, 67716780. Neurosci. 26, 66276636.
Walker, A.K., Rivera, P.D., Wang, Q., Chuang, J.C., Yang, B., Migliati, E., Parsha, K., Schaar, K., Xi,
Tran, S., Osborne-Lawrence, S., Estill, S.J., X., Aronowski, J., and Savitz, S.I. (2013).
Starwalt, R., Huntington, P., Morlock, L., et al. Intra-arterial delivery is not superior to intrave-
(2014) The P7C3 class of neuroprotective com- nous delivery of autologous bone marrow mono-
pounds exerts antidepressant efficacy in mice nuclear cells in acute ischemic stroke. Stroke 44,
by increasing hippocampal neurogenesis. Mol. 34633472.
Psychiatry 20, 500508. Yao, J., Mu, Y., and Gage, F.H. (2012). Neural stem
Walter, C., Murphy, B.L., Pun, R.Y., Spieles- cells:Mechanisms and modeling. Protein Cell. 3,
Engemann, A.L., and Danzer, S.C. (2007). 251261.
228 Part II:Homeostatic Control

Yassa, M.A., and Stark, C.E. (2011). Pattern separa- contribute to stroke-induced neurogenesis
tion in the hippocampus. Trends Neurosci. 34, and neuroblast migration toward the infarct
515525. boundary in adult rats. J. Cereb. Blood Flow
Yuan, T.F., and Arias-Carrion, O. (2011). Adult neu- Metab. 24, 441448.
rogenesis in the hypothalamus:Evidence, func- Zhang, R.L., Chopp, M., Roberts, C., Liu, X., Wei,
tions, and implications. CNS Neurol. Disord. M., Nejad-Davarani, S.P., Wang, X., and Zhang,
Drug Targets 10, 433439. Z.G. (2014). Stroke increases neural stem cells
Zhang, C., McNeil, E., Dressler, L., and Siman, R. and angiogenesis in the neurogenic niche of the
(2007). Long-lasting impairment in hippocam- adult mouse. PloS One 9:e113972.
pal neurogenesis associated with amyloid depo- Zhao, C., Teng, E.M., Summers, R.G. Jr., Ming, G.L.,
sition in a knock-in mouse model of familial and Gage, F.H. (2006). Distinct morphological
Alzheimers disease. Exp. Neurol. 204,7787. stages of dentate granule neuron maturation in the
Zhang, C.L., Zou, Y., He, W., Gage, F.H., and Evans, adult mouse hippocampus. J. Neurosci. 26,311.
R.M. (2008). A role for adult TLX-positive neural Zhao, M., Momma, S., Delfani, K., Carlen, M.,
stem cells in learning and behaviour. Nature 451, Cassidy, R.M., Johansson, C.B., Brismar, H.,
10041007. Shupliakov, O., Frisen, J., and Janson, A.M.
Zhang, L., Hu, X., Luo, J., Li, L., Chen, X., Huang, R., and (2003). Evidence for neurogenesis in the adult
Pei, Z. (2013). Physical exercise improves functional mammalian substantia nigra. Proc. Natl. Acad.
recovery through mitigation of autophagy, attenua- Sci. U.S.A. 100, 79257930.
tion of apoptosis and enhancement of neurogenesis Zhu, D.Y., Lau, L., Liu, S.H., Wei, J.S., and Lu, Y.M. (2004).
after MCAO in rats. BMC Neurosci. 14,46. Activation of cAMP-response-element-binding
Zhang, R., Zhang, Z., Wang, L., Wang, Y., Gousev, protein (CREB) after focal cerebral ischemia stim-
A., Zhang, L., Ho, K.L., Morshead, C., and ulates neurogenesis in the adult dentate gyrus.
Chopp, M. (2004). Activated neural stem cells Proc. Natl. Acad. Sci. U.S.A. 101, 94539457.
PARTIII

Homeostatic Manipulators

Preventative and Restorative Opportunities


14
Systems (Network) Pharmacology forBrain
Functionality Restoration
DORU GEORG M ARGINE A NU

OVERVIEW doped by various psychostimulant drugs, whose


The Neurologic and psychiatric diseases etiolo- use became common for many healthy people,
gies are as yet poorly understood; medications though it raises serious safety concerns. On the
merely treat the symptoms, and the prevail- other hand, nootropic drugs were originally
ing reductionist focus fails to grasp the brains meant to activate/restore deficient higher brain
complexity and the intricacy of its dysfunctions. activity while not inducing toxic phenomena.
Reductionism has led to some chief successes The nootropic concept, tailored on the unusual
in molecular biology, but it fails to account for pharmacology of piracetam, remained largely
integrative biological functions that are emer- overlooked due to the lack of a straightforward
gent properties occurring only at the organis- drug-receptor mechanism, while a multitude
mic level. Also, the reductionist goal of single of weak interactions have been documented for
target-selective drugs, instead of substantiat- piracetam-like nootropics. Though at odds with
ing an efficient drug design, caused a persistent the analytical quest for a definite molecular
decrease in new drug discoveries, particularly interaction accounting for the effect, the original
for brain diseases. In contrast to target-selective nootropic concept does fit the holistic-oriented
pharmacology, emergent systems (or network) framework of network pharmacology.
pharmacology holds that drug action results Network-oriented drug discovery requires
from multiple interactions with manifold targets optimization of multiple positive and negative
in a complex network underlying therapeutic influences on several targets while balancing
and collateral effects. This chapter exposes the drug-like properties and controlling unwanted
necessity of holistic-type systems biologybased effectsan endeavor by far more complex
approaches that embrace the intrinsic complexity than the much simpler single-target strategy.
of brain functional disturbances. Nonetheless, significant bioinformatics and
Brain functionality disorders are complex dis- chemoinformatics resources and computational
eases whose multifactorial and polygenic nature methods, briefly outlined in this chapter, have
do suggest a disturbance in networks of interac- already been put forward and are progressing
tions, and all major neuropsychiatric drugs do rapidly.
in fact interact with multiple molecular targets.
The multimechanistic feature derives from the Introduction:Brain Functionality
development of those drugs based on phenotypic and Its Complexity
observations in integrated in vivo systems, rather Brain function to ensure an organisms inte-
than target-oriented rational drug discovery. But gration into its milieu and to exert centralized
systems pharmacology ultimately aims to design control over all body organs is an exquisite and
ab initio drugs that would appropriately act on demanding communicational activity that led
the ensemble of cell components disturbed in a to emergence of the most complex system in the
given pathology, normalizing their network. universethe human brainwhose complexity
Pharmacologic support of cognitive functions is unanimously acknowledged and increasingly
impaired in dementia-type degenerative disor- approached with concepts from mathematics,
ders is by now hardly capable of any cognitive physics, and computer science (Sporns, 2003;
restoration. But normal cognition can be actually Singer, 2007). Brain functionality ultimately
232 Part III:Homeostatic Manipulators

relies on the cellular property of excitability, suggestion that principles derived from particu-
which is truly prototypal for neurons and sub- lar cases might be applied to make general pre-
tle but present also in some glia (Volterra and dictions later evolved into the inductive method.
Meldolesi, 2005), and on highly structured inter- Descartes proposal that each difficult problem
cellular (interneuronal, glia-neuronal, and inter- should be divided into as many simpler parts
glial) communication. The astounding structural necessary for its resolution (the analysis rule) and
intricacy of the human brain is accompanied by that the effort to understand should start with the
an exceptionally intense metabolism, represent- simplest and easiest to know objects, gradually
ing a drain of over 20% of resting consumption rising to the knowledge of those composed (the
on an organisms energy budget (Laughlin and synthesis rule), had a foremost and long-lasting
Sejnowski, 2003). These very features render impact on all Western science. This methodologi-
the brain subject to various types of dysfunc- cal reductionism, which posits reducing explana-
tion: degeneration, physicochemical damage, tions to the smallest possible entities as the best
excitability troubles, and intercellular com- scientific strategy, entails epistemological reduc-
munication alterations, largely deriving from tionism, which admits that the knowledge of a
disruption of the homeostatic environment scientific domain about more complex objects
needed for proper neuronal function (Boison of study can be reduced to knowledge in scien-
etal., 2013). They also entail the complexity and tific domains about simpler objects, and derives
heterogeneity of neurological (e.g., Tejada etal., from ontological reductionism, which consid-
2013) and psychiatric (Yang and Tsai, 2013; ers reality as composed of a minimum number
Takahashi, 2013)diseases, as yet poorly mastered. of kinds of simplest entities (Brigandt and Love,
Consequently, the etiology of brain-specific 2012; Fang and Casadevall, 2011). Descartes
pathologies is but insufficiently understood, with reductionist thinking, relying on his belief that
the corollary that brain disease medications are God made the world as a clockwork mechanism,
mostly symptomatic and hardly curative. triumphed in the classical mechanics of Newton
This chapter highlights the idea that, for and culminated in the mathematically elegant
advancing the means to restore disturbed brain rational mechanics developed in the late eight-
functionality, neuropsychopharmacology should eenth and early nineteenth centuries. Its full suc-
transcend the currently prevailing reduction- cess to explain the movement of all bodies, from
ist focus and develop integrative, holistic-type an apple falling from the tree to manmade vehi-
approaches aimed at embracing the intrinsic cles to celestial bodies, was so impressive that it
complexity of brain functional disturbances. initiated the search for the mechanism of every
Such a conceptual move is brought about by the phenomenon as the ultimate goal of all sciences.
fairly recent ascent of systems biology (SB; Kitano, Descartes was so fascinated by the exquisite
2002; Sauer et al., 2007) that goes beyond the clock-like automata of his time that he believed
mere analytic (reductionist) focus on the molecu- that animals function as machines, albeit more
lar components, taking into account the complex advanced, since being made by the hands of God
and dynamic interactions within the organisms, (Discours de la mthode, part5). The accumula-
considered in their environment. Consequently, tion of discoveries on cellular organization of
the pathology is seen to reflect alterations of the animals and plants culminated in the middle of
network of interactions that underlie organismic the 19th century in Theodor Schwanns cellular
functions, and the corresponding systems (or net- theory, so that it is aptly said that biology gradu-
work) pharmacology is aimed at renormalizing ated from natural history by holding the hand
that network. This chapter explores the coming of reductionism (Bose, 2013). Furthermore,
out, the current status, and the prospects of neu- as every organism is made up of molecules, for
ropsychopharmacology in this conceptualshift. understanding phenomena at the organism level
biochemistry focused on biomolecules and on
The Rise ofSystems Biology biochemical reactions. The climax of the reduc-
and Integrative Approaches tionist approach was reached in the third quarter
inNeuroscience of the 20th century by the brilliant achievements
The advent of modern science essentially relies on of molecular biology. These seemed to substanti-
Francis Bacons Novum Organon (1620) and Ren ate the belief that all biology might be explained
Descartes Discours de la mthode (1637) that laid in terms of chemistry and physics, as claimed
the bases of Western scientific inquiry. Bacons by Francis Crick (1966) in a provocative book.
Systems (Network) Pharmacology for Brain Functionality Restoration 233

Along those lines, an extreme reductionist view detrimental effect of de facto forgetting the syn-
is the belief that consciousness and mental states thesis rule, equally stated in Descartes Method,
can be reduced to the chemical reactions occur- while largely relying only on his analysis rule, is
ring in the brain (Van Regenmortel,2004). particularly obvious in neuroscience. The insuf-
Notwithstanding the wealth of knowledge ficiency of the current understanding is evident
about individual cellular components and their not only for the most challenging brain activity of
roles generated by the reductionist (analytic) giving rise to consciousness and thought (Singer,
approach, it is obvious that no biological func- 2007)but even for apparently much simpler and
tion can be attributed to an individual mole- long-studied subjects, such as the mechanism of
cule, because all the functions of organisms are general anesthesia (see later discussion).
emergent properties that occur only at a given The necessity that neuroscience transcends
level of complexity, while not existing at lower reductionism was strongly enhanced by the
levels (OConnor and Wong, 2012). This sim- awareness of the complexity of brain pathologies
ple conclusion was imposed, among others, by causality. Apart from the fairly rare channelo-
the unexpected results or just the lack of effect pathies associated with a single mutated gene,
of many knockout experiments performed to genetic studies have indicated that the etiologies
reveal the functional role of a single gene (for of common neurological (e.g. epilepsy) and psy-
discussion, see Hopkins, 2008; Mazzocchi, chiatric (e.g. bipolar disorder and schizophrenia)
2012). It was further reinforced when, following diseases involve many genes interacting mutu-
the complete mapping of the human genome, ally and with environmental risk factors, each
it appeared that the number of protein-coding of which contributes only a small risk on its
genes is not larger than 21,000 (Pennisi, 2012), own (Berkovic et al., 2006; Huang and Mucke,
which is less than four times the number of genes 2012; Szczepankiewicz, 2013; Jiang et al., 2013).
of the unicellular eukaryote Saccharomyces cere- A recent example is the study of the genetic
visiae (Goffeau et al., 1996), though the human architecture of Tourette syndrome and obses-
genome has to encode for ~100 trillion cells of sive compulsive disorder (Davis etal., 2013)that
many different types in the human body. This provides evidence that the two pathologies are
evidently suggests that the observed complex- highly heritable and polygenic, and that a signifi-
ity is not encoded as such in the relatively few cant majority of the heritability of both disorders
genes, but it arises from the large set of mutual is associated with single nucleotide polymor-
interactions that they are capable of generating. phism variants. Previous attempts to identify
Thus the richness of information derives not the individual genetic risk factors involved in
only from genes themselvesas it was the ear- these two neuropsychiatric disorders have been
lier tenet of molecular biologybut also from mostly unsuccessful, likely because there are
the interaction between genes and between their many genetic variations scattered throughout the
respective products (see discussion in Ahn etal., genome, each contributing only a small amount
2006; Sinha et al., 2009). Beyond the informa- to the overall risk. The complexity of the etiology
tion stored in the nucleotide sequence, epigenetic of brain dysfunction is illustrated in Figure 14.1
mechanismschiefly DNA methylation and his- for the case of epileptic syndromes.
tone modifications but also dynamic reorganiza- Epigenetic mechanisms and gene-
tion of nuclear architecture and RNA and DNA environmental interactions mediated by epige-
editinglargely influence the way genes are netic mechanisms have been recognized as fun-
expressed without altering the underlying DNA damental causes of different neurodevelopmental
sequence. Most notably, epigenetic mechanisms disorders, including autism spectrum disorders.
account for cellular differentiation during mor- Likewise, there is strong evidence that neurode-
phogenesis by selective inhibition/activation of generative diseases are largely mediated by aber-
various genes (Reik,2007). rant epigenetic mechanisms, which have been
In neuroscience, while the Cartesian ana- also identified in diverse neuropsychiatric condi-
lytic approach could ensure an in-depth current tions (for a review see Mehler, 2008). Moreover,
knowledge of how individual brain cells function, the complexity of brain pathologies causality
it was much less successful in deciphering their is enhanced by the confirmed participation in
cooperation in large ensembles and could not various neurodevelopmental diseases associated
further our understanding of higher brain activi- with epilepsy, autism disorders, and intellectual
ties that integrate a huge number of neurons. The disability of somatic mutations not present in
234 Part III:Homeostatic Manipulators

GeneticEnvironmental Head trauma

Gene 1 risk interaction


Perinatal
injury
Epileptogenic mutations and variants

Epileptogenic acquired factors


Gene 2 Monogenic
(chanelopathies) Stroke

Structural/
metabolic
Gene 3 Developmental

Genetic
lesions
Epilepsy

Tumor

Polygenic (complex
Gene N1 inheritance) Infectious
lesions

Gene N Previous
seizures

FIGURE14.1: Convergence of multiple genetic and environmental factors in the etiology of epilepsy. Single gene
mutations, mostly in genes coding channel proteins, underlie only rare epileptic syndromes, while multiple gene
variants contribute to frequent polygenic epilepsies. The neurobiological spectrum of epilepsies appears fairly
continuous from a predominantly genetic to a predominantly acquired causation.
Slightly modified from Margineanu, 2012; reproduced with permission

all body cells, as they arise during embryonic function and, consequently, considers that a dis-
development and are undetectable in the parents ease expresses a disturbed network of interac-
(Poduri etal.,2013). tions rather than alteration of a critical molecular
Integrative approaches in biology date from component. Specifically, multi-omics and power-
19th-century embryology, but the origins of a ful bioinformatics computational tools make the
holistic approach of animal description can be systemic approach to a better understanding/
traced back to Aristotle (384322 bc). He stated treatment of nervous system diseases promising
in his treatise Metaphysics that the whole is (Noorbakhsh etal., 2009; Villoslada etal., 2009;
more than the sum of its parts, thus affirming Hu etal.,2013).
that a whole has emergent properties that can be An extra-scientific factor that forcefully
neither reduced nor deduced from the qualities pushed amendment of the reductionist
of its individual parts (Bothwell, 2006; Brigandt approaches is the lowered yield of drug discovery
and Love, 2012). This view, now held in a vari- in the past two decades (Pammolli et al., 2011),
ety of scientific fields (Mazzocchi, 2008; Brigandt especially obvious with respect to the discovery
and Love, 2012), is at the core of the SB that origi- of novel drugs for brain diseases (Miller, 2010;
nates in the general systems theory that L. von Arrowsmith, 2011). The productivity decrease in
Bertalanffy developed (1950, 1968)in the 1950s. pharmaceutical research and development is sig-
The awareness of the limits of reductionism in nificantly concomitant with the replacement of
biology became so overt in the 1990s that it led to the traditional phenotypic-based drug discovery
symposia and monographs specifically devoted with a target-based one (Sams-Dodd, 2005). This
to that subject (e.g., Bock and Goode, 1998). latter relies on the simplifying idea that a single
Holistic approaches to biological data received mutated gene, giving rise to one altered protein
mainstream attention (Ideker etal., 2001; Kitano, causes a disease that should be treated by a selec-
2002; Barabsi and Oltvai, 2004) after the pro- tive drug targeted at that protein. In phenotypic
gress of omics technologies permitted the simul- screening in vivo, the compounds are selected on
taneous probing of thousands of genes and their the basis of their ability to produce desired meas-
products, thus allowing SB to reach the molecular urable changes in the physiopathological states
level of description. SB deals with the networks of animals. In target-based screening, however,
of interactions that underlie each biological the whole physiological context is replaced by
Systems (Network) Pharmacology for Brain Functionality Restoration 235

16.0 14.3

% of Sys. Pharmacol. entries


14.0
12.0
10.0
7.3
8.0
6.0
3.7
4.0 2.7
1.3 1.9
2.0 0.7 0.7 0.7 0.2 0.2
0.0

e
se

e d in

ke

ia

r d nt

er

r
as

ps

as

e
io

en
Pa

rd
a

nc
ro

ss

m
ise
ise

se
ile

iso
r
St

Ca
r
e

h
di

Bi pai
Ep
ld

pr

p
zo
ric

De
a

im
iv

la
hi
ic

at
at

po
g

Sc

ve
hi
r
lo

ne

iti
yc
o
ur

ge

gn
Ps
Ne

de

Co
u ro
Ne

FIGURE 14.2: The percentage of the total number of papers indexed in the PubMed databank under the med-
ical subject heading (MeSH) systems pharmacology (Sys. Pharmacol.) that also include as MeSH as various
brain-specific pathologies. The percentage of systems pharmacology papers referring to cancer is given for com-
parison. The respective percentage value is given at the top of each column. Search completed on November 10,
2013.

the molecular interaction with only one target but the scientific interestassessed by the num-
(Pujol etal., 2010), the compounds being chosen ber of papers devoted to these topicsis already
to optimize the affinity and selectivity toward a noteworthy (Figure 14.2), justifying optimistic
single protein, whose involvement in a pathologi- expectations.
cal process is deemed critical.
The reductive philosophy of target-based The Necessary Track ofMultitargets
drug discovery was expected to substantiate an inNeuropsychopharmacology
efficient rational drug design, but it turned bit- Pharmacologys truly great idea of drugreceptor
terly illusory (Sams-Dodd, 2005, 2013). The poor interaction (Rang, 2006) suggested the meta-
productivity in the past decades of the pharma- phoric concept of a magic bullet drug that might
ceutical industry lured by the target-centered treat a disease upon selectively hitting a decisive
approach favors a shift toward the SB-relying molecular target. Put forward by Paul Ehrlich as
systems pharmacology (Hopkins, 2008; Berger a logical objective of antimicrobial therapy, that
and Iyengar, 2009; Boran and Iyengar, 2010)that metaphor largely seduced the ensemble of drug
replaces drug discovery on realistic biological discovery practitioners by the hope of a concep-
grounds (Butcher, 2005). Systems pharmacol- tually simple rational drug discovery. However,
ogy has a view of drug action basically different for complex systemic diseasesas is the case
from traditional pharmacology. Traditionally, with all disturbances of brain functionalitythe
a drug is meant to interact specifically with a intrinsic multifactorial and polygenic nature of
given therapeutic target that mediates the ther- the pathology turned the magic bullet into a lure.
apeutic effect and only accessorily some side In contrast, the SB view of diseases as arising
effects. Any other putative interaction is con- from the disturbance of networks of interactions
sidered off-target and mainly carrying the risk voids of sense the search for clean drugs that
of unwanted effects. On the other hand, systems would specifically interact with unique molecu-
pharmacology conceives drug action as result- lar targets.
ing from multiple interactions, possibly of weak The realistic alternative appears to be a
strength, with manifold targets pertaining to a multitarget pharmacology (or polypharmacol-
complex network that underlies both therapeu- ogy) by compounds that interact in a concerted
tic and collateraloccasionally adverseeffects. manner with multiple targets (Keith etal., 2005;
Systems pharmacology approaches aimed to dis- Hopkins, 2008). Drugs nonselective for a sin-
cover novel drugs for treating brain pathologies gle target but that selectively interact with sev-
are too recent for already-launched medicines, eral targets involved in a given pathology have
236 Part III:Homeostatic Manipulators

been pertinently dubbed by Roth etal. (2004) as earlier. In a meeting in 1990 addressing the
magic shotguns. An overview of the antipsy- question of whether high selectivity is always
chotic drugs used in the treatment of schizophre- beneficial (Fredholm and Abbott, 1990), Arvid
nia and similar severe mental disorders, recorded Carlsson highlighted the merit of correcting the
in DrugBank, revealed for most of them multi- imbalance between receptors rather than a single
ple targets, peaking at no less than 26 targets for deficit. Likewise, in the circumscribed domain
clozapine and quetiapine and 25 for olanzapine of AEDs, at the same time that the fascination
and aripiprazole (Sun et al., 2012). Remarkably, with the rational drug design was generalized,
it was, on the other hand, highlighted that the Lscher and Schmidt (1994) noted that, for AED
enriched pharmacology imparted on clozap- development, the target-centered rational
ine by the multiplicity of its molecular targets, strategies did not show any superiority over the
rather than the lack of extra-pyramidal side older screening methods in animal models, and
effects, is the origin of the clinical superiority of they emphasized that the most clinically useful
this second-generation atypical antipsychotic AEDs are multimechanistic. However, before the
(Grnder etal.,2009). recent rise of the theoretical underpinning of SB,
The label magic shotgun, coined by Roth such lucid observations and opinions, though
etal. (2004) upon evaluating the mechanisms of expressed by high-ranking scientists, could not
the drugs for mood disorders and schizophrenia, deter the reductionist trend in neuropharmacol-
is actually fit for most (if not all) of the drugs ogy, largely because a target-centered drug dis-
effective in treating psychiatric and neurologi- covery comforts the marketing-driven leaning of
cal troubles. Thus, the antipsychotic drugs have a pharmaceutical industry toward clean drugs,
wide pharmacological promiscuity, as they mod- deemed to be advantageous (Margineanu,2014).
ulate (at nMM concentrations) the function of Most clinically effective drugs currently
quite numerous ion channels, both ligand-gated used for treating neurological and psychiatric
and voltage-gated. Also the antidementia drugs troubles interact with multiple targets, because
have been reported to interact with multiple ion these drugs have been developed starting from
channels (Bianchi, 2010). Likewise, the fact that phenotypic observations, largely serendipitous,
all the reference antiepileptic drugs (AEDs) and in integrated in vivo systems, rather than by
the more clinically useful new generation AEDs target-oriented rational drug discovery. The vast
exert their hypoexcitatory effects by interacting ensemble of neuropsychiatric drugs serendipi-
with multiple ion channels was repeatedly high- tously discovered that are highly multimecha-
lighted (Bianchi etal., 2009; Margineanu, 2012). nistic is illustrated in Figure 14.3 by the case of
Interestingly, functional modulation of multi- valproate, which is truly emblematical, though
ple ion channels has been clearly attested even not singular, for the multiplicity of biochemi-
for the AED levetiracetam (Margineanu and cal and cellular effects exerted by a fairly sim-
Klitgaard, 2002), which has a well-defined bind- ple (MW=144.2) molecule. The systemic-type
ing site, the synaptic vesicle protein SV2A (Lynch phenotypic screening in CNS drug discovery
etal., 2004), which is not a channel protein. remarkably resisted the reductionist thinking
The pharmacological promiscuity of psychi- that underlies target pharmacology, as proven
atric, anticonvulsant, and anesthetic drugs that by an analysis of the discovery approaches that
modulate multiple molecular targets, particu- generated the new molecular entities approved
larly the ligand-gated ion channels GABA A, Gly, by the US Food and Drug Administration (FDA)
nACh, and 5-HT3 of the cys-loop family of recep- in the decade from 1999 to 2008 (Swinney and
tors, appears so marked that it was hypothesized Anthony, 2011). Surprisingly, given the main
that it effectively contributes to clinical efficacy, focus on target-based approaches, phenotypic
so that the promiscuity maps of current neurop- screening led to the discovery of more first-in-
harmaceuticals might provide templates against class small-molecule drugs than target-based
which to screen candidate compounds (Bianchi approaches (28 vs. 17 drugs, respectively).
and Botzolakis,2010). Moreover, an assessment of the drug targets of
Systems pharmacology provides the ration- the new molecular entities approved by FDA
ale to using multimechanistic drugs, but the in the decade 2000 to 2009 revealed a trend
idea that absolute selectivity should not be a toward multitarget drugs, with the average
sought-after objective for drugs aimed to treat number of targets per approved drug rising to
central nervous system (CNS) pathologies that 2.5 from a previously estimated value of 1.8
involve complex changes has been put forward (Lu etal.,2012).
Systems (Network) Pharmacology for Brain Functionality Restoration 237

/NaF; P /T metabolism (Keith et al., 2005), to unforeseen


drugdrug interactions, as well as to possible
Brain
cCMP
multiplied side effects. Accordingly, the focus is
GABA
turnover on the design by knowledge-based modification
O
GHB
of drug structure to achieve multifunctionality of
HO
VPA
formation single-drug molecules that target multiple etio-
& release
... ... logical targets specific to a particular neuropsy-
!!!
Brain chiatric disorder (Van der Schyf etal., 2006; see
Asp later discussion).
HDAC1
Concerning the reality that the most effi-
Brain
Gly; Taurine cient CNS drugs are multimechanistic, one
Brain
5-HT; DA
should notice that those drug molecules pos-
sess that clinically beneficial feature simply by
FIGURE14.3: Apart of the dozens of biochemical and chance, with the merit of the drug discovery
cellular effects of valproic acid (or valproate) that likely process being only of having duly selected them
contribute to the large-spectrum antiepileptic activity by informed phenotypic screening and serendip-
(Lscher, 2002), mood-stabilizing and antimigraine ity and occasionally having improved them by
activity (Rosenberg, 2007; Chiu etal., 2013), potential ADME (absorption, distribution, metabolism,
anticancer activity, and toxicity (Chateauvieux et al., and excretion)-guided medicinal chemistry and
2010)of this drug. The actions are designed by either: efficacy criteria. On the other hand, the ultimate
(activation), | (inhibition), or (modulation). Red letters objective of systems pharmacology is, as men-
on yellow background indicate relevance for anticon- tioned, the ab initio design of drugs that will
vulsant activity and brown letters on green background appropriately act on the ensemble of cell compo-
indicate potential anticancer relevance. Asp = aspar- nents disturbed in a given pathology, normaliz-
tic acid, Cgmp= cyclic guanosine monophosphate, ing their network.
DA=dopamine, GABA=gamma-aminobutyric acid,
GHB = gamma-hydroxybutyric acid, Gly =glycine, Might Systems Pharmacology
HDAC1 = histone deacetylase 1, 5-HT = serotonin, Rescue theControversial Nootropic
INa F = fast sodium current, INa P = lasting (permanent) Drug Concept?
sodium current, IT = low-voltage activated (transient) Cognitive enhancement is an unquestionable
calcium current. therapeutic goal in all pathology-related treat-
ment, whether or not it involves age-linked cogni-
tive impairment. At the same time, an enhanced
Apart from the multitargeted drugs, poly- and longer lasting high cognitive activity is that
pharmacology is also performed by multidrug typical human aspiration that begets education
therapies. This is in no way a novelty in the treat- and culture and whose appeal derives from both
ment of brain diseases, since polypharmacy is a the subjective self-confidence and the competitive
long-standing and widespread clinical practice social advantage imparted by advanced mental
both in psychiatry (Correll and Gallego, 2012; capability. The marked increase in life expectancy
Kukreja etal., 2013)and neurology (e.g., French of the population in developed countries is par-
and Faught, 2009; Brodie and Sills, 2011). The aim alleled by the increased prevalence of cognitive
of any rational polytherapy is to find the com- decline and dementia, causing cognitive troubles
bination of drugs with complementary simple to emerge as a major health threat of this cen-
mechanisms that will obtain additive, preferably tury (Bishop etal., 2010). Cognitive impairment
synergistic effects with infra-additive toxicity. is emblematic for dementia-associated neurode-
But the sobering reality is that by now the choice generative conditions, but it is also prominent
of drug combinations remains largely empirical, in psychiatric disorders (Millan et al., 2012),
until a consistent systemic characterization of the with cognitive enhancement standing a major
addressed pathology can substantiate a proper challenge to their treatment (Insel et al., 2013).
mechanistic complementarity and an integrated Pharmacological therapeutic approaches aimed
view of the multitude of potential molecular to support cognitive functions pathologically
interactions can allow mastering the toxicity. affected are quite numerous (Froestl etal., 2012;
Regardless, combining several drug molecules Froestl et al., 2013a, 2013b), but their success
raises many risks due to the potentially different notoriously lags behind the desire. Apart from
degrees of bioavailability, pharmacokinetics, and the restoration of impaired cognition, normal
238 Part III:Homeostatic Manipulators

neurocognitive function too can be doped by indeed such a low acute toxicity in all animal
various psychostimulant drugs (Lanni et al., species tested in preclinical studies that no LD50
2008). The use of psychostimulants such as meth- could be determined. Likewise, it appeared very
ylphenidate and modafinil actually became a well tolerated at therapeutic doses of the order
common fact of life for many healthy people who of several grams per day in several thousands of
have to cope with demanding mental activities patients included in controlled clinical studies
(Sahakian and Morein-Zamir, 2007), including (Winblad, 2005). It is noteworthy that both the
professional scientists (Maher, 2008). This reality lack of toxicity and the unusually high dosages
raises serious ethical concerns (Farah etal., 2004; necessary to achieve therapeutic effects imply
Cakic, 2009; Hyman etal., 2013)related not only very weak affinity of piracetam for the molecular
to social justice but chiefly to the safety of using components in the cells of the receiving organ-
strong promoters of attention and wakefulness. ism. Illustrative for this is the fact that piracetam
On the other hand, 40 years ago, Corneliu almost completely lacks affinity (pK=4.1) for the
Giurgea, from the Belgian pharmaceutical firm SV2A proteinthe binding site of the pyrroli-
UCB, put forward the concept of nootropic dine AEDs levetiracetam (pK=6.1)and brivar-
drugs (Giurgea, 1972), tailored to account for acetam (pK=7.1)in spite of the close chemical
the unusual pharmacology of piracetam, a pyr- kinship of the three compounds.
rolidine derivative synthesized at UCB in 1964 The mechanisms by which piracetam exerts
and marketed in 1971. That Pavlovian neuro- its actions remained obscure for a long time
physiologist coined the term nootropic (join- (Giurgea, 1982) and they still appear multifari-
ing the Greek words for mind and toward) ous, as this small molecule (MW = 142.16) has
to designate drugs that would directly activate been reported to influence cholinergic, sero-
the integrative activities of the brain, restoring toninergic, noradrenergic, and glutamatergic
deficient higher brain activity and opposing systems (for a review see Winblad, 2005). But
brain impairment by agents such as barbiturates none of its effects results from a direct agonism
and scopolamine while not inducing any toxic or antagonism, with piracetam showing no
phenomenon. Since his definition was simply noticeable affinity for the receptors of those neu-
phenomenological, did not make any mechanis- rotransmitters, in contrast to the usual psycho-
tic reference, and appeared to neglect the core tropic drugs. More recently, Ahmed and Oswald
idea of drugreceptor interaction, the nootropic (2010) revealed a weak positive allosteric modu-
concept hardly found any place in the major trea- lation by piracetam of the AMPA receptors. Such
tises of pharmacology and remained controver- a weak modulation might conceivably relate to
sial. This apparent neglect is strongly contrasted the repeatedly reported piracetam-induced
by the numerous early attempts to synthesize changes in physical properties of cellular mem-
other piracetam-like nootropics, as well as by the branes (Mller et al., 1997; Eckert et al., 1999;
steadily rising scientific interest and widespread Mingeot-Leclercq et al., 2003). A recent analy-
popularity in the lay public. Thus, even 20years sis of piracetam modulation of dopaminergic,
ago more than 1,650 piracetam-like compounds glutamatergic, or cholinergic brain systems
had been synthesized worldwide in view of the pinpointed the interaction with membrane
development of nootropics, many of them con- phospholipids as the most plausible mechanism
taining the same 2-oxo-1-pyrrolidine acetic acid explaining piracetam action on the activities
(racetam) substructure (Gouliaev and Senning, of neurotransmitter systems (Slais et al., 2012).
1994). Also, currently PubMed indicates 27,754 Arecent neurochemical study of the prevention
entries under the term nootropic (as of October 17, by piracetam of scopolamine-induced memory
2013), with 910 entries in2012. impairment (Marisco et al., 2013) suggests that
Presently, the term nootropic is taken piracetam-induced improvement of memory
as equivalent to cognitive enhancer (see, e.g., is associated with protection against oxidative
Lanni etal., 2008; Froestl etal., 2012)and to the stress. Indeed, piracetam was shown to improve
mass-medias catchy term smart drug. However, mitochondrial dysfunction associated with oxi-
the equivalence is but loose (Margineanu, 2011), dative stress and/or aging (Keil et al., 2006),
as it leaves aside the requirement of lack of toxic- with mitochondrial stabilization and protection
ity that was explicit in the original definition of providing a likely justification of the putative
nootropic. Piracetam, which is unanimously beneficial effect of piracetam on the cognition
acknowledged as the forerunner of nootropic of elderly patients. Moreover, a restoration by
drugs (Shorvon, 2001; Froestl et al., 2012), has piracetam of age-altered or pathology-altered
Systems (Network) Pharmacology for Brain Functionality Restoration 239

fluidity of plasma membranes of various cells, pleiotropic effects and that, for its action, high
not only of neurons, via a nonspecific interac- clinically relevant tissue concentrations (~10 mM)
tion with cell membrane phospholipids could are required makes it unlikely that any single
account for the reported effects of piracetam molecular mechanism, or its corresponding tar-
beyond the cognitive disorders, in a variety of get, might explain all the pharmacology of that
pathologies from vertigo to myoclonus to stroke drug (Paul,2006).
and even sickle cell anemia (see Winblad, 2005). The explanatory shift in the case of inha-
Furthermore, the restoring effect of piracetam lational anesthetics and alcohol from a purely
on the drug-impaired glucose uptake in eryth- nonspecific interaction with membrane lipids
rocytes (Naftalin etal., 2004)might also derive to a primary interaction with various types of
from its effects on membrane fluidity. membrane proteins makes such an evolution
Piracetams lack of specificity and its putative of the mechanistic understanding of the effects
fluidizing effect on membranes remind us of the of piracetam and chemically related nootrop-
classical case of inhalational general anesthetics, ics conceivable as well. This is indeed suggested
for which a membrane lipid-located action, as sug- by the already-mentioned allosteric interaction
gested by the Meyer-Overton linear correlation with AMPA receptors and by a recent report that
of anesthetic potency with lipophilicity, was long piracetam induces in rat brain synaptosomes
accepted as the basic mechanism. More recently, a plasma membrane depolarization sensitive
deviations from that rule (Cantor, 2001) led to to GABA receptors inhibitors and to AMPA/
a referral to the lateral pressure of lipids on ion kainate receptor inhibitors (Fedorovich, 2013).
channels, after which the lipid theory was largely Whether or not such an explanatory shift will
superseded in the past decade by the alternative proceed with identifying other weak interactions
view that anesthetics act by binding directly to with proteins, it cannot question the rescue of the
target proteins (Franks, 2006). However, not a original nootropic concept within the systemic
single but rather several targets have been identi- frame of network pharmacology. That holis-
fied as the most likely candidates, and currently tic type of concept, referring to the integrative
a multiple-receptor interaction is favored (Chau, activity of the brain, is at odds with the analyti-
2010). Apart from the inhalational general anes- cal quest of a straightforward molecular interac-
thetics, another prototypal example of molecules tion that would account for all the effects. But
exerting a multitude of effects on the organism, within the holistic-oriented conceptual frame-
in particular on brain functions, while not hav- work of network pharmacology, the multiplic-
ing any single ligandreceptor interaction that ity of weak interactions is no longer a drawback,
would account all its effects, is alcoholone of and the weakly interacting and poorly specific
the oldest and most widely (ab)used of all psycho- compoundsas in the case of most natural
active drugs. Similarly to the case of general anes- products and many brain pharmaceuticalsare
thetics, it has long been held that the effects on no longer disregarded as merely dirty drugs but
the brain of the amphiphilic molecule of ethanol instead are considered multimechanistic agents
are produced by acting primarily on membrane potentially efficient to restore disturbed func-
lipids. However, within the past two decades it tional networks. The future might show whether
appears that the primary sites of alcohol action and to what extent this possible conceptual res-
are membrane proteins (Peoples et al., 1996), a cue will have a practical outcome. On the other
view that has gained widespread acceptance (e.g., hand, if one leaves aside the piracetam-inspired
Spanagel, 2009; Komrekov and Jank, 2012). initial definition and uses the term nootropic
The primary targets of ethanol in the brain are simply as equivalent to cognitive enhancer,
multiple: NMDA, GABA A, glycine, 5-HT3, and irrespective of the requirement of lack of toxic-
nACh receptors, as well as L-type Ca 2+ channels ity, the term becomes a huge umbrella covering
and G protein-activated inwardly rectifying K+ a plethora of compounds (Froestl et al., 2012;
channels, with the initial effect on these receptors Froestl etal., 2013a, 2013b) actually or supposedly
and ion channels being followed by a second wave enhancing one or another of the multiple mental
of indirect effects of ethanol on monoamines, functionsmemory, attention, executive func-
neurosteroids, opioids, and endocannabinoids tions, perception, language, psychomotor, and/
(see Spanagel, 2009, for a review). Consequently, or executive functions (Figure 14.4)involved in
it was highlighted that the fact that a molecule the complex concept of cognition (Nehlig, 2010;
as simple as the ethanol (MW=46.07) has such Millan etal.,2012).
240 Part III:Homeostatic Manipulators

Cognition For systems pharmacology, attaining the


final goal of finding the drug(s) able to restore
Nootropic
E a certain pathology-disturbed functionality,

ns r
fu xecu

tio oto
nc ti

nc m
without entailing other dysfunctions, imposes
fu cho
tio ve
ns
y
Ps
gaining a global view of the network of molecu-
lar interactions underlying that disease state as
n

Language Memory
ptio

Me
enhancer the first landmark. This implies a large-scale

mo
ce
Per

ry
Attention integration, via bioinformatics, of diverse data
acquired from multiple levels of biological
Stimulant
complexity, from molecular-cellular (geno-
Neurotransmission
promoting wake/enhancing mood type, gene expression, DNAprotein binding,
proteinprotein interaction) to physiological
Neuroprotec- and clinical levels. The data, warehoused and
Brain metabolism & homeostasis tive/recon-
structive
annotated within a comprehensive database,
have to be further transferred to visualization
FIGURE 14.4: Cognition as a highly complex con- programs to construct the network of interac-
struct that can be impacted at several levels. Cognition tions involved in the syndrome tackled and the
emerges from multiple levels of integration of con- molecular networks that define toxicity path-
tributing factors, whose pharmacological modulation ways and subtypes of disease (Schadt et al.,
might result in a cognitive-enhancing effect. While the 2009; Arrell and Terzic, 2010; Iskar etal., 2011).
dietary supplements, nutraceuticals, and functional Large networks can be visualized with programs
foods claiming a procognitive effect (purportedly) act that implement algorithms for drawing nodes
at the basic level (e.g., via free-radicals scavenging) and and performing basic analysis of data from
the current most popular smart drugs are stimulants, any field, be it social or biological, such as, for
the original definition of a nootropic drug envisioned example, the freely downloadable and evolving
a direct action at the uppermost level. Such an action programs NetDraw (https://sites.google.com/
remained hardly conceivable as a straightforward inter- site/netdrawsoftware/home) and Pajek (http://
action with a given molecular target but becomes plau- pajek.imfm.si/doku.php?id=pajek). The charac-
sible in terms of influencing a network of interactions. terization of the molecular network undergo-
ing a disease is an ongoing process of iterative
modelexperimentation cycling, with biological
Practical Matters ofSystems measurements leading to models amenable to
Pharmacology ofCNSDrugs computational simulation, prediction, and then
A core concept of neuropsychopharmacology is experimentation for validation that potentially
that molecules influencing behavior and those that substantiates model refinement.
improve the functional state of patients afflicted In addition, to devise a disease-associated
by psychopathologies chiefly act by increasing molecular network, compounds must be char-
or reducing the effect of specific combinations of acterized by their action on that molecular net-
actions exerted by synaptic mediators. This princi- work. This characterization and annotation of
ple is also held to largely underlie the pharmacology compounds should also be iteratively refined
of some neuropathologies, though less gener- on identification of networks associated with
ally than in psychopharmacology. The principle off-target effects. Large-scale open-access data-
is fully compatible with systems pharmacology, bases of chemicals and associated bioactivity
provided it is not reduced to aiming just one neu- data, as well as the bioinformatics tools for the
rotransmitter, as is customary in drug-discovery analysis of small molecules properties in the con-
projects. But optimizing multiple positive and text of cellular networks, would become increas-
negative influences on several targets, while try- ingly popular and rapidly grow and diversify. The
ing to balance drug-like properties and control National Health Institutes databank PubChem
unwanted effects, as implied by network-oriented (http://pubchem.ncbi.nlm.nih.gov/), launched
drug discovery, appears to be a fairly intimidating in 2004 as three linked databases (Substance,
endeavor when compared with the much simpler Compound, and BioAssay) with a search tool of
single-target strategy. Nonetheless, significant structure similarity, quickly became a resource
bioinformatics and chemoinformatics resources for data on several hundreds of thousands chemi-
and computational methods have already been put cals and their bioactivities, made readily acces-
forward and are progressing rapidly. sible (Wang et al., 2009). ChemBank (http://
Systems (Network) Pharmacology for Brain Functionality Restoration 241

chembank.broadinstitute.org/) is a knowledge Nevertheless, the chemoinformatics compu-


environment created by the Chemical Biology tational tools involved in hit identification and
Program of Broad Institute (http://www.broa- lead selection by machine-learning methods that
dinstitute.org/) to guide chemists synthesizing can make reliable in silico predictions of drug
novel compounds and biologists searching for polypharmacology are evolving rapidly. Keiser
small molecules that perturb specific biologi- et al (2009) elaborated a similarity ensemble
cal pathways. It currently stores information approach that predicts numerous unanticipated
on hundreds of thousands of small molecules drug-target associations on the basis of chemical
and hundreds of biomedically relevant assays. similarities between a drug and the set of ligands
STITCH (short for Search Tool for Interactions of a target. Some of the more likely predictions,
of Chemicals; http://stitch.embl.de/) is a data- such as the antagonism of the 1 adrenergic
base of interactions between some 300,000 small receptor by the antidepressives fluoxetine and
molecules and about 2.6 million proteins from paroxetine (selective serotonin recapture inhibi-
1,133 organisms (Kuhn etal., 2012). ChEMBL is tors), have been actually confirmed experimen-
a very large open-access repository of annotated tally. Likewise, a computational implementation
compounds activity data taken from medici- based on the similarity of drugs against sets of
nal chemistry sources (https://www.ebi.ac.uk/ molecules for which binding data is available
chembldb/), which currently houses more than was able to predict with high-level precision the
5.4 million bioactivity measurements for more entire affinity profile of a representative collec-
than 1million small molecules and 5,200 protein tion of antipsychotic drugs (Vidal and Mestres,
targets (Gaulton etal., 2012). These are but a few 2010), confirming the efficiency of in silico recep-
representative examples from a larger set of data- torome screening. On the other hand, Campillos
bases of interest to systems pharmacology (Iskar etal. (2008) have shown that targets for drugs can
et al., 2011; Taboureau et al., 2012; Panagiotou be predicted not only by exploiting similarity in
and Taboureau, 2012). Their growth largely relies chemical structure or in activity across cell lines
on the fact that data from in vitro binding assays but also by using phenotypic side-effect similari-
and chemical perturbation experiments are rou- ties as an indication that two drugs might share
tinely deposited in public repositories, a trend a target. Overall, it can be taken as proven that
expected to extend in the future to multipara- chemoinformatics computational approaches are
metric readouts and clinical and histopathologi- able to predict the interaction with multiple tar-
cal parameters describing response to drugs at gets so as to potentially suggest putative indica-
tissue and organismic levels (Iskar etal.,2011). tions and side effects (Hopkins,2009).
Once the network of molecular interactions The repeatedly revealed reality that modula-
involved in a given pathology is assembled with tion of single-protein targets is not sufficient for
reasonable reliability, the next step toward a sys- obtaining the desired restoration, and that partial
tems pharmacology-derived drug is to identify modulation of several targets can be more effec-
the chemical structure(s) likely to duly impact tive than the complete inhibition of a single tar-
that network. In principle, a network can be get (Csermely etal., 2005; Morphy and Rankovic,
impacted either by hitting a hub node of it or 2007), has received widespread acceptance in the
by influencing the whole network via multiple case of neuropsychiatric dysfunctions that always
milder influences (Csermely et al., 2013). While reflect complex molecular alterations. But the
a central hit strategy seems indicated when aim- design of multitarget drugs that have balanced
ing to selectively damage the integrity of an activity at each target of interest while simulta-
infectious agent or malignant cells, the search of neously achieving a suitable pharmacokinetic
medicines for complex neurological and psychi- profile is a medicinal chemistry task by far more
atric syndromes seems likely to require a network challenging than designing a best-binding ligand
influence strategy, aiming to renormalize a faulty on the basis of the three-dimensional structure
network by mildly impacting multiple targets. At of a single-target. The successful in silico recep-
this point, however, the understanding of net- torome screening, as in the examples cited ear-
work dynamics in healthy versus diseased states lier, enhances the prospect of multitarget drug
has not yet achieved the required reliability, and, design, but it does not actually represent drug
moreover, the methods of the network influence designing. However, the design of ligands against
strategy are as yet less developed than those of profiles of multiple drug targets is clearly emerg-
the central hit strategy (Csermely etal.,2013). ing. For example, Besnard etal. (2012) developed
242 Part III:Homeostatic Manipulators

an automated algorithm for ligand design that of smaller molecular size than target-selective
adapts by multi-objective evolution, progressing compounds (Morphy and Rankovic, 2007), as
toward novel compounds with predefined mul- illustrated by the highly multimechanistic val-
titarget profiles. That algorithm was validated proate or alcohol. This might be also the case
by starting from the structure of the acetylcho- of the old prototype nootropic drug piracetam,
linesterase inhibitor donepezil (approved for a fairly small molecule showing no noticeable
cognitive enhancement in Alzheimers disease), specific binding and almost no acute toxicity
which has only minimal activity at D2 receptors, but reported to exert multiple weak effects on
and evolving with a dual objective of improving brain neurotransmitters. Incidentally, the very
D2 activity while achieving blood-brain barrier concept of a nootropic drug, deemed fuzzy in
penetration. A large proportion of the affinity conventional molecular pharmacology, appears
predictions of prospectively designed ligands uncontroversial within the frame of systems
have been confirmed experimentally, therefore pharmacology.
attesting to the feasibility of designing drug leads The beneficial feature of most efficient cur-
with multitarget profiles to achieve a desired rent CNS drugs as multimechanistic is not as yet
polypharmacology. the result of a purposeful drug design but of the
fact that those drug molecules have been duly
Concluding Remarks and Outlook selected by serendipity and phenotypic screening
All organismic functions are emergent proper- in integrated in vivo systems, rather than emerg-
ties that cannot be understood outside of their ing from target-oriented drug discovery. Due to
inherent complexity, and this reality is particu- the unequaled complexity of brain functional-
larly obvious when dealing with human brain ity, in CNS drug discovery the systemic-type
functions and their alteration. The current insuf- phenotypic screening remarkably resisted the
ficient understanding of the multifactorial etiol- lure of reductionist thinking that underlies tar-
ogy of neuropsychiatric diseases largely derives get-centered pharmacology. Beyond the domain
from the reductionist failure to grasp the intrin- of CNS drugs, the striking concomitancy of
sic complexity and heterogeneity of brain dys- a marked decline in pharmaceutical develop-
functions and has as a corollary that the existing ment of new drugs with the replacement of tra-
drugs to treat them are merely symptomatic, not ditional phenotypic-based drug discovery by a
basically restorative. target-based one led to an increasing awareness
However, the progress of omics technologies that phenotypic drug discovery is a viable strat-
and of the bioinformatics computational tools egy with the potential to enhance innovation
have rendered possible holistic approaches of (Lee etal.,2012).
the networks of interactions that underlie each The ultimate objective of systems pharmacol-
biological function, entailing a fairly recent but ogy is the ab initio design of drugs that would
rapidly mounting conceptual move from reduc- appropriately act on the pathology-disturbed
tionism toward SB. Within the systemic concep- network of molecular interactions, normaliz-
tual frame, a disease is seen to reflect alteration ing it. This task requires elaborating and ana-
of a whole network of interactions, rather than lyzing the network of molecular interactions
of a critical molecular component that might underlying that disease state, which is a complex
be selectively targeted by a magic bullettype of activity, implying a large-scale computational
drug. Consequently, the systems (or network) integration of diverse biological data and a pro-
pharmacology aims to renormalize the disturbed cess of iterative model-experimentation cycling.
network by polypharmacology via multimecha- Subsequently, compounds must be characterized
nistic drugs that would interact in a concerted by their action on that molecular network, which
manner with multiple targets or via suitable drug also involves iterative refinement. The design of
combinations. multitarget drugs with due activity at several
In fact, most clinically effective drugs that targets and suitable pharmacokinetic profiles
treat neurological and psychiatric troubles actu- involves in silico receptorome screening and
ally interact with multiple targets, being promis- adaptive chemoinformaticstools.
cuous magic shotguns rather than magic bullets, Obviously, all of ideas constitute an endeavor
even in cases when the drug might have a defined so highly challenging that it is not reasonable to
binding site. Interestingly and in some ways par- expect quick outcomes. However, the progress
adoxically, promiscuous compounds are typical so far has been quite rapid. Also, the awareness
Systems (Network) Pharmacology for Brain Functionality Restoration 243

that systems pharmacology is the only way to anticonvulsant pharmacotherapy. Med. Hypoth.
restore altered brain functionality (not just alle- 72, 297305.
viate symptoms) with drugs is already manifest Bishop, N.A., Lu, T., and Yankner, B.A. (2010).
in connection with such diverse pathologies as Neural mechanisms of ageing and cognitive
schizophrenia, dementia, depression, stroke, and decline. Nature 464, 529535.
epilepsy. Thus it does not seem a gratuitous state- Bock, G.R., and Goode, J.A. (1998). The limits of
ment to expect that, in the not-too-distant future reductionism in biology. Novartis Foundation
system thinking might replace mere serendipity Symposium 213 (Chichester, UK:Wiley).
as the main, driving force of drug discovery in Boison, D., Sandau U.S., Ruskin, D.N., Kawamura, M.
neuropsychopharmacology. Jr., and Masino, S.A. (2013). Homeostatic control
of brain functionnew approaches to under-
stand epileptogenesis. Front. Cell. Neurosci. 7,
References 109. doi:10.3389/fncel.2013.00109
Ahmed, A.H., and Oswald, R.E. (2010). Piracetam defines Boran, A.D.W., and Iyengar R. (2010). Systems phar-
a new binding site for allosteric modulators of -am macology. Mt. Sinai J.Med. 77, 333344.
ino-3-hydroxy-5-methyl-4-isoxazole-propionic acid Bose, B. (2013). Systems biology:Abiologists view-
(AMPA) receptors. J. Med. Chem. 53, 21972203. point. Prog. Biophys. Molec. Biol. 113, 358368.
Ahn, A.C., Tewari, M., Poon, C.S., and Phillips, Bothwell, J.H.F. (2006). The long past of systems biol-
R.S. (2006). The limits of reductionism in medi- ogy. New Phytol. 170,610.
cine: Could systems biology offer an alterna- Brigandt, I. and Love, A. (2012). Reductionism in
tive? PLoS Med. 3(6): e208. doi:10.1371/journal. biology. In: Stanford encyclopedia of philoso-
pmed.0030208 phy, E.N. Zalta, ed. http://plato.stanford.edu/
Arrell, D.K., and Terzic, A. (2010). Network systems archives/sum2012/entries/reduction-biology/.
biology for drug discovery. Clin. Pharmacol. Brodie, M.J., and Sills, G.J. (2011). Combining anti-
Therap. 88, 120125. epileptic drugsrational polytherapy? Seizure
Arrowsmith, J. (2011). Phase III and submission fail- 20, 369375.
ures 20072010. Nat. Rev. Drug Disc.10,1. Butcher, E.C. (2005). Can cell systems biology res-
Barabsi, A.L., and Oltvai, Z.N. (2004). Network cue drug discovery? Nat. Rev. Drug Discov. 4,
biology: Understanding the cells functional 461467.
organization. Nat. Rev. Genet. 5, 101113. Cakic, V. (2009). Smart drugs for cognitive enhance-
Berger, S.I., and Iyengar, R. (2009). Network analy- ment: Ethical and pragmatic considerations in
ses in systems pharmacology. Bioinformatics 25, the era of cosmetic neurology. J. Med. Ethics 35,
24662472. 611615.
Berkovic, S.F., Mulley, J.C., Scheffer, I.E., and Petrou, S. Campillos, M., Kuhn, M., Gavin, A.-C., Jensen,
(2006). Human epilepsies: Interaction of genetic L. J., and Bork, P. (2008). Drug target identifi-
and acquired factors. Trends Neurosci. 29, 391397. cation using side-effect similarity. Science 321,
Besnard, J., Ruda, G.F., Setola, V., Abecassis, K., 263266.
Rodriguiz, R.M., Huang, X.-P., Norval1, S., Sassano, Cantor, R. (2001). Breaking the MeyerOverton
M.F., Shin, A.I., Webster, L.A., Simeons, F.R.C., rule: Predicted effects of varying stiffness and
Stojanovski, L., Prat, A., Seidah, N.G., Constam, interfacial activity on the intrinsic potency of
D.B., Bickerton, G.R., Read, K.D., Wetsel, W.C., anesthetics. Biophys. J. 80, 22842297.
Gilbert, I.H., Roth, B.L., and Hopkins, A.L. (2012). Chau, P.-L. (2010). New insights into the molecu-
Automated design of ligands to polypharmacolog- lar mechanisms of general anaesthetics. Br.
ical profiles. Nature 492, 215220. J.Pharmacol. 161, 288307.
Bianchi, M.T. (2010). Promiscuous modulation of ion Chateauvieux, S., Morceau, F., Dicato, M., and
channels by anti-psychotic and anti-dementia Diederich, M. (2010). Molecular and thera-
medications. Med. Hypotheses 74, 297300. peutic potential and toxicity of valproic
Bianchi, M.T., and Botzolakis, E.J. (2010). Targeting acid. J. Biomed. Biotech., Article ID 479364.
ligand-gated ion channels in neurology and psy- doi:10.1155/2010/479364
chiatry: Is pharmacological promiscuity an obsta- Chiu, C.-T., Wang, Z., Hunsberger, J.G., and Chuang,
cle or an opportunity? BMC Pharmacol. 10, 3. D.-M. (2013). Therapeutic potential of mood sta-
http://www.biomedcentral.com/1471-2210/10/3 bilizers lithium and valproic acid:beyond bipo-
Bianchi, M.T., Pathmanathan, J., and Cash, S.S. lar disorder. Pharmacol. Rev. 65, 105142.
(2009). From ion channels to complex networks: Correll, C.U., and Gallego, J.A. (2012). Antipsychotic
Magic bullet versus magic shotgun approaches to polypharmacy: A comprehensive evaluation of
244 Part III:Homeostatic Manipulators

relevant correlates of a long-standing clinical Overnigton, J.P. (2012). ChEMBL:Alarge-scale


practice. Psychiatr. Clin. North Am. 35, 661681. bioactivity database for drug discovery. Nucl.
Crick, F.H.C. (1966). Of molecules and men Acids Res. 40, D1100D1107.
(Seattle:University of Washington Press). Giurgea, C. (1972). Vers une pharmacologie de
Csermely, P., Agoston, V., and Pongor, S. (2005). lactivit intgrative du cerveau: Tentative du
The efficiency of multi-target drugs: The net- concept nootrope en psychopharmacologie.
work approach might help drug design. Trends Actualits Pharmacologiques 25, 115156.
Pharmacol. Sci. 26, 178182. Giurgea, C.E. (1982). The nootropic concept and
Csermely, P., Korcsmros, T., Kiss, H.J.M., London, G., its prospective implications. Drug Dev. Res. 2,
and Nussinov, R. (2013). Structure and dynam- 441446.
ics of molecular networks:Anovel paradigm of Goffeau, A., Barrell, B.G., Bussey, H., Davis, R.W.,
drug discovery. Pharmacol. Ther. 138, 333408. Dujon, B., Feldmann, H., Galibert, F., Hoheisel,
Davis, L.K., Yu, D., Keenan, C.L., Gamazon, E.R., J.D., Jacq, C., Johnston, M., Louis, E.J., Mewes,
Konkashbaev, A.I., etal. (2013). Partitioning the H.W., Murakami, Y., Philippsen, P., Tettelin, H.,
heritability of Tourette syndrome and obsessive and Oliver, S.G. (1996). Life with 6000 genes.
compulsive disorder reveals differences in genetic Science 274, 546567.
architecture. PLoS Genet. 9(10): e1003864. Gouliaev, A., and Senning, A. (1994). Piracetam and
doi:10.1371/journal.pgen.1003864 other structurally related nootropics. Brain Res.
Eckert, G.P., Cairns, N.J., and Mller, W.E. (1999). Rev. 19, 180222.
Piracetam reverses hippocampal membrane Grnder, G., Hippius, H., and Carlsson, A. (2009).
alterations in Alzheimers disease. J. Neural The atypicality of antipsychotics: A concept
Transm. 106, 757761. re-examined and re-defined. Nat. Rev. Drug
Fang, F.C., and Casadevall, A. (2011). Reductionistic Discov. 8, 197202.
and holistic science. Infect. Immun. 79, Hopkins, A.L. (2008). Network pharmacology:The
14011404. next paradigm in drug discovery. Nat. Chem.
Farah, M.J., Illes, J., Cook-Deegan, R., Gardner, H., Biol. 4, 682690.
Kandel, E. King, p., Parens, E., Sahakian, B., and Hopkins, A. L. (2009). Predicting promiscuity.
Wolpe, P.R. (2004). Neurocognitive enhance- Nature 462, 167168.
ment:What can we do and what should we do? Hu, X., Zhao, D., and Strotmann, A. (2013). Mapping
Nat. Rev. Neurosci. 5, 421425. molecular association networks of nervous sys-
Fedorovich, S.V. (2013). Piracetam induces plasma tem diseases via large-scale analysis of published
membrane depolarization in rat brain synapto- research. PLoS One 8(6), e67121. doi:10.1371/
somes. Neurosci. Lett. 553, 206210. journal.pone.0067121
Franks, N.P. (2006). Molecular targets underly- Huang, Y., and Mucke, L. (2012). Alzheimer mech-
ing general anaesthesia. Br. J. Pharmacol. 147, anisms and therapeutic strategies. Cell 148,
S72S81. 12041222.
Fredholm, B., and Abbott, A. (1990). New targets for Hyman, S., Volkow, N., and Nutt, D. (2013).
drug action:Is high selectivity always beneficial? Pharmacological cognitive enhancement
Trends Pharmacol. Sci. 11, 175178. in healthy people: Potential and concerns.
French, J.A., and Faught, E. (2009). Rational poly- Neuropharmacology 64,812.
therapy. Epilepsia 50 (Suppl 8),6368. Ideker, T., Galitski, T., and Hood, L. (2001). A
Froestl, W., Muhs, A., and Pfeifer, A. (2012). new approach to decoding life: Systems biol-
Cognitive enhancers (nootropics) Part 1: Drugs ogy. Annu. Rev. Genomics Hum. Genet. 2,
interacting with receptors. J. Alzheimers Dis. 32, 343372.
793887. Insel, T., Krystal, J., and Ehlers, M. (2013). New
Froestl, W., Muhs, A., and Pfeifer, A. (2013a). drug development for cognitive enhancement
Cognitive enhancers (nootropics) Part2:Drugs in mental health:Challenges and opportunities.
interacting with enzymes. J. Alzheimers Dis. 33, Neuropharmacology 64,27.
547658. Iskar, M., Zeller, G., Zhao, X.-M., van Noort, V.,
Froestl, W., Muhs, A., and Pfeifer, A. (2013b). and Bork, P. (2011). Drug discovery in the age
Cognitive enhancers (nootropics) Part3:Drugs of systems biology: The rise of computational
interacting with targets other than receptors or approaches for data integration. Curr. Opinion
enzymes. J. Alzheimers Dis. 34,1114. Biotechnol. 23,18.
Gaulton, A., Bellis, L.J., Bento, A.P., Chambers, J., Jiang, Z., Cowell, R.M., and Nakazawa, K. (2013).
Davies, M., Hersey, A., Light, Y., McGlinchey, S., Convergence of genetic and environmental fac-
Michalovitch, D., Al-Lazikani, B., and tors on parvalbumin-positive interneurons in
Systems (Network) Pharmacology for Brain Functionality Restoration 245

schizophrenia. Front. Behav. Neurosci. 7, 116. Maher, B. (2008). Poll results: look whos doping.
doi:10.3389/fnbeh.2013.00116 Nature 452, 674675.
Keil, U., Scherping, I., Hauptmann, S., Schuessel, K., Margineanu, D.G. (2011). A weird concept with
Eckert, A., and Mller, W.E. (2006). Piracetam unusual fate: Nootropic drug. Rev. Quest. Sci.
improves mitochondrial dysfunction following 182,3352.
oxidative stress. Br. J.Pharmacol. 147, 199208. Margineanu, D.G. (2012). Systems biology impact on
Keith, C.T., Borisy, A.A., and Stockwel, B.R. (2005). antiepileptic drug discovery. Epilepsy Res. 98,
Multicomponent therapeutics for networked sys- 104115.
tems. Nat. Rev. Drug Discov. 4,7178. Margineanu, D.G. (2014). Systems biology, complex-
Keiser, M.J., Setola, V., Irwin, J.J., Laggner, C., Abbas, ity, and the impact on antiepileptic drug discov-
A.I., Hufeisen, S.J., Jensen, N.H., Kuijer, M.B., ery. Epilepsy Behav. 38, 131142. http://dx.doi.
Matos, R.C., Tran, T.B., Whaley, R., Glennon, R.A., org/10.1016/j.yebeh.2013.08.029
Hert, J., Thomas, K.L., Edwards, D.D., Shoichet, Margineanu, D.G., and Klitgaard, H. (2002).
B.K., and Roth, B.L. (2009). Predicting new Levetiracetam mechanisms of action. In:
molecular targets for known drugs. Nature 462, Antiepileptic drugs, 5th ed., R.H. Levy etal., eds.
175181. (Philadelphia: Lippincott Williams & Wilkins),
Kitano, H. (2002). Systems biology:Abrief overview. pp. 419427.
Science 295, 16621664. Marisco, P.C., Carvalho, F.B., Rosa, M.M., Girardi, B.A.,
Komrekov, I., and Jank, M. (2012). Effects of alco- Gutierres, J.M., Jaques, J.A., Salla, A.P., Pimentel,
hol on the brain biomembranes:Areview. Soud V.C., Schetinger, M.R., Leal, D.B., Mello, C.F.,
Lek. 57,7577. and Rubin, M.A. (2013). Piracetam prevents
Kuhn, M., Szklarczyk, D., Franceschini, A., von scopolamine-induced memory impairment and
Mering, C., Jensen, L.J., and Bork, P. (2012). decrease of NTPDase, 5-nucleotidase and ade-
STITCH 3: Zooming in on proteinchemical nosine deaminase activities. Neurochem. Res.
interactions. Nucl. Acids Res. 40, D876D880. 38, 17041714.
Kukreja, S., Kalra, G., Shah, N., and Shrivastava, A. Mazzocchi, F. (2008). Complexity in biology. EMBO
(2013). Polypharmacy in psychiatry: A review. Rep. 9,1014.
Mens Sana Monogr. 11,8299. Mazzocchi, F. (2012). Complexity and the
Lanni, C., Lenzken, S.C., Pascale, A., Del Vecchio, I., reductionism-holism debate in systems biol-
Racchi, M., Pistoia, F., and Govoni, S. (2008). ogy. Wiley Interdiscip. Rev. Syst. Biol. Med. 4,
Cognition enhancers between treating and dop- 413437. doi:10.1002/wsbm.1181
ing the mind. Pharmacol. Res. 57, 196213. Mehler, M.F. (2008). Epigenetic principles and mech-
Laughlin, S.B., and Sejnowski, T.J. (2003). anisms underlying nervous system functions in
Communication in neuronal networks. Science health and disease. Prog. Neurobiol. 86, 305341.
301, 18701874. Millan, M.J., Agid, Y., Brne, M., Bullmore, E.T.,
Lee, J.A., Uhlik, M.T., Moxham, C.M., Tomandl, D., Carter, C.S., Clayton, N.S., Connor, R., Davis,
and Sall, D.J. (2012). Modern phenotypic drug S., Deakin, B., DeRubeis, R.J., Dubois, B., Geyer,
discovery is a viable, neoclassic pharma strategy. M.A., Goodwin, G.M., Gorwood, P., Jay, T.M.,
J. Med. Chem. 55, 45274538. Jols, M., Mansuy, I.M., Meyer-Lindenberg, A.,
Lscher, W. (2002). Valproic acid mechanisms of Murphy, D., Rolls, E., Saletu, B., Spedding, M.,
action. In:Antiepileptic drugs, 5th ed., R.H. Levy Sweeney, J., Whittington, M., and Young L.J.
etal., eds. (Philadelphia:Lippincott Williams & (2012). Cognitive dysfunction in psychiatric
Wilkins), pp. 767779. disorders: Characteristics, causes and the quest
Lscher, W., and Schmidt, D. (1994). Strategies in for improved therapy. Nat. Rev. Drug Discov. 11,
antiepileptic drug development:Is rational drug 141168.
design superior to random screening and struc- Miller, G. (2010). Is pharma running out of brainy
tural variation? Epilepsy Res. 17, 95134. ideas? Science 329, 502504.
Lu, J.-J., Pan, W., Hu, Y.-J., and Wang, Y.-T. (2012). Mingeot-Leclercq, M.-P., Lins, L., Bensliman, M.,
Multi-target drugs: The trend of drug research Thomas, A., Van Bambeke, F., Peuvot, J.,
and development. PLoS One 7(6):e40262. Schanck, A., and Brasseur, R. (2003). Piracetam
doi:10.1371/journal.pone.0040262 inhibits the lipid-destabilising effect of the amy-
Lynch, B.A., Lambeng N., Nocka K., Kensel-Hammes, loid peptide AP C-terminal fragment. Biochim.
P., Bajjalieh, S.M., Matagne, A., and Fuks, B. Biophys. Acta 1609,2838.
(2004). The synaptic vesicle protein SV2A is the Morphy, R., and Rankovic, Z. (2007). Fragments,
binding site for the antiepileptic drug levetirace- network biology and designing multiple ligands.
tam. Proc. Nat. Acad. Sci. U.S.A. 101, 98619866. Drug Discov. Today 12, 156160.
246 Part III:Homeostatic Manipulators

Mller, W.E., Koch, S., Scheuer, K., Rostock, A., and Sahakian, B., and Morein-Zamir, S. (2007).
Bartsch, R. (1997). Effects of piracetam on mem- Professors little helper. Nature 450, 11571159.
brane fluidity in the aged mouse, rat, and human Sams-Dodd, F. (2005). Target-based drug discov-
brain. Biochem. Pharmacol. 53, 135140. ery:Is something wrong? Drug Discov. Today 10,
Naftalin, R.J., Cunningham, P., and Afzal-Ahmed, I. 139147.
(2004). Piracetam and TRH analogues antagonise Sams-Dodd, F. (2013). Is poor research the cause of
inhibition by barbiturates, diazepam, melatonin the declining productivity of the pharmaceuti-
and galanin of human erythrocyte D-glucose cal industry? An industry in need of a paradigm
transport. Br. J.Pharmacol. 142, 594608. shift. Drug Discov. Today 18, 211217.
Nehlig, A. (2010). Is caffeine a cognitive enhancer? J. Sauer, U., Heinemann, M., and Zamboni, N. (2007).
Alzheimers Dis. 20 (Suppl. 1), S85S94. Getting closer to the whole picture. Science 316,
Noorbakhsh, F., Overall, C.M., and Power, C. (2009). 550551.
Deciphering complex mechanisms in neuro- Schadt, E.E., Friend, S.H., and Shaywitz, D.A. (2009).
degenerative diseases: the advent of systems biol- A network view of disease and compound screen-
ogy. Trends Neurosci. 32, 88100. ing. Nat. Rev. Drug Discov. 8, 286295.
OConnor, T., and Wong, H.Y. (2012). Emergent Shorvon, S. (2001). Pyrrolidine derivatives. Lancet
properties. In: Stanford encyclopedia of phi- 358, 18851892.
losophy, E.N. Zalta, ed. (Spring 2012 ed.), http:// Singer, W. (2007). Understanding the brain. EMBO
plato.stanford.edu/archives/spr2012/entries/ Rep. 8, S16S19.
properties-emergent/ Sinha, S., Jesan, T., and Chatterjee, N. (2009). Systems
Pammolli, F., Magazzini, L., and Riccaboni, M. biology: From the cell to the brain. In: Current
(2011). The productivity crisis in pharmaceutical trends in science: Platinum Jubilee Special, N.
R&D. Nat. Rev. Drug Discov. 10, 428438. Mukunda, ed. (Bangalore: Indian Academy of
Panagiotou, G., and Taboureau, O. (2012). The Sciences), pp. 199205.
impact of network biology in pharmacology and Slais, K., Machalova, A., Landa, L., Vrskova, D., and
toxicology. SAR QSAR Environ Res. 23, 221235. Sulcova, A. (2012). Could piracetam potentiate
Paul, S.M. (2006). Alcohol-sensitive GABA receptors behavioural effects of psychostimulants? Med.
and alcohol antagonists. Proc. Nat. Acad. Sci. Hypoth. 79, 216218.
U.S.A. 103, 83078308. Spanagel, R. (2009). Alcoholism:Asystems approach
Pennisi, E. (2012). ENCODE project writes eulogy from molecular physiology to addictive behav-
for junk DNA. Science 337, 11591160. ior. Physiol. Rev. 89, 649705.
Peoples, R.W., Li, C., and Weight, F.F. (1996). Lipid vs. Sporns, O. (2003). Network analysis, complexity and
protein theories of alcohol action in the nervous sys- brain function. Complexity 8,5660.
tem. Annu. Rev. Pharmacol. Toxicol. 36, 185201. Sun, J., Xu, H., and Zhao, Z. (2012). Network-assisted
Poduri, A., Evrony, G.D., Cai, X., and Walsh, C.A. investigation of antipsychotic drugs and their
(2013). Somatic mutation, genomic variation, targets. Chem. Biodiv. 9, 900910.
and neurological disease. Science 341, 1237758. Swinney, D.C., and Anthony, J. (2011). How were new
Pujol, A., Mosca, R., Farrs, J., and Aloy P. (2010). medicines discovered? Nat. Rev. Drug Discov. 10,
Unveiling the role of network and systems biol- 507519.
ogy in drug discovery. Trends Pharmacol. Sci. 31, Szczepankiewicz, A. (2013). Evidence for single
115122. nucleotide polymorphisms and their associa-
Rang, H.P. (2006). The receptor concept: tion with bipolar disorder. Neuropsychiatric Dis.
Pharmacologys big idea. Br. J. Pharmacol. 147, Treat. 9, 15731582.
S9S16. Taboureau, O., Baell, J.B., Fernndez-Recio, J., and
Reik, W. (2007). Stability and flexibility of epigenetic Villoutreix, B.O. (2012). Established and emerg-
gene regulation in mammalian development. ing trends in computational drug discovery in the
Nature 447, 425432. structural genomics era. Chem. Biol. 19, 2941.
Rosenberg, G. (2007). The mechanisms of action of Takahashi, T. (2013). Complexity of spontane-
valproate in neuropsychiatric disorders:Can we ous brain activity in mental disorders. Prog.
see the forest for the trees? Cell. Mol. Life Sci. 64, Neuropsychopharmacol. Biol. Psychiatry 45,
20902103. 257265.
Roth, B.L., Sheffler, D.J., and Kroeze, W.K. (2004). Tejada, J., Costa, K.M., Bertti, P., and Garcia-
Magic shotguns versus magic bullets:Selectively Cairasco, N. (2013). The epilepsies: Complex
non-selective drugs for mood disorders and challenges needing complex solutions. Epilepsy
schizophrenia. Nat. Rev. Drug Discov. 3, 353359. Behav. 26, 212228.
Systems (Network) Pharmacology for Brain Functionality Restoration 247

Van der Schyf, C.J., Geldenhuys, W.J., and Youdim, von Bertalanffy, L. (1950). An outline of general sys-
M.B.H. (2006). Multifunctional drugs with dif- tem theory. Br. J.Philos. Sci. 1, 139164.
ferent CNS targets for neuropsychiatric disor- von Bertalanffy, L. (1968). General system theory:
ders. J. Neurochem. 99, 10331048. Foundations, developments, applications (New
Van Regenmortel, M.H.V. (2004). Reductionism and York:Braziller).
complexity in molecular biology. EMBO Rep. 5, Wang, Y., Xiao, J., Suzek, T.O., Zhang, J., Wang, J.,
10161020. and Bryant, S.H. (2009). PubChem: A pub-
Vidal, D., and Mestres, J. (2010). In silico recepto- lic information system for analyzing bioac-
rome screening of antipsychotic drugs. Mol. Inf. tivities of small molecules. Nucl. Acids Res. 37,
29, 543551. W623W633.
Villoslada, P., Steinman, L. and Baranzini, S.E. Winblad, B. (2005). Piracetam:Areview of pharma-
(2009). Systems biology and its application to the cological properties and clinical uses. CNS Drug
understanding of neurological diseases. Ann. Rev. 11, 169182.
Neurol. 65, 124139. Yang, A.C., and Tsai, S.-J. (2013). Is mental ill-
Volterra, A., and Meldolesi J. (2005). Astrocytes, ness complex? From behavior to brain. Prog.
from brain glue to communication elements. Neuropsychopharmacol. Biol. Psychiatry 45,
Nat. Rev. Neurosci. 6, 626640. 252256.
15
Ketogenic Diets forNeurological Disorders
L I N D S E Y B . G A N O , M A N I S H A PAT E L , A N D J O N G M . R H O

INTRODUCTION potentially be amenable to treatments that


The ketogenic diet (KD) is a high-fat, low- restore normal metabolism. A common thread
carbohydrate therapy for drug-resistant epilepsy of such diet-based therapies for brain diseases
(Neal, 2008; Vining, 1998) and is increasingly is that metabolic substrates and nutrients can
being studied for therapeutic efficacy in a num- exert profound effects on neuronal plasticity,
ber of neurological disorders, including epilepsy, modifying neural circuits and cellular proper-
headache, neurotrauma, Alzheimers disease ties to enhance and normalize function. Further,
(AD), Parkinsons disease (PD), sleep disorders, as there is increasing evidence for diet-induced
brain cancer, autism, pain, and amyotrophic lat- epigenetic mechanisms contributing causally to
eral sclerosis (ALS) (Baranano & Hartman, 2008; the development of common chronic diseases
Stafstrom & Rho, 2012). This is a result of growing (McKay & Mathers, 2011; Roopra, Dingledine, &
experimental evidence for the broad neuroprotec- Hsieh, 2012), greater knowledge of processes and
tive properties of the KD and mechanistic linkages players such as DNA methylation, histone modi-
to key cellular signaling pathways and fundamen- fications, and noncoding microRNAs will be
tal bioenergetics processes, notably within mito- needed to understand the relationships between
chondria (Milder & Patel, 2012; Waldbaum & energy dysregulation and therapeutic strategies
Patel, 2010). In recent years, the field of neurome- to counter such impairment (McKay & Mathers,
tabolism has been greatly amplified by interest in 2011; Urdinguio, Sanchez-Mut, & Esteller,2009).
dietary treatments such as the KD (Milder & Patel, This chapter explores the rationale and evidence
2012)and by the recognition that bioenergetic dys- for using the KD and related dietary treatments in
regulation may be a critical pathophysiological fac- a broad range of neurological disorders and high-
tor in diseases of the nervous system (Lin & Beal, lights novel mechanisms that have been implicated
2006; Pathak, Berthet, & Nakamura, 2013). Indeed, in their actions. However, it is important to recog-
there is increasing appreciation for the concept of nize that much of the data discussed here remain
energy failureprincipally from mitochondrial preliminary in nature. Nevertheless, the therapeu-
dysfunctionas a key mechanism resulting in tic potential for dietary therapies for neurological
neuronal death seen in neurodegenerative diseases disorders remains almost limitless when viewed
(Pathak, Berthet, & Nakamura,2013). from the perspective of salvaging neuronal bioen-
That diet and nutrition influence brain func- ergetic dysfunction (Masino & Rho, 2012; Milder
tion should not be altogether surprising, and there & Patel, 2012; Pathak, Berthet, & Nakamura,2013).
exist much clinical and laboratory data linking
disturbances in energy metabolism to a vari- KETOGENIC DIET AND
ety of clinical disorders (Roth, Szulc, & Danoff, E P I L E P S Y: H I S T O R I C A L
2011; Schiff et al., 2011; Waldbaum & Patel, ASPECTS
2010). Fundamentally, any disease in which the The use of dietary manipulations to treat
pathogenesis is affected by disturbances in cel- epilepsyin particular, controlling seizures
lular energy utilizationand this could apply to through sustained fastingdates back to the
almost every known medical conditionwould time of Hippocrates (Bailey, Pfeifer, & Thiele,

This research was originally published in the Journal of Lipid Research. Gano LB, Patel M, Rho JM. Ketogenic
diets, mitochondria, and neurological diseases. Journal of Lipid Research. 2014 Nov; 55(11):221128. the
American Society for Biochemistry and Molecular Biology.
Ketogenic Diets and Mitochondria and Neurological Diseases 249

2005; Wheless, 2008). In modern times, reports for a dietician to implement and monitor the
of modifying diets to treat seizures emerged in patient while he or she is on the diet (Kossoff &
the early 20th century both in France and in the Rho, 2009; Kossoff, Zupec-Kania, & Rho, 2009;
United States (Wheless, 2008). Importantly, in Kossoff et al., 2009). The classic KD is primarily
the 1920s, Drs. Stanley Cobb and W.G. Lennox based on ingestion of saturated long-chain fatty
at Harvard Medical School conducted research acids (FA) (Kossoff & Rho, 2009). Upon restric-
on the effects of starvation as a treatment for epi- tion of carbohydrates, ketogenesis occurs in the
lepsy (Wheless, 2008). The effects of fasting were liver and ketone bodies are exported to the circu-
seen within 2 to 3days, and hence they concluded lation (Figure 15.1). Circulating concentrations of
that a lack of carbohydrates in the diet prompted the major ketone bodies BHB, ACA, and acetone
a change in metabolism to force the utilization have been shown to significantly increase within 1
of fats to produce energy (Wheless, 2008). It was to 3days after initiation of theKD.
also recognized that in the fasted state, the body A key aspect of the KD includes partial caloric
produced ketone bodies (-hydroxybutyrate restriction (CR). Prior to starting the diet, a fast-
[BHB], acetoacetate [ACA], and acetone) through ing period of 24 to 48 hours is typical (Freeman,
the liver and that a diet high in fats but low in car- Kossoff, & Hartman, 2007), but the need for this
bohydrates could mimic fasting (Wheless,2008). requirement has been debated (Bergqvist et al.,
Dr.Russell Wilder at Mayo Clinic first referred 2005). An initial period of fasting may accelerate
to this special high-fat diet as the ketogenic diet seizure control and is therefore recommended for
(Wheless, 2008). During an era when antiseizure patients with a greater need for immediate seizure
drugs (ASDs) were scarce, the KD became quickly reduction (Freeman & Vining, 1999; Kossoff etal.,
popularized in large medical centers. However, 2008). Adecrease in daily caloric intake of 10% to
with the advent of diphenylhydantoin in 1938, the 25% is typical with the diet. Gluconeogenesis may
KD quickly fell out of favor due to the simplicity result from consumption of excess calories, and
of prescribing an oral medication as opposed to a therefore this slight reduction in calories is thought
strict and exacting dietary regimen. Nevertheless, to increase the efficacy of the diet by maintaining
a variation of the KDthat is, the medium-chain ketosis (Bough & Rho, 2007; Masino & Rho,2012).
triglyceride (MCT) dietemerged later as yet After initiation of the diet, seizure control
another dietary option for medically intractable slowly increases within the first few days to weeks
epilepsy (Huttenlocher, Wilbourn, & Signore, (Freeman & Vining, 1999; Kossoff et al., 2008).
1971)and remains today as an alternative to the This is believed to be due to the gradual elevation
classic KD (Neal etal.,2009). of circulating ketone bodies. However, since serum
In recent years, there has been an explosion levels of ketone bodies do not correlate tightly
in clinical use of the KD and its variants (Kossoff with seizure control, it is unknown whether these
etal., 2013; Muzykewicz etal., 2009), as well as substrates are directly responsible for the clinical
in scientific interest regarding the mechanisms effects observed (Kossoff & Rho, 2009). Despite
underlying their action (Lutas & Yellen, 2013; this uncertainty, it is known that a break from the
Masino & Rho, 2012). Even beyond this resur- diet by ingestion of carbohydrates rapidly reverses
gence in popularity for epilepsy, the diet has been the antiseizure effects of the diet. In fact, the
increasingly found to exert protective effects in onset of seizures can occur less than an hour after
a variety of neurological diseases (Baranano & administration of glucose (Huttenlocher,1976).
Hartman, 2008; Stafstrom & Rho, 2012), and new Historically, the KD has been primarily used
mechanistic insights have steadily emerged. to treat epilepsy in pediatric patients. Efficacy
in seizure control with the KD is assumed to
Efficacy inEpilepsy:Clinical Studies be enhanced in younger patients and is thought
The classic KD utilizes a fat-to-carbohydrate to reflect age-dependent changes in the expres-
plus protein ratio of 4:1 by weight, with approxi- sion of monocarboxylate transporters, which
mately 90% of daily caloric intake coming from transfer ketone bodies across the blood-brain
fat, and the inclusion of a small amount of protein barrier from the systemic circulation (Pierre
(1 g/kg BW/day) to ensure adequate growth in & Pellerin, 2005; Prins, 2008) (Figure 15.1).
pediatric patients (Kossoff & Rho, 2009; Kossoff, However, despite challenges in maintaining the
Zupec-Kania, & Rho, 2009; Kossoff et al., 2009). diet in older patientsmostly due to compli-
A fat-to-carbohydrate ratio of 3:1 may be uti- ance issuesimproved seizure control has been
lized based on a patients needs and efficacy, reported as well in adolescents and adults (Klein
which underscores the importance of the need etal., 2010; Mady etal., 2003; Payne etal.,2011).
250 Part III:Homeostatic Manipulators

Ketogenic Diet

ATP
Fatty acids (FA)

UCP-2
Krebs cycle

FA ?
CAT CPT-1
FAO 1
Acetone Acetone BHB ACA

MRC
Acetyl-CoA ACA ACA 2
BHB BHB MCT-1 Acetoacetyl-CoA
3
BHB Acetyl-CoA
Krebs cycle Glial cell
Pyruvate Pyruvate
Glucose
Oxaloacetate Mitochondrion
Glucose Glycolysis
GLUT-1
Mitochondrion
Glucose Neuron
Hepatocyte Capillary BBB

Figure15.1 Metabolic pathways involved in ketogenic diet treatment. CAT=carnitine-acylcarnitine translo-


case; FAO=fatty acid oxidation; ACA=acetoacetate; BHB=-hydroxybutyrate; MCT-1=monocarboxylate transporter-1;
GLUT-1=glucose transporter-1; BBB=blood-brain barrier; CPT-1=carnitine palmitoyl transferase; UCP=uncoupling
protein; ATP=adenosine triphosphate; =3-hydroxybutyrate dehydrogenase, =succinyl-CoA3-oxoacid CoA trans-
ferase; = mitochondrial acetoacetyl-CoA thiolase; MRC=mitochondrial respiratory complex.
Reprinted with permission:Kim do Y, Rho JM. The ketogenic diet and epilepsy. Curr Opin Clin Nutr Metab Care 2008
Mar;11(2):11320. Lippincott, Williams & Wilkins.

Despite numerous clinical reports document- PTZ and flurothyl, a volatile convulsant (Bough
ing efficacy of the KD against intractable epilepsy etal., 2006). The KD also increased resistance in
(Kossoff & Rho, 2009), very few class 1 and 2 stud- the 6Hz model of epilepsy in mice, but the effects
ies exist. After nearly a century of use, the strong- required 5days on the diet and protection was lost
est evidence became available only as recently as after 3 weeks (Hartman etal., 2008). Differences
2008. In this randomized controlled trial of 145 in age-dependent protection and latency to seizure
children, ages 2 to 16 years with daily seizures control were shown in flurothyl-induced seizures
and who did not respond to at least two ASDs, it in juvenile and adult mice (Rho etal., 1999). The
was shown that those who maintained the KD for KD increased time to the first generalized (clonic)
greater than 3months had a significant reduction seizure in juvenile mice after 7 and 12days but not
in the mean percentage of baseline seizures (Neal after 3days in young mice or after 15days in adult
etal., 2008). Seven percent of children on the KD mice. However, 15days on the diet protected adult
demonstrated a greater than 90% reduction in sei- mice against second generalized (tonic extension)
zures, compared with 0% on the control diet, and seizures, whereas juvenile mice showed no differ-
there was a more than 50% reduction in seizures ence after 3, 7, and 12days on the diet. These and
in 38% of the patients on the KD versus 6% on the other studies emphasize the inherent variability of
control diet (Neal etal.,2008). the effects of the KD as a function of the different
rodent models used, the heterogeneous methods
Efficacy inEpilepsy: used for inducing seizures, as well as the dietary
Preclinical Studies protocols adopted.
Efficacy of the KD has also been shown in multi-
ple animal models of seizures and epilepsy. Rats Variations onthe KetogenicDiet
fed the KD for nearly 2 weeks exhibited increased Despite documented efficacy of the KD against
seizure thresholds after repeated intravenous infu- seizure disorders, implementation remains chal-
sion of pentylenetetrazol (PTZ), a chemoconvul- lenging (e.g., poor compliance due to lack of pal-
sant that blocks -aminobutyric acid (GABA) type atability and concerns regarding long-term health
Areceptors, whereas a 3-week treatment increased consequences such as increases in cholesterol and
thresholds in response to a single exposure to both triglycerides, slow growth in pediatric patients,
Ketogenic Diets and Mitochondria and Neurological Diseases 251

etc.) (Kossoff & Dorward, 2008; Kossoff, Laux, children under 10years of age on a 60% MCT diet
et al., 2008; Kossoff et al., 2008). Hence clinical with the classic 3:1 KD, circulating levels of BHB
researchers have sought alternative therapies that were found to be similar (Huttenlocher, 1976;
retain antiseizure effects that are devoid of these Huttenlocher, Wilbourn, & Signore,1971).
limitations. In addition to the MCT diet, two dis- Medium-chain trigylerides produce a higher
tinct variations on the KD theme include the mod- level of ketosis compared with long-chain trigly-
ified Atkins diet (MAD) and the low-glycemic erides, and this allows for a decrease in the intake
index treatment (LGIT) (Cross & Neal, 2008) of these fats and a greater consumption of carbo-
(Figure 15.2). Finally, there is CR, a dietary prac- hydrates and protein on this modified diet (Neal
tice known to reduce the progression of many et al., 2009).
age-related diseases. Interestingly, this particu- The first long-term randomized trial com-
lar dietary strategy, which the KD was originally pared the effects of the classic KD and the MCT
designed to mimic, has been an intense focus for diet in children with intractable epilepsy after
researchers interested in the mechanisms under- 3, 6, and 12 months on these regimens (Neal
lying aging/longevity, neurological diseases, and a etal., 2009). The 4:1 classic KD was used for the
host of other conditions (Bishop & Guarente, 2007; majority of the children, although some chil-
Fusco & Pani, 2013; Libert & Guarente,2013). dren were on a 3:1 ratio. The MCT diet was com-
posed of ~15% carbohydrates, ~10% protein, 30%
The Medium-Chain TriglyerideDiet long-chain FA, and 40% to 45% MCT fat. There
The MCT diet, whichas the name implies were no differences between dietary groups in
utilizes medium-chain triglycerides as the fat com- percentage of reduction in baseline seizures after
ponent, was first described in 1971 and was reported 3, 6, and 12months. Additionally, no differences
to induce antiseizure effects similar to the classic were found between the two diets in those achiev-
KD (Huttenlocher, 1976; Huttenlocher, Wilbourn, ing seizure reductions greater than 50% or 90%.
& Signore, 1971). In an initial comparison of Serum levels of ACA and BHB were greater in
children on the classic KD after 3 and 6months,
Classic ketrogenic Medium-Chain Triglyceride but only levels of ACA remained elevated in those
(4:1)
on the KD after 12months (Neal etal.,2009).
3%

7% The Modified AtkinsDiet


20% The MAD was designed to mimic the high-fat
KD while allowing more liberalized intake of
10% protein, fluids, and calories to increase compli-
70% ance, especially in adults (Kossoff & Dorward,
90%
2008)(Figure 15.2). However, the MAD still relies
on carbohydrate restriction, initially 10 g/day
in children and 15 g/day in adults; this can
5%
be increased to 20 to 30 g/day after a couple of
27%
months on the diet depending on seizure control
25%
(Kossoff & Dorward, 2008). The MAD was origi-
45% nally developed at the Johns Hopkins Hospital
70% and is considered to be a modified version of
28% the original Atkins diet because weight loss is not
the principal goal and protein intake is liberal-
ized (Kossoff & Dorward,2008).
Modified Atkins Low-Glycemic Index
Seizure control with the MAD was originally
Fat Protein Carbohydrates shown in 6 children and adults with intractable
epilepsy (Kossoff et al., 2003). In the 5 patients
Figure 15.2 Comparison of four major keto- able to maintain ketosis for 6 to 24 months, 3
genic diets. Pie charts depict relative proportion of demonstrated a significant reduction in seizures
calories provided by fat, protein, and carbohydrates for and the opportunity to reduce concomitant ASDs
the classic ketogenic diet (4:1 ratio by weight of fats to (Kossoff etal., 2003). Following this initial report,
carbohydrate plus protein), the medium-chain triglyc- the Dr.Robert C.Atkins Foundation sponsored
eride (MCT) diet, the modified Atkins diet (MAD), the first prospective study on the MAD in 20
and the low-glycemic index therapy (LGIT). children who experienced at least three seizures
252 Part III:Homeostatic Manipulators

per week and had prior use of at least two ASDs and circulating levels of BHB during follow-ups
(Kossoff etal., 2006). In those that maintained the (Muzykewicz etal.,2009).
diet for at least 6months, 4 became seizure-free,
13 (65%) had a greater than 50% reduction in sei- Caloric Restriction
zures, and 7 (35%) demonstrated a greater than As the KD was originally designed to mimic the
90% decrease (Kossoff etal., 2006). Efficacy of the effects of fasting, it was postulated that CR (or
MAD on seizure control in adults was also shown intermittent fasting) may provide similar results
in a prospective study of 30 adults with at least without the need for a large regular intake of fats.
weekly seizures and previous use of at least two CR involves a reduction in total daily caloric intake
ASDs (Kossoff etal., 2008). Of those that main- below the ad libitum level without the risk of mal-
tained the diet, a greater than 50% reduction nutrition (Bishop & Guarente, 2007; Guarente,
in seizures was found in 14 (47%) patients after 2013). CR is well known to ameliorate many
3months and in 10 (33%) adults after 6months age-related and metabolic diseases and is the only
(Kossoff etal.,2008). natural method known to increase lifespan across
multiple species (Libert & Guarente, 2013). As
The Low-Glycemic Index Treatment such, comparisons of the KD and CR on seizure
The glycemic index (GI) is a measure of the rise in control have been performed in animal models of
circulating levels of glucose in response to inges- epilepsy. However, clinical studies involving CR
tion of a specific food. The index estimates how or intermittent fasting for epilepsy have yet to be
each gram of available carbohydrate in a food conducted (Hartman & Stafstrom,2013).
source will elevate levels of blood glucose com- A study in rats of varying ages treated with mild
pared to the consumption of pure glucose, which calorically restricted standard chow diet (90% of
is assigned a GI of 100. The rationale for the LGIT recommended daily calories) and an isocaloric
was based on the clinical observation that the KD KD showed protection against PTZ-induced sei-
led to a sustained drop in blood glucose levels, zures (Bough etal., 1999). Rats fed the isocaloric
and on laboratory studies showing decreased sei- KD exhibited a greater threshold to PTZ-induced
zure susceptibility in epileptic (EL) mice induced seizures compared to CR-standard chow animals,
by CR (Greene et al., 2001). The LGIT prevents which in turn demonstrated greater protection
large postprandial increases in blood glucose, than those fed the same standard chow ad libitum
resulting in more stable circulating levels of glu- (Bough etal.,1999).
cose, and in early studies has been shown to pro- In the EL genetic mouse model of epilepsy,
vide seizure control in a broad range of patients CR (15% or 30%) delayed the development and
(Pfeifer, Lyczkowski, & Thiele, 2008; Pfeifer & frequency of seizures in both juvenile and adult
Thiele, 2005). This diet also allows for a greater animals (Greene et al., 2001). Additionally,
intake of carbohydrates (~4060 g/day), but this is when compared with a previous study by the
restricted to those foods with a GI of <50 (Pfeifer, same group that utilized the typical KD in EL
Lyczkowski, & Thiele, 2008; Pfeifer & Thiele, mice (Todorova et al., 2000), the authors found
2005). Total caloric intake is determined based on that the mild form of CR (15%) resulted in
patients needs, with 20% to 30% of calories com- greater protection than the KD in juvenile mice.
ing from protein and the remaining 60% from fats Afollow-up study in adult EL mice showed that
(Pfeifer, Lyczkowski, & Thiele, 2008)(Figure15.2). CR in the context of both a ketogenic and a
In an initial study of 20 epileptic patients high-carbohydrate/low-fat diet reduced seizure
(534 years old), 10 out of 20 patients demon- susceptibility (Mantis et al., 2004), leading the
strated a >90% seizure reduction while on the authors to conclude that restriction of calories,
LGIT, despite achieving lower levels of circulating not the composition of the diet per se, is the key
ketone bodies (Pfeifer & Thiele, 2005). In a larger determinant of seizure control.
cohort of children on the LGIT, 89% of whom
failed to respond to at least three ASDs, seizure M EC H A N I ST IC OV E RV I E W:
frequencies were reduced by greater than 50% in KETOGENESIS
42%, 50%, 54%, 64%, and 66% in patients after 1, Sustained intake of a high-fat, low-carbohydrate
3, 6, 9, and 12months of treatment, respectively diet increases the rates of FA oxidation (FAO)
(Muzykewicz etal., 2009). This study also showed and gluconeogenesis. The end product of FAO is
an inverse relation between efficacy of the diet acetyl-Coenzyme A(acetyl-CoA), which can enter
and serum glucose at certain time-points but did the tricarboxylic acid (TCA) cycle and reacts with
not find a correlation between seizure reduction oxaloacetate to form citrate. However, under these
Ketogenic Diets and Mitochondria and Neurological Diseases 253

metabolic conditions, oxaloacetate is also diverted studies showing antiseizure properties of the
to gluconeogenesis and exported out of the mito- major ketone BHB have not yet been forthcoming.
chondria as malate. In the liver, increased produc-
tion of acetyl-CoA results in levels that exceed the Neurotransmitters and Ion
amount of oxaloacetate available for entry into the Channel Regulation
TCA cycle, and ketogenesis is then initiated when A long-standing theory for KD action involves
two acetyl-CoA molecules are combined to form changes in the levels of certain neurotransmitters
acetoacetyl-CoA (Figure 15.1). Acetoacetyl-CoA (NT), as a result of altered synthesis and/or clear-
is then condensed with another molecule of ance from the synaptic cleft. The production of the
acetyl-CoA to form 3-hydroxy-3-methylglutaryl major excitatory NT glutamate is paradoxically
CoA (HMG-CoA), in a nonreversible step cata- linked to the synthesis of the main inhibitory NT
lyzed by the rate-limiting enzyme HMG-CoA syn- GABA via the action of the biosynthetic enzyme
thase 2 (HMG-CoAS2). The ketone body ACA is for GABA, glutamate decarboxylase (GAD). The
then produced via the breakdown of HMG-CoA, KD has been proposed to alter the metabolism of
releasing a molecule of acetyl-CoA. ACA can then glutamatein response to ketosisresulting in
be exported from the liver into the circulation for increased levels of GABA and enhanced inhibi-
uptake by tissues with high metabolic demands, tory neurotransmission (Yudkoff etal.,2005).
such as the heart, skeletal muscle, and the brain. Glutamate is cleared from the synaptic
Alternatively, ACA can be further reduced to the space by astrocytes, which convert glutamate
ketone body BHB by BHB dehydrogenase (BDH) to glutamine through the action of the glial
in a reaction that is coupled to the ratio of the oxi- enzyme glutamine synthetase. Glutamine is
dized to reduced forms of nicotinamide adenine then exported to neurons where it is hydrolyzed
dinucleotide (NAD+; i.e., NAD+/NADH) or the to glutamate and can then either be converted
spontaneous decarboxylation of ACA that yields to GABA or transaminated to aspartate, in a
acetone, another ketone body. BHB and acetone reaction that also requires oxaloacetate. Since
are also excreted from the liver and taken up from the KD induces metabolic changes that require
the circulation into other tissues. In extrahepatic available oxaloacetate for either incorporation of
tissues, BDH catalyzes the first reaction in ketone acetyl-CoA into the TCA cycle or for gluconeo-
body oxidation from BHB to ACA, which makes it genesis, the production of aspartate from gluta-
an important regulator of mitochondrial NAD+/ mate is reduced. This may result in enhanced flux
NADH status (Cotter, Schugar, & Crawford, 2013). through GAD to increase the synthesis of GABA
ACA is then further oxidized to acetyl-CoA for (Yudkoff etal.,2005).
entry into the TCAcycle. In children fed the KD, cerebrospinal fluid
levels of GABA were increased but without a
KetoneBodies change in glutamate concentrations (Dahlin
The earliest demonstration of ketone bodies et al., 2005). However, rats fed the KD for 3
inducing antiseizure effects was made by Keith weeks showed a reduction in brain glutamate
in the 1930s. Acetoacetate was shown to pro- levels, with no change in GABA (Melo, Nehlig,
tect against thujone-induced seizures in rabbits & Sonnewald, 2006). Another study in rats fed
(Keith, 1933). This was followed decades later the KD for 3 weeks found increased levels of glu-
by two additional studies demonstrating in vivo tamate and glutamine in the hippocampus, but
antiseizure effects of ketone bodies (Likhodii this was also associated with an overall decrease
etal., 2003; Rho etal., 2002). In the Frings audio- in the transcripts of genes involved in synaptic
genic seizure-susceptible mouse (a model of transmission (Bough etal., 2006). Further, using
sensory-evoked reflex seizures), acute adminis- both mild CR (i.e., 90% of daily energy require-
tration of acetone and ACA led to an elevation ments, or 10% CR) and an isocaloric KD, inves-
in seizure threshold, whereas the more prevalent tigators found significant increases in the mRNA
ketone body BHB had no effect on sound-induced expression of both isoforms of GAD (GAD65
seizures. In a separate study, acetone displayed and GAD67) in several brain regions and inde-
dose-dependent antiseizure effects in four diverse pendent of ketogenic effects (Cheng etal., 2004).
rodent models of epilepsy, including maximal Additionally, reduced levels of aspartate were
electroshock seizures, subcutaneous PTZ, amyg- found in a mouse model of ketosis, along with
dala kindling, and the AY-9944 (an inhibitor the expected increases in acetyl-CoA, with no
of cholesterol biosynthesis) model of atypical change in the levels of GABA and glutamate
absence seizures (Likhodii etal., 2003). Curiously, (Yudkoff etal., 2005). However, this same study
254 Part III:Homeostatic Manipulators

also showed increases in glutamine and GABA metabolism of glucose versus ketone bodies, as
upon infusion of the nitrogen donors alanine or neurons and astrocytes from Bad -/- or BadS155A
leucine (Yudkoff etal.,2005). mice exhibited reduced mitochondrial oxida-
As another component of the NT theory, it tive metabolism of glucose but enhanced mito-
was hypothesized that the KD might also enhance chondrial respiration in the presence of BHB
reuptake of glutamate from the synaptic cleft by (Gimenez-Cassina etal.,2012).
astrocytes (Bough etal., 2007). However, in rats fed Interestingly, this change in metabolism was
a KD for 4 to 5 weeks, there were no differences in implicated in neuronal excitability, as Bad -/- or
the levels of brain glutamate transporters and no BadS155A mice also demonstrated increased
change in the reuptake activity of glutamate (Bough resistance to KA and PTZ-induced seizures,
etal., 2007). With regard to glutamatergic neuro- effects that required the opening of K ATP chan-
transmission, a recent study demonstrated a possi- nels (Gimenez-Cassina et al., 2012). While K ATP
ble alternative mechanism through which the KD channel opening induced by low adenosine
could suppress neuronal excitability. The ketone triphosphate (ATP) levels is indeed an intrigu-
bodies BHB and ACA were shown to directly influ- ing mechanism that couples the metabolic state
ence presynaptic glutamate release by directly com- of the cell to neuronal excitability, the fact that
peting with Cl for allosteric activation of vesicular the KD and ketone bodies actually increase ATP
glutamate transporters, resulting in diminished production needs to be reconciled with the K ATP
release of glutamate (Juge etal.,2010). channel hypothesis (DeVivo etal., 1978; Kim do,
In the same study, direct application of the Vallejo, & Rho, 2010; Ma, Berg, & Yellen,2007).
potassium channel blocker 4-aminopyridine to Increases in the levels of the purine nucleo-
rat brain in vivo evoked seizures with concurrent tide adenosine may also potently modulate neu-
secretion of glutamate, and these effects were ronal activity. Adenosine is produced from ATP
blocked by ACA (Juge etal.,2010). and itself produces antiseizure effects through
Despite these observations, whether activation of inhibitory adenosine A1 receptors
ketone bodies exert direct effects on excita- (A1R) (Masino etal., 2012). As the KD increases
tory or inhibitory neurotransmission remains cellular levels of ATP (DeVivo et al., 1978; Kim
controversial. In the hippocampus, BHB and do, Vallejo, & Rho, 2010), this may result in
ACA did not acutely affect GABAA, -amino- elevated levels of adenosine and subsequent
3-hydroxy-5-methylisoxazole-4-propionate, enhanced activation of A1R. In mice, targeted
N-methyl-D-aspartate, kainate, or glycine recep- deletion of A1R receptors (i.e., A1R+/- and A1R
tors (Thio, Wong, & Yamada, 2000). However, -/-) or increased expression of adenosine kinase
ACA and BHB were later shown in vitro to reduce (Adk-Tg), an enzyme that enhances clearance of
the spontaneous firing rate of GABAergic neu- adenosine, causes spontaneous electrographic
rons in the substantia nigra pars reticulata, a seizures (Masino etal., 2011). A3-week treatment
putative subcortical seizure gate, and this action with a KD led to decreased electrographic sei-
was dependent on opening of ATP-sensitive zures in Adk-Tg and A1R+/- mice but not in ani-
potassium (K ATP) channels and GABAB receptor mals missing A1R receptors (A1R-/-), indicating
activation (Ma, Berg, & Yellen, 2007). The same that adenosine may be an important mediator of
group then showed that the open probability of the KDs antiseizure effects (Masino etal.,2011).
K ATP channels in the hippocampus in vitro was
enhanced in the presence of BHB (Tanner etal., Bioenergetic and Mitochondrial
2011). Further support for ketone body effects on Changes
neuronal excitability was recently demonstrated Pathological changes in mitochondrial energy
in sympathetic neurons in vitro. Here, BHB was metabolism and reactive oxygen species (ROS)
shown to be an agonist of the free FA receptor 3, production are known to occur with epilep-
through which inhibition of N-type Ca2+ channels togenesis, and intriguingly the KD has been
was documented (Won etal.,2013). Interestingly, found to profoundly affect these processes
the short-chain FA acetate, proprionate, and (Rowley & Patel, 2013). In addition to enhancing
butyrate are known agonists of free FA receptor 3. energy reserves, ATP levels, and the expression
Another link between the KD and K ATP chan- of many enzymes involved in multiple metabolic
nels was recently revealed. BCL-2-associated pathways in the mitochondria, the KD has also
agonist of cell death (BAD), a member of the been shown to increase mitochondrial biogen-
Bcl-2 proteins that govern apoptotic cell death, esis in the hippocampus (Bough et al., 2006).
was found to mediate a switch in the oxidative Additionally, multiple studies have demonstrated
Ketogenic Diets and Mitochondria and Neurological Diseases 255

elevated antioxidant activity, diminished produc- capacity can critically affect apoptosis, neuronal
tion of ROS, and decreased ROS-induced dam- excitability, and seizure susceptibility.
age with the KD (Masino & Rho, 2012; Milder &
Patel,2012). Antioxidant Activity, Reactive
Figure 15.3 summarizes the substrates and Oxygen Species, and theRedoxState
pathways linking mitochondrial redox changes Several studies indicate that the KD decreases
to the TCA cycle and respiratory chain func- the production of ROS and limits ROS-mediated
tion. Impairment of mitochondrial bioenergetics damage, possibly by enhancing antioxidant

Figure 15.3 Mitochondrial function and neuronal excitability. Various aspects of the mitochondria
can lead to impairment of its bioenergetic capacity affecting neuronal excitability, apoptosis, and an increase
in seizure susceptibility. O2 production by complex Iand III of the electron transport chain (ETC) leads to the
production of ONOO in a reaction with NO, and H 2O2 through dismutation by the antioxidant MnSOD. H2O2 is
membrane-permeable and able to diffuse out of the mitochondria causing widespread oxidative damage. Excessive
O2 production also damages Fe-S containing enzymes involved in the tricarboxylic acid cycle such as aconitase.
OH can be formed from H2O2 through Fenton chemistry and lead to further oxidative damage of macromol-
ecules such as ETC complexes and mtDNA. Oxidative damage to mtDNA can lead to increased mutation rates
and a decrease in ETC subunit expression encoded by the mitochondrial genome. Alterations in the redox status of
GSH/GSSG and CoASH/CoASSG can cause an inability to protect against the deleterious effects of reactive oxy-
gen species. Modification of neurotransmitter biosynthesis within the mitochondria can affect levels of neuronal
excitability/inhibition. Oxidative damage to these targets can result in increased neuronal excitability resulting
from decreased mitochondrial membrane potential and adenosine triphosphate (ATP) levels affecting the Na+/
K+-ATPase and the release of cytochrome C, leading to apoptosis. mNa+C2+E=mitochondrial sodium calcium
exchanger; mCU = mitochondrial calcium uniporter; mNICE = mitochondrial sodium independent calcium
exchanger; MPT=mitochondrial permeability transition pore; GSH=glutathione; GSSG=glutathione disulfide;
CoASH=coenzyme A; CoASSG=coenzyme Aglutathione disulfide; GR=glutathione reductase; GPx=glu-
tathione peroxidase; cyto C=cytochrome C; m=mitochondrial membrane potential.
Reprinted with permission:Waldbaum S, Patel M.Mitochondria, oxidative stress, and temporal lobe epilepsy. Epilepsy Res 2010
Jan;88(1):2345. Elsevier.
256 Part III:Homeostatic Manipulators

activity. Rats fed the KD for 8 weeks showed and this was associated with increased activity
tissue-specific alterations in lipid peroxida- of NAD(P)H:quinone oxidoreductase (NQO), a
tion, total antioxidant capacity, and antioxi- prototypical Nrf2 target. Despite a depletion of
dant enzyme activities (Ziegler et al., 2003). GSH in liver homogenates at various time-points
Specifically, total antioxidant capacity was ele- (3days, 1 week, 3 weeks), levels of reduced coen-
vated in the hippocampus, lipid peroxidation was zyme A, a measure of mitochondrial antioxidant
increased in the cerebellum, and glutathione per- capacity, was decreased at 3days but elevated after
oxidase activity was enhanced in the hippocam- 3 weeks. Likewise, in the liver, nuclear extracts
pus (Ziegler etal.,2003). demonstrated increased Nrf2 expression after 1
The KD has also been shown to change the and 3 weeks on the diet, accompanied by eleva-
mitochondrial production of hydrogen perox- tions in both NQO activity and the expression
ide (H2O2) as a function of treatment duration. of Nrf2 target heme oxygenase-1 (HO-1) after 3
A decrease in substrate-driven mitochondrial weeks on the diet (Milder, Liang, & Patel, 2010).
H2O2 production was shown in rats after 3 Interestingly, a recent study found that increas-
weeks on the diet, and this was associated with ing Nrf2 expression in a rat model of temporal
increased mitochondrial glutathione (GSH) lev- lobe epilepsy decreased spontaneous seizures
els, depletion of which is known to occur with (Mazzuferi etal.,2013).
seizures (Jarrett et al., 2008). The increase in Acute application of the ketone bodies BHB
GSH was associated with elevated activity of and ACA in hippocampal slices enhanced cata-
the rate-limiting enzyme in GSH biosynthesis, lase activity in response H2O2 (Kim do, Vallejo, &
glutamate cysteine ligase (GCL), and enhanced Rho, 2010) and decreased oxidation of carboxy-
expression of the GCL catalytic subunit, GCLC, H2DCFDA, a dye often used as an indicator of
and modulatory subunit, GCLM, in rats fed the intracellular ROS (Maalouf & Rho, 2008). In iso-
KD (Jarrett etal.,2008). lated mitochondria, ACA and BHB have been
The increase in GSH and levels of the GCL shown to decrease ROS levels in response to gluta-
subunits GCLC and GCLM observed by Jarrett mate by enhancing oxidation of NADH (Maalouf
etal. (2008) prompted an investigation to exam- etal., 2007). Additionally, ACA and BHB reduced
ine the role of NF E2-related factor 2 (Nrf2), as mitochondrial ROS both basally and in response
activation of this redox-sensitive transcription to the ATP synthase inhibitor oligomycin (Kim do
factor is the primary mechanism that induces etal.,2007).
this antioxidant pathway (Milder, Liang, & A possible mechanism mediating the decrease
Patel, 2010). Nrf2 is activated by cellular stress in mitochondrial ROS production with the KD and
and initiates transcription of a diverse set of ketone bodies is enhanced expression of uncou-
genes, such as antioxidant defense, drug trans- pling proteins (UCP). Increased activity of UCP
porters, metabolic enzymes, and transcription can diminish the mitochondrial membrane poten-
factors, by binding to the antioxidant or electro- tial (), resulting in a decrease in ROS production,
phile response elements (Suzuki, Motohashi, & and this has been associated with increased resist-
Yamamoto, 2013). This study demonstrated that ance to KA-induced seizures (Sullivan etal.,2003).
production of mitochondrial-derived H2O2 was Additionally, FA can induce increases in UCP
initially enhanced after 1day on the diet but was expression possibly through enhanced activity
significantly decreased at the 3-week time-point of transcription factors, such as the peroxisome
(Milder, Liang, & Patel,2010). proliferator-activated receptor (PPAR) and the
This initial increase in H2O2 was accom- forkhead box (FOXO) families of transcription
panied by an elevation in the lipid peroxida- factors (Azzu & Brand, 2010). In mice fed the KD,
tion product 4-hydroxy-2-nonenal (4-HNE), UCP activity was enhanced, and this was associ-
both of which stimulate Nrf2 activity through ated with increased levels of UCP2, UCP4, and
oxidation of the inhibitory binding partner UCP5 in the hippocampus (Sullivan etal., 2004).
Kelch-like ECH-associated protein 1, result- Additionally, ROS productionassessed in the
ing in the release and nuclear translocation of presence of oligomycin to maximize was
Nrf2 (Sekhar, Rachakonda, & Freeman, 2010). reduced in mice fed the KD (Sullivan etal.,2004).
The acute rise in H2O2 and 4-HNE with the KD Recently, BHB was shown to be an inhibitor
coincided with increased hippocampal nuclear of class I histone deacetylases (HDAC) in vitro
expression of Nrf2 after 1 week on the KD, and in vivo (Shimazu etal., 2013), and this activity
indicative of enhanced Nrf2 activation. Levels of was associated with increased resistance to oxida-
Nrf2 remained elevated after 3 weeks on the KD, tive stress. Specifically, BHB increased acetylation
Ketogenic Diets and Mitochondria and Neurological Diseases 257

of histone H3 lysine 9 and histone H3 lysine 14 seizure control. In vitro application of 2-deoxy-D-
and enhanced transcription of genes regulated by glucose (2-DG), an inhibitor of phosphoglu-
FOXO3A, including the antioxidant enzymes man- cose isomerase, resulted in antiseizure effects
ganese superoxide dismutase and catalase. Further, against the GABA A receptor antagonist bicucul-
BHB (administered in vivo for 24 hours via an line, 4-aminopyridine, and increased extracel-
osmotic pump) decreased protein carbonylation, lular K+ (Stafstrom etal., 2009). Additionally, in
4-HNE, and lipid peroxides in the kidney. Although vivo administration of 2-DG reduced seizures
the authors did not report such effects in neuronal induced by audiogenic and 6Hz stimulation in
tissue or cells, it is possible that direct inhibition of mice (Stafstrom et al., 2009) and chronically in
HDACs and the ensuing transcriptional changes a rat kindling model (Garriga-Canut etal., 2006;
may mediate some of the antioxidant effects known Stafstrom etal., 2009). The antiseizure effects of
to occur in the brain with theKD. 2-DG may be partially mediated by changes in
the expression of genes encoding brain-derived
Mitochondrial Permeability neurotrophic factor and its receptor TrkB, both
Transition of which are regulated by the activity of the tran-
ACA and BHB both blocked neuronal death in scription factor neural restrictive silencing factor
response to diamide, a mitochondrial permeabil- (NRSF), which represses transcription by bind-
ity transition (mPT) activator, in a mechanism ing to the neuron restrictive silencing element
independent of oxidative stress. Additionally, ACA (NRSE) in promoter regions (Garriga-Canut
and BHB mimicked the effects of the mPT blocker, et al., 2006). NRSF transcriptional repression
cyclosporin A, as all three increased the threshold was enhanced by 2-DG, and this was associ-
for calcium-induced mPT opening (Kim do etal., ated with a reduction in acetylation and an
2007). More recently, it was shown that prolonged increase in methylation of histone H3 lysine 9
in vitro seizure-like activity induced with at the NRSE promoter, epigenetic modifications
low-Mg2+ in rat glio-neuronal cocultures resulted associated with suppression of gene transcrip-
in depolarization of and mPT opening, with tion. NRSF-mediated repression required an
subsequent cell death (Kovac etal.,2012). interaction with the transcriptional corepressor
These effects were reversed by CsA, a known carboxyl-terminal binding protein that was dis-
inhibitor of mPT. Despite these observations, the rupted with increasing concentrations of NADH,
causal relationships between mPT, the KD, and a cofactor that is elevated upon increased gly-
epilepsy have yet to be clearly delineated. colytic flux. This suggests that the antiseizure
actions of 2-DG may be mediated by a decrease
Glycolytic Restriction/Diversion in cytosolic and nuclear levels of NADH and
A key feature of the KD is a relative reduction in subsequent influences on histone modifications
glycolysis and an increase in nonglucose sources (Garriga-Canut etal.,2006).
of fuel through the oxidation of FA and ketone Interestingly, changes in histone acetylation
bodies, which ultimately feed the TCA cycle have also shown to be mediated by the activ-
through a process known as anaplerosis (i.e., the ity of ATP-citrate lyase (ACL), which converts
replenishing of depleted metabolic cycle inter- glucose-derived citrate to acetyl-CoA (Wellen
mediates). Glycolytic restriction is thought to be etal.,2009).
an important mechanism mediating the antisei- Enhanced flux of acetyl-CoA from ACL
zure properties of the KD. As mentioned, CR has increased acetylation of H3, and this resulted in
been shown in a mouse model of epilepsy to ren- increased expression of glycolytic enzymes. This
der antiseizure, and possibly antiepileptogenic, effect was found to be specific for acetyl-CoA
effects (Mantis etal.,2004). produced from glycolysis, as changes in histone
The earliest clinical observation supporting acetylation did not occur in cells supplemented
this notion is the rapid reversal of seizure con- with FA under glucose-replete conditions
trol upon ingestion of carbohydrates or glucose (Wellen etal.,2009).
in patients on the KD (Huttenlocher, 1976). Acetyl-CoA has yet another role as the
Additionally, studies utilizing labeled metabolic cofactor for acetylation of the -lysine residues
precursors have shown reduced oxidative metab- on histones and nonhistone enzymes by lysine
olism of glucose (Melo, Nehlig, & Sonnewald, acetyltransferases (KAT) (Albaugh, Arnold, &
2006; Yudkoff etal.,2005). Denu, 2011). Three families of KATs have been
Use of glycolytic inhibitors has enabled fur- found to alter the acetylation state of a diverse
ther insights into this possible mechanism for group of substrates with varying influences
258 Part III:Homeostatic Manipulators

on cellular functions (Albaugh, Arnold, & levels of BHB, cortisol, and free fatty acids (Fraser
Denu, 2011). However, these three families etal., 2003). Most PUFA were also increased in the
of KATs have primarily been localized to the serum, including the triglyceride and phospholipid
nucleo-cytoplasmic compartments, and a mito- forms of linoleic acid, arachidonic acid (AA), and
chondrial KAT has yet to be confirmed despite docosahexaenoic acid (DHA); triglyceride levels
estimates that 65% of mitochondrial proteins are of stearic acid; and palmitic acid in phospholipids
acetylated, especially those involved in energy (Fraser etal., 2003). Additionally, these alterations
metabolism and antioxidant defenses (Albaugh, in FA were associated with seizure reduction in
Arnold, & Denu, 2011; Hebert et al., 2013). 78% of the children. Interestingly, seizure control
Recently, it was shown that the high alkaline pH was correlated with circulating AA levels but not
of the mitochondria in the presence of elevated EEG changes (Fraser etal.,2003).
levels of acetyl-CoA allows for nonenzymatic Whether PUFA supplementation can ren-
acetylation to occur in vitro (Wagner & Payne, der antiseizure effects remains controversial.
2013). This has led to speculation that compart- Supplementation with 5 g of omega-3 PUFA for
mental differences in acetyl-CoA concentra- 6months appeared to reduce the frequency and
tion can direct changes in acetylation (Albaugh, severity of seizures in a small observational study
Arnold, & Denu,2011). (Schlanger, Shinitzky, & Yam, 2002). However, in
Fructose-1,6-bisphosphate (FBP) also inhibits a 12-week randomized, placebo-controlled paral-
glycolysis by diverting the metabolism of glucose lel group study in adults with epilepsy, adminis-
to the pentose phosphate pathway and also affords tration of eicosapentaenoic (EPA) and DHA (1 g
broad antiseizure effects in vivo (Lian, Khan, & EPA, 0.7 g DHA daily) reduced seizure frequency
Stringer, 2007). FBP was shown to provide the for the first 6 weeks of treatment, but this effect
greatest protection when compared to the KD, did not persist despite sustained increases in
2-DG, and the commonly prescribed ASD valproic DHA and EPA and decreases in AA and linoleic
acid in the pilocarpine-, KA-, and PTZ-induced acid in the plasma (Yuen etal., 2005). In a further
seizure models in rats (Lian, Khan, & Stringer, randomized, blinded trial of PUFA supplementa-
2007). Surprisingly, the KD did not demonstrate tion (EPA plus DHA, 2.2 mg/day in a 3:2 ratio over
antiseizure activity in this study, and 2-DG was 12 weeks) in adults with uncontrolled epilepsy,
only protective in the pilocarpine model, whereas there was a lack of clear efficacy when compared
valproic acid demonstrated limited protection in to placebo (Bromfield et al., 2008). However, it
all three models. Co-administration of lactate, to is unclear whether different doses, duration, or
relieve the glycolytic inhibition induced by either ratios of PUFA might have been effective.
2-DG or FBP, abolished 2-DG-mediated protec- In rats fed the KD, there were marked reduc-
tion but only slightly reduced the antiseizure tions of PUFA levels in plasma and adipose tis-
activity of FBP (Lian, Khan, & Stringer, 2007). sue but enhanced mobilization of the PUFA AA
This indicates that the effects of FBP are not solely and DHA to the liver and brain (Taha, Ryan, &
due to inhibition of glycolysis but may possibly Cunnane, 2005). This was accompanied by an ini-
be a result of enhanced flux through the pentose tial increase in the plasma levels of BHB, followed
phosphate pathway. by a reduction by day 10 of the diet. Following a
calorie-restricted KD, mRNA expression of the
Fatty Acid Oxidation rate-limiting enzyme for ketone body production,
Intake of a high-fat diet, such as the KD, inherently HMG-CoAS2, was found to increase in both the
increases the rate of FAO, and this also changes liver and the brain, in contrast to isocaloric stand-
the levels and types of polyunstaturated fatty acids ard chow, which showed enhanced expression only
(PUFA) in the circulation, liver, and brain. These in the liver (Cullingford, Eagles, & Sato,2002).
alterations in PUFA are a result of endogenous Collectively, these and other animal studies
production and export of these FA into the circu- suggest that diet-induced changes in brain con-
lation since large quantities of these lipids are not tent and metabolism of PUFA may be impor-
a typical component of the KD (Fraser etal., 2003). tant contributors to antiseizure effects of the KD
PUFA are known to possess neuroprotective prop- (Taha, Burnham, & Auvin,2010).
erties (Michael-Titus & Priestley, 2013); therefore
it has been speculated that this lipid species may Bioenergetics Reserve and
mediate the antiseizure effects of theKD. Mitochondrial Respiration
In pediatric patients with epilepsy, 3 to 4 weeks Alterations in bioenergetic metabolites by the
of KD treatment led to elevations in circulating KD have been shown in several animal studies.
Ketogenic Diets and Mitochondria and Neurological Diseases 259

DeVivo and colleagues first demonstrated sig- pathways, it is also likely that levels of meta-
nificant increases in ATP and the ATP/ADP ratio bolic intermediates may be altered. Specifically,
(and other parameters of bioenergetic reserve the KD may enhance neuronal ATP production
capacity) in the brain of rats fed the KD for 3 through refilling of TCA cycle intermediates
weeks (DeVivo etal., 1978). Further, decreases in from the increase in acetyl-CoA levels from the
creatine with no change in phosphocreatine were oxidation of ketone bodies. These actions may
noted in this study. However, while a later study contribute to antiseizure effects. In this regard,
failed to confirm elevations in ATP and ATP/ADP the anaplerotic substrate triheptanoin has been
ratios, the KD was shown to increase the ratio shown to raise seizure thresholds in mouse sei-
of phosphocreatine to creatine (Bough et al., zure models and to help restore levels of TCA
2006). In vitro studies have further supported intermediates (Hadera et al., 2014; Willis et al.,
the concept that KD metabolites can enhance 2010). Further, replenishment of the TCA cycle
bioenergetic function. For example, BHB and may counter seizure-induced energy failure
ACA prevented depletion of ATP in hippocam- and augment inhibitory neurotransmission
pal slices in response to H2O2 and inhibitors of (Kovac, Abramov, & Walker, 2013). For example,
complex I (e.g., rotenone) and complex II (e.g., -ketoglutarate, which serves a dual function as
3-nitropropionic acid) of the electron transport a TCA cycle intermediate and as the immediate
chain (ETC) (Kim do, Vallejo, & Rho, 2010). In a precursor to glutamate, which can be converted
mouse model of global cerebral ischemia, admin- to GABA, directly links anaplerosis to neuronal
istration of BHB immediately following bilateral excitability (Kovac, Abramov, & Walker,2013).
common carotid artery ligation ameliorated the
decline in tissue levels of ATP (Suzuki et al., A C T I VAT I O N O F
2001). Thus, collectively, there is both in vivo and E N E R G Y- S E N S I N G
in vitro evidence for the KD and ketone bodies S I G N A L I N G PAT H WAY S
enhancing ATP production inbrain. It is well established that multiple signaling path-
Consistent with the KDs favorable effects on ways have evolved to sense states of diminished
energy production, changes in the expression of energy stores, such as seen during exercise and
enzymes involved in metabolic pathways have CR, and that activation of these pathways results
also been reported. Two studies revealed changes in restoration of cellular homeostasis and integ-
in gene expression in the hippocampus of mice rity (Libert & Guarente, 2013; Mattson, 2012).
and rats fed the KD (Bough et al., 2006; Noh Data regarding the association of the KD with
et al., 2004), and both demonstrated enhanced these energy-sensing pathways are limited, but
expression of numerous enzymes involved in the similarities between the protective mecha-
mitochondrial metabolism (Noh et al., 2004), nisms evoked by the KD and those known to be
such as the TCA cycle and oxidative phosphoryl- influenced by these pathways indicates substan-
ation (Bough etal., 2006). However, there was not tial overlap in their mechanisms and downstream
complete concordance with respect to the gene effects.
expression changes for all metabolic enzymes
(Bough etal.,2006). Peroxisome Proliferator-Activated
Mitochondrial respiration and ETC activity Receptors
have been shown to be elevated in animals fed the The PPAR family of nuclear receptors represents
KD or with in vitro application of ketone bodies. one signaling pathway that may mediate some
BHB increased oxygen consumption and ATP of the beneficial effects of the KD (Sampath &
production in purified mitochondria in the pres- Ntambi, 2005). PPARs are transcription factors
ence of two inhibitors of complex I, 1-methyl-4- that bind to the PPAR response element (PPRE)
phenyl-1,2,3,6-tetrahydropyridine (MPTP) and of promoter regions. They include three iso-
rotenone (Tieu etal., 2003). Further, in a model of forms: PPAR, PPAR, and PPAR/ (Sampath
glutamate excitotoxicity, ACA and BHB increased & Ntambi, 2005). FA are endogenous ligands
complex I-driven oxygen consumption through for PPARs, and PPAR agonists are used in the
increased NADH oxidation (Maalouf etal.,2007). treatment of lipid disorders such as type II dia-
betes mellitus and high cholesterol (Sampath &
TCA Cycle Effects and Anaplerosis Ntambi, 2005). The induction of FAO is a known
As numerous studies have shown that the KD can metabolic effect of PPAR activation, which ren-
increase ATP levels, bioenergetic capacity, and ders these transcription factors an attractive
transcription of enzymes in energy-producing mediator for the clinical effects of theKD.
260 Part III:Homeostatic Manipulators

Fenofibrate, a PPAR agonist, was shown to KD, but this was not associated with enhanced
exhibit antiseizure properties similar to the KD activation in the brain (McDaniel etal.,2011).
(Porta etal., 2009). Rats fed the KD and a diet con-
taining fenofibrate showed increased threshold to Mammalian Target ofRapamycin
PTZ-induced seizures and an increased latency The mammalian target of rapamycin (mTOR) is
to spike-and-wave discharges lasting longer than another protein kinase that exerts multiple effects
5 minutes in the lithium-pilocarpine model, on energy metabolism through the actions of two
compared with control-fed animals (Porta etal., distinct complexes, mTOR complex 1 (mTORC1)
2009). In a separate study, acute injection of the and mTOR complex 2 (mTORC2) (Johnson,
PPAR agonist FMOC-L-leucine protected adult Rabinovitch, & Kaeberlein, 2013). Unlike the PPARs
magnesium-deficient mice against audiogenic and AMPK, however, mTOR is activated during
seizures, and this was reversed by administra- high-energy states, and this results in an induction
tion of the PPAR antagonist GW9662 (Maurois of protein synthesis and mRNA translation, among
et al., 2008). In contrast, FMOC-L-leucine did other actions that promote growth and cellular
not exhibit antiseizure activity in another sei- proliferation (Johnson, Rabinovitch, & Kaeberlein,
zure model, the 6 Hz test, and another PPAR 2013). Importantly, AMPK inhibits mTORC1
ligand, rosiglitazone, did not reduce audiogenic through direct phosphorylation of both a subunit
seizures (Maurois et al., 2008). Rosiglitazone of this complex and an upstream regulatory protein
has been shown to reduce oxidative stress in the (Johnson, Rabinovitch, & Kaeberlein, 2013). In rats,
lithium-pilocarpine model of epilepsy, and this the KD inhibited activation of the mTOR path-
was associated with an amelioration of neuronal way in the liver and brain (McDaniel etal., 2011).
death in the hippocampus and an enhancement Additionally, in the KA model, enhanced activation
of SOD activity and levels of GSH, as well as a of mTOR was found in the hippocampus of rats fed
reduction in the Nrf2 target HO-1 (Yu et al., a standard diet, and this effect was blocked after
2008). Further, the PPAR-selective agonist 7days on the KD (McDaniel etal., 2011). Given that
ciprofibrate was shown to selectively increase mTOR inhibition is believed to retard the processes
mRNA levels of HMG-CoAS2, the rate-limiting of epileptogenesis (Wong, 2013) and that the KD
enzyme in the production of ketone bodies, and can decrease mTOR signaling, it is conceivable that
that of two other known PPAR targets, acyl CoA metabolism-based treatments could render antiepi-
oxidase and medium chain acyl-CoA dehydroge- leptogenic effects.
nase (Cullingford, Dolphin, & Sato,2002).
Sirtuins
AMP-ActivatedKinase The sirtuins (SIRT17) are a family of NAD-
AMP-activated kinase (AMPK) is induced by dependent enzymes known to influence mul-
low levels of AMP and inhibited by high con- tiple aspects of cellular homeostasis, including
centrations of ATP; therefore, AMPK functions metabolism and antioxidant activity. They were
as a direct sensor of the energy state of the cell. originally classified as Class III HDACs; how-
Activation of AMPK leads to the phosphorylation ever, it is now known that individual isoforms
of multiple substrates, resulting in concurrent also demonstrate desuccinylase, demalonylase,
inhibition of anabolic pathways and increases and ADP-ribosyltransferase activities (Libert &
in catabolic metabolism to enhance the produc- Guarente, 2013). The sirtuins are direct sensors of
tion of ATP (Hardie & Sakamoto, 2006). These the energetic and redox state of the cell, principally
effects are known to include an upregulation of through NAD+, which is required for their cata-
FAO and increased mitochondrial biogenesis lytic activity. Additionally, the beneficial effects
(Hardie & Sakamoto, 2006). Similar to PPARs, of CR may be mediated by the sirtuins, as well
agonists of AMPK are used to treat type II diabe- as the AMPK and mTOR pathways (Guarente,
tes mellitus and metabolic syndrome (Hardie & 2013; Houtkooper, Williams, & Auwerx, 2010).
Sakamoto,2006). The diversity and abundance of their histone and
The KD increased AMPK activity in liver nonhistone substrates, as well as their specific
and skeletal muscle of mice, and this was associ- localization within different cellular compart-
ated with increased gene expression of enzymes ments, reflect the extent of their broad influence on
involved in FAO and a reduction in transcript lev- energy metabolism (Guarente, 2013; Houtkooper,
els of enzymes in lipid synthesis pathways in the Williams, & Auwerx, 2010). Collectively, the sir-
liver (Kennedy etal., 2007). Increased activation tuins are known to enhance oxidative phosphoryla-
of AMPK was also found in the liver of rats on the tion and reduce glycolysis, in addition to providing
Ketogenic Diets and Mitochondria and Neurological Diseases 261

increased resistance to oxidative stress (Libert & of the ETC and elevations in the production of
Guarente, 2013). The substantial overlap in the mitochondrial-derived ROS, which can alter
activities of these regulators of cellular homeosta- cellular signaling. Additionally, given the simi-
sis with the known mechanisms of the KD suggests lar mechanisms of these processes in neurode-
possible involvement of the sirtuins in the effects of generative diseases, and the beneficial effects of
this dietary modification. Certainly, the observa- the KD and ketone bodies on multiple aspects
tion the BHB is a ClassIHDAC inhibitor (Shimazu of mitochondrial function, there has been an
etal., 2013)is compelling in this regard. increase in the use of metabolism-based treat-
One study examined levels of SIRT1 and SIRT3 ments for neurological diseases (Baranano &
in interscapular brown adipose tissue from mice Hartman, 2008; Stafstrom & Rho,2012).
on the KD for 1 month (Srivastava et al., 2013).
Protein expression of SIRT1 was elevated, but lev- Alzheimers Disease
els of SIRT3 were reduced compared to mice on AD is characterized by an accumulation of
a standard diet (Srivastava et al., 2013). A sepa- neurofibrillary tangles and amyloid plaques
rate study examining gene expression changes in comprised of misfolded aggregates of tau and
the hippocampus of rats fed the KD for 3 weeks amyloid- (A) proteins, respectively, result-
found increased levels of Sirt5, although this was ing in neuronal death (Mattson, 2012). AD is
reported in the supplemental results section and an age-related neurodegenerative disease and is
not discussed by the authors (Bough etal.,2006). known to arise from both genetic and environ-
Further, SIRT3 and SIRT5, major regulators of mental influences. Older adults with AD are at
mitochondrial energy metabolism, exhibit extensive increased risk for the development of epilepsy,
regulation of the acetylation and succinylation state and similar mechanisms, such as deficits in
of numerous enzymes in the TCA cycle, FAO, and mitochondrial energy metabolism and eleva-
ketogenesis (Rardin, Newman, etal., 2013; Rardin, tions in oxidative stress, are thought to contrib-
He, et al., 2013). Interestingly, activity of HMG- ute to both pathological states (Mattson, 2012;
CoAS2, the rate-limiting enzyme in ketogenesis, is Palop & Mucke, 2009). Therefore, there is grow-
regulated by both deacetylation through SIRT3 and ing interest in the use of the KD to delay the
desuccinylation by SIRT5, two posttranslational progressionofAD.
modifications thought to inhibit enzymatic activ- In fasted patients with AD or mild cogni-
ity (Rardin, Newman, etal., 2013; Rardin, He, etal., tive impairment (MCI), acute ingestion of a
2013; Shimazu etal., 2010). Finally, separate studies MCT drink increased BHB levels and cogni-
in Sirt3-/- and Sirt5-/- mice demonstrated reduc- tive function compared to placebo (Reger etal.,
tions in the circulating levels of BHB under fasting 2004). Further, in a separate study of older
but not basal conditions (Rardin, He, et al., 2013; adults with MCI, a low-carbohydrate (5%10%
Shimazu etal.,2010). of calories) diet for 6 weeks enhanced a meas-
ure of verbal memory versus preintervention
E V I D E N C E A N D I M P L I C AT I O N S scores, whereas there was no change in those
FOROTHER NEUROLOGICAL assigned to the high-carbohydrate (50% of calo-
DISORDERS ries) diet (Krikorian et al., 2012). Additionally,
the improvement in memory in those on the
Mitochondrial Dysfunction low-carbohydrate diet was positively correlated
inNeurological Diseases with levels of urinary ketones (Krikorian et al.,
Mitochondrial dysfunction has recently been 2012). Finally, Henderson and colleagues treated
recognized as a common mechanism under- 152 patients having mild to moderate AD daily
lying many neurological disorders (Cali, with 20 g of MCT for 3months and found that
Ottolini, & Brini, 2012; de Castro, Martins, & MCT-treated patients (lacking the APOE4 allele)
Tufi, 2010; Lin & Beal, 2006; Pathak, Berthet, achieved significantly higher scores on the
& Nakamura, 2013). The role for mitochondrial cognitive subscale of the Alzheimers Disease
energetics and signaling as important media- Assessment Scale at two different time-points
tors of neuronal death, a common feature of versus placebo-treated controls. Interestingly,
neurodegenerative diseases (Johri & Beal, postdose serum BHB levels correlated posi-
2012), is increasingly being understood. Specific tively with improvement in scores (Henderson
pathological changes include a reduction in etal.,2009).
ATP production via oxidative phosphoryla- However, in a mouse model of AD, consump-
tion or direct inhibition of specific complexes tion of a KD high in saturated FA reduced total
262 Part III:Homeostatic Manipulators

levels of A but did not affect cognitive func- coined the Warburg effect (Warburg, 1956).
tion (Van der Auwera et al., 2005). In vitro, Defects in mitochondrial function resulting
BHB reduced hippocampal neuronal cell death in diminished oxidative phosphorylation are
from exposure to the proteolytic fragment of the thought to be main contributors to cancer cell
-chain of the amyloid precursor protein, A1-42 metabolism. Since cancer cells preferentially use
(Kashiwaya etal., 2000). Additionally, it has been glucose for energy, and the KD reduces glycolytic
shown that a ketone ester diet lessens amyloid flux and enhances oxidative metabolism, high-fat
and tau pathologies and improves learning and KDs may represent potentially viable treat-
memory performance in a mouse model of AD ments to limit oncogenesis (Seyfried etal., 2011).
(Kashiwaya etal., 2013). Taken together, there are Indeed, this conceptual approach was demon-
an increasing number of studies pointing to the strated in a compelling manner in a mouse astro-
neuroprotective benefits of the KD and its meta- cytoma model, indicating that plasma glucose is
bolic substrates. an accurate predictor of tumor growth more than
the specific origin of dietary calories (Seyfried
Parkinsons Disease etal.,2003).
The hallmark neuropathological finding in PD is In a later study employing a mouse model of
the degeneration of dopaminergic neurons in the malignant glioma, the KD reduced tumor growth
substantia nigra. Amain mechanism thought to and increased survival, and this was associated
contribute to this excitotoxic cell death is defects with a decrease in tissue levels of ROS (Stafford
in complex I of the ETC (Johri & Beal, 2012). et al., 2010). Additionally, the KD induced gene
Hence it follows that metabolism of ketone bod- expression changes in the tumor tissue to more
ies may circumvent this deficit and allow for oxi- closely resemble the pattern found in the normal
dative phosphorylation to occur (Maalouf etal., brain. Further, the KD also caused an upregu-
2007). In PD patients treated with the KD for 4 lation of enzymes involved in oxidative stress
weeks, scores on Unified Parkinsons Disease resistance, such as glutathione peroxidase 7 and
Rating Scale improved, although this study had peroxiredoxin 4, in tumor tissue but not normal
a very small sample size and did not include con- specimens (Stafford etal.,2010).
trols (Vanitallie etal., 2005). Moreover, this study The same research group later showed in a
showed that switching monounsaturated FA and similar model that administration of KetoCal, a
PUFA for saturated fats prevented the increase in commercially available 4:1 KD, greatly increased
cholesterol expected from intake of a high-fat diet survival (Abdelwahab et al., 2012). When
(Vanitallie etal., 2005). In a prospective study of KetoCal was given in conjunction with radiation
older adults, increased consumption of total fats, therapy, a supra-additive effect was found as vis-
monounsaturated FA, and PUFA were associated ualization of tumor cells diminished below the
with a lower risk of PD (de Lau etal.,2005). levels of detection in 9 out of 11 animals. Further,
Following treatment with MPTP, an inhibitor when these animals were placed back on a stand-
of complex Icommonly used to model PD, BHB ard diet, no regrowth of tumors was found after
reduced dopaminergic neuronal cell death in 100days (Abdelwahab etal.,2012).
substantia nigra pars compacta in vivo, and this In a more recent study, expression of cytosolic
was associated with increased levels of dopamine glycolytic enzymes and mitochondrial ketolytic
and improved rotarod performance, demonstrat- enzymes were altered in sections of malignant
ing an attenuation of motor deficits (Tieu etal., gliomas compared with adjacent normal brain
2003). Additionally, BHB reduced cell death tissue from adults (Chang, Olson, & Schwartz,
in vitro in mesencephalic neurons exposed to 2013). The most common pattern observed was
1-methyl-4-phenylpyridinium, the active metab- upregulation of the cytosolic glycolytic enzymes
olite of MPTP (Kashiwaya etal.,2000). with a reduction in the mitochondrial ketolytic
enzymes in the brain tumor tissue.
BrainCancer
Cancer cells are known to undergo dramatic Neurotrauma
metabolic alterations, including a preference for Acute neurotrauma results in metabolic changes
ATP production via glycolysis and enhanced lac- in the brain, including diminished glycolysis, and
tic acid production, despite the presence of oxy- also activates excitotoxic and neuroinflamma-
gen for oxidative phosphorylation (Seyfried & tory cascades (Algattas & Huang, 2014; Babikian
Shelton, 2010). This shift in metabolism was first et al., 2010). Despite the glycolytic restriction
noted by Otto Warburg and was subsequently observed, the KDthrough a multiplicity of
Ketogenic Diets and Mitochondria and Neurological Diseases 263

other neuroprotective mechanismsmay coun- colleagues, who showed that fasting for 24 hours
ter the pathophysiological changes seen after following controlled cortical impact in rats
traumatic brain injury (TBI). In an animal model resulted in increased tissue sparing and improve-
of TBI, cortical contusion volume was reduced in ments in mitochondrial function and that these
rats fed the KD (Prins, Fujima, & Hovda,2005). effects were a result of ketones and not hypogly-
Further, infusion of BHB 3 hours after injury cemia (Davis etal.,2008).
increased cerebral uptake of BHB and amelio-
rated the injury-induced reduction in cortical Amyotrophic Lateral Sclerosis
ATP levels (Prins et al., 2004). A recent study Mitochondrial dysfunction is also thought to
in a rat model of TBI demonstrated that the KD contribute to the progression of ALS, a disease
decreased mRNA levels of Bax, a Bcl-2 protein characterized by degeneration of motor neurons
mediating apoptosis, and reduced cerebral edema in the cortex and spinal cord (Rothstein, 2009).
and apoptosis (Hu etal., 2009). Additionally, ini- In ALS mice, the KD improved motor func-
tiation of the KD 4days before insulin-induced tion, as evidenced by increased time to failure
hypoglycemia reduced neuronal death in rats on rotorod performance, and this was associated
(Yamada, Rensing, & Thio,2005). with preservation of motor neurons in the ven-
Further support for a metabolic approach tral horn of the spinal cord (Zhao et al., 2006).
toward TBI treatment is provided by Davis and In mitochondria isolated from the spinal cord of

Ketogenic Diet
LGIT MAD MCT CR

Glycolysis/ Fatty Acid Oxidation

Ketogenesis/
Ketone Bodies

mTOR AMPK PPARs Sirtuins HDACs

TAC Cycle Antioxidant Mitochondrial Histone


Ion Channels
Anaplerosis Capacity Function Acetylation

Synaptic Integrity Bioenergetics ROS

Neuroprotection

Figure15.4 Proposed mechanisms for the neuroprotective effects of the ketogenic diet (KD) and
its variants. The dietary interventions are shown in orange; the metabolic effects of the diets are shown in blue;
the energy-sensing pathways that may mediate the effects of the dietary alterations are shown in red; the cellular
effects resulting from the diets and/or the energy-sensing pathways are shown in green; the broad protective effects
of the diets and the resulting cellular effects are in cyan. LGIT=low-glycemic index treatment; MAD=modified
Atkins diet; MCT=medium-chain triglyeride diet; CR=caloric restriction; mTOR=mammalian target of rapa-
mycin; AMPK=AMP-activated kinase; PPARs=peroxisome proliferator-activated receptors; HDACs=histone
deacetylases; TCA=tricarboxylic; ROS=reactive oxygen species. Solid black lines indicate links proven in the
literature; dashed black lines represent possible but as yet unproven links. See text for further details.
264 Part III:Homeostatic Manipulators

a transgenic mouse model of ALS (SOD1-G93A), frequency; unfortunately, the small study size
addition of BHB in vitro enhanced ATP produc- and limited number of patients completing the
tion, and this effect was maintained in the pres- 3-month study prevent any definitive conclusions
ence of the complex Iinhibitor rotenone but not (Kossoff etal.,2010).
the complex II inhibitor malonate (Zhao et al.,
2006). Additionally, the preservation of ATP lev- Summary
els by BHB was associated with increased neu- The KD is a broad-spectrum therapy for multiple
ronal survival in the presence of rotenone but not forms of epilepsy in both children and adults. The
malonate (Zhao etal.,2006). utility of the KD and variations of this diet for
the treatment of a variety of neurodegenerative
Pain and Inflammation disorders suggests common central mechanisms
Multiple lines of experimental evidence suggest that restore imbalances in energy metabolism
shared fundamental mechanisms responsible for (Figure 15.4). The numerous mechanisms known
chronic pain syndromes and epilepsy, particularly to partially mediate the effects of the KD (such
the involvement of cellular membrane-bound ion as increases in FAO, reductions in glycolysis,
channels (Bialer, 2012). Specifically, both chronic enhancement of the cellular responses to oxida-
pain and epilepsy are characterized by enhanced tive stress, etc.), indicate that manipulation of
neuronal excitability, and, whatever the relevant these specific pathways may represent an attrac-
mechanisms may be, metabolic approaches tive paradigm for experimental therapeutics.
toward treatment (e.g., the KD, inhibition of gly- Specifically, bioenergetic substrates and enzymes
colysis through fasting or 2-DG, etc.) can alleviate may be desirable drug targets for the treatment
neuropathic pain (Masino & Rho, 2012; Ruskin, of many neurological diseases. Additionally, the
Kawamura, & Masino, 2009). Additionally, signaling pathways that evolved to sense the cel-
ASDs are often prescribed for chronic pain, lular energetic state and provide resistance to
further suggesting similarities in the patho- metabolic stress may provide the best means to
physiology of these two disorders (Masino & mimic the KD. Future research in this burgeon-
Ruskin, 2013). In juvenile and adult rats, the KD ing area may lead to the elucidation of additional
reduced pain, as assessed by increased latency to novel mechanisms that mediate the pleiotropic
hind-paw withdrawal in a test of thermal nocio- neuroprotective effects of theKD.
ception, and inflammation, as measured by
hind-paw swelling in response to injection of the References
immune-potentiator Freunds complete adjuvant Abdelwahab, M.G., K. E.Fenton, M. C.Preul, J. M.R ho,
(Ruskin, Kawamura, & Masino, 2009). Despite A. Lynch, P. Stafford, and A. C.Scheck. 2012. The
attaining similar levels of circulating ketone ketogenic diet is an effective adjuvant to radiation
bodies, the anti-inflammatory and hypo-algesic therapy for the treatment of malignant glioma.
effects of the KD were more prominent in juvenile PLoS One 7:e36197.
mice (Ruskin, Kawamura, & Masino,2009). Albaugh, B.N., K. M.Arnold, and J. M.Denu. 2011.
KAT(ching) metabolism by the tail:insight into
the links between lysine acetyltransferases and
Other Disorders metabolism. Chembiochem 12:290298.
There are a number of other reports suggesting Algattas, H., and J. H.Huang. 2014. Traumatic brain
that the KD (or other dietary manipulations) can injury pathophysiology and treatments: Early,
effectively treat diverse neurological disorders intermediate, and late phases post-injury. Int J
such as autism and migraine. For example, in the Mol Sci 15:309341.
BTBR mouse, an inbred model that recapitulates Azzu, V., and M. D.Brand. 2010. The on-off switches
the core behavioral features of autism spectrum of the mitochondrial uncoupling proteins. Trends
disorder and that does not exhibit altered suscep- Biochem Sci 35:298307.
tibility to seizures, the KD increased sociability Babikian, T., M. L.Prins, Y. Cai, G. Barkhoudarian,
and communication while reducing repetitive I. Hartonian, D. A.Hovda, and C. C.Giza. 2010.
behaviors (Ruskin et al., 2013). A small study Molecular and physiological responses to juve-
examined the use of the MAD in treating chronic nile traumatic brain injury:Focus on growth and
headaches in adolescents and demonstrated metabolism. Dev Neurosci 32:431441.
reduced headache severity in the three patients Bailey, E.E., H. H.Pfeifer, and E. A.Thiele. 2005. The
that completed the study (Kossoff et al., 2010). use of diet in the treatment of epilepsy. Epilepsy
However, the MAD did not reduce headache Behav 6:48.
Ketogenic Diets and Mitochondria and Neurological Diseases 265

Baranano, K.W., and A. L.Hartman. 2008. The keto- alpha-selective activator ciprofibrate upregu-
genic diet:uses in epilepsy and other neurologic lates expression of genes encoding fatty acid
illnesses. Curr Treat Options Neurol 10:410419. oxidation and ketogenesis enzymes in rat brain.
Bergqvist, A. G., J. I. Schall, P. R. Gallagher, A. Neuropharmacology 42:724730.
Cnaan, and V. A. Stallings. 2005. Fasting versus Cullingford, T.E., D. A.Eagles, and H. Sato. 2002.
gradual initiation of the ketogenic diet: A pro- The ketogenic diet upregulates expression of the
spective, randomized clinical trial of efficacy. gene encoding the key ketogenic enzyme mito-
Epilepsia 46:18101819. chondrial 3-hydroxy-3-methylglutaryl-CoA syn-
Bialer, M. 2012. Why are antiepileptic drugs used thase in rat brain. Epilepsy Res 49:99107.
for nonepileptic conditions? Epilepsia 53 Suppl Dahlin, M., A. Elfving, U. Ungerstedt, and P. Amark.
7:2633. 2005. The ketogenic diet influences the levels of
Bishop, N.A., and L. Guarente. 2007. Genetic links excitatory and inhibitory amino acids in the CSF
between diet and lifespan: Shared mechanisms in children with refractory epilepsy. Epilepsy Res
from yeast to humans. Nat Rev Genet 8:835844. 64:115125.
Bough, K. J., and J. M. Rho. 2007. Anticonvulsant Davis, L.M., J. R.Pauly, R. D.Readnower, J. M.Rho,
mechanisms of the ketogenic diet. Epilepsia and P. G.Sullivan. 2008. Fasting is neuroprotec-
48:4358. tive following traumatic brain injury. J Neurosci
Bough, K. J., J. Wetherington, B. Hassel, J. F. Pare, Res 86:18121822.
J. W. Gawryluk, J. G. Greene, R. Shaw, Y. de Castro, I. P., L. M. Martins, and R. Tufi. 2010.
Smith, J. D. Geiger, and R. J. Dingledine. 2006. Mitochondrial quality control and neurological
Mitochondrial biogenesis in the anticonvulsant disease:an emerging connection. Expert Rev Mol
mechanism of the ketogenic diet. Ann Neurol Med 12:e12.
60:223235. de Lau, L. M., M. Bornebroek, J. C. Witteman, A.
Bough, K.J., M. Paquet, J. F.Pare, B. Hassel, Y. Smith, Hofman, P. J. Koudstaal, and M. M. Breteler.
R. A. Hall, and R. Dingledine. 2007. Evidence 2005. Dietary fatty acids and the risk of
against enhanced glutamate transport in the Parkinson disease: The Rotterdam study.
anticonvulsant mechanism of the ketogenic diet. Neurology 64:20402045.
Epilepsy Res 74:232236. DeVivo, D.C., M. P.Leckie, J. S.Ferrendelli, and D.
Bough, K.J., R. Valiyil, F. T.Han, and D. A.Eagles. B.McDougal, Jr. 1978. Chronic ketosis and cere-
1999. Seizure resistance is dependent upon age bral metabolism. Ann Neurol 3:331337.
and calorie restriction in rats fed a ketogenic Fraser, D. D., S. Whiting, R. D. Andrew, E.
diet. Epilepsy Res 35:2128. A. Macdonald, K. Musa-Veloso, and S.
Bromfield, E., B. Dworetzky, S. Hurwitz, Z. Eluri, L. C. Cunnane. 2003. Elevated polyunsaturated
Lane, S. Replansky, and D. Mostofsky. 2008. A fatty acids in blood serum obtained from children
randomized trial of polyunsaturated fatty acids on the ketogenic diet. Neurology 60:10261029.
for refractory epilepsy. Epilepsy Behav 12:187190. Freeman, J. M., and E. P. Vining. 1999. Seizures
Cali, T., D. Ottolini, and M. Brini. 2012. decrease rapidly after fasting:preliminary stud-
Mitochondrial Ca(2+) and neurodegeneration. ies of the ketogenic diet. Arch Pediatr Adolesc
Cell Calcium 52:7385. Med 153:946949.
Chang, H.T., L. K.Olson, and K. A.Schwartz. 2013. Freeman, J. M., E. H. Kossoff, and A. L. Hartman.
Ketolytic and glycolytic enzymatic expression 2007. The ketogenic diet: One decade later.
profiles in malignant gliomas: Implication for Pediatrics 119:535543.43
ketogenic diet therapy. Nutr Metab (Lond) 10:47. Fusco, S., and G. Pani. 2013. Brain response to calo-
Cheng, C.M., K. Hicks, J. Wang, D. A.Eagles, and rie restriction. Cell Mol Life Sci 70:31573170.
C. A.Bondy. 2004. Caloric restriction augments Garriga-Canut, M., B. Schoenike, R. Qazi, K.
brain glutamic acid decarboxylase-65 and -67 Bergendahl, T. J. Daley, R. M. Pfender, J.
expression. J Neurosci Res 77:270276. F. Morrison, J. Ockuly, C. Stafstrom, T. Sutula,
Cotter, D. G., R. C. Schugar, and P. A. Crawford. and A. Roopra. 2006. 2-Deoxy-D-glucose reduces
2013. Ketone body metabolism and cardiovas- epilepsy progression by NRSF-CtBP-dependent
cular disease. Am J Physiol Heart Circ Physiol metabolic regulation of chromatin structure. Nat
304:H1060H1076. Neurosci 9:13821387.
Cross, J. H., and E. G. Neal. 2008. The ketogenic Gimenez-Cassina, A., J. R. Martinez-Francois,
diet:Update on recent clinical trials. Epilepsia 49 J. K. Fisher, B. Szlyk, K. Polak, J. Wiwczar,
Suppl 8:610. G. R. Tanner, A. Lutas, G. Yellen, and N.
Cullingford, T.E., C. T.Dolphin, and H. Sato. 2002. N. Danial. 2012. BAD-dependent regulation of
The peroxisome proliferator-activated receptor fuel metabolism and K(ATP) channel activity
266 Part III:Homeostatic Manipulators

confers resistance to epileptic seizures. Neuron Jarrett, S. G., J. B. Milder, L. P. Liang, and M.
74:719730. Patel. 2008. The ketogenic diet increases mito-
Greene, A. E., M. T. Todorova, R. McGowan, and chondrial glutathione levels. J Neurochem
T. N. Seyfried. 2001. Caloric restriction inhib- 106:10441051.
its seizure susceptibility in epileptic EL mice by Johnson, S.C., P. S.Rabinovitch, and M. Kaeberlein.
reducing blood glucose. Epilepsia 42:13711378. 2013. mTOR is a key modulator of ageing and
Guarente, L. 2013. Calorie restriction and sirtuins age-related disease. Nature 493:338345.
revisited. Genes Dev 27:20722085. Johri, A., and M. F. Beal. 2012. Mitochondrial
Hadera, M. G., O. B. Smeland, T. S. McDonald, K. dysfunction in neurodegenerative diseases.
N. Tan, U. Sonnewald, and K. Borges. 2014. J Pharmacol Exp Ther 342:619630.
Triheptanoin partially restores levels of tricar- Juge, N., J. A.Gray, H. Omote, T. Miyaji, T. Inoue, C.
boxylic acid cycle intermediates in the mouse Hara, H. Uneyama, R. H.Edwards, R. A.Nicoll,
pilocarpine model of epilepsy. J Neurochem and Y. Moriyama. 2010. Metabolic control
129:107119. of vesicular glutamate transport and release.
Hardie, D.G., and K. Sakamoto. 2006. AMPK:Akey Neuron 68:99112.
sensor of fuel and energy status in skeletal mus- Kashiwaya, Y., C. Bergman, J. H. Lee, R. Wan, M.
cle. Physiology (Bethesda) 21:4860. T.King, M. R.Mughal, E. Okun, K. Clarke, M.
Hartman, A. L., and C. E. Stafstrom. 2013. P.Mattson, and R. L.Veech. 2013. A ketone ester
Harnessing the power of metabolism for seizure diet exhibits anxiolytic and cognition-sparing
prevention:focus on dietary treatments. Epilepsy properties, and lessens amyloid and tau patholo-
Behav 26:266272. gies in a mouse model of Alzheimers disease.
Hartman, A. L., M. Lyle, M. A. Rogawski, and M. Neurobiol Aging 34:15301539.
Gasior. 2008. Efficacy of the ketogenic diet in the Kashiwaya, Y., T. Takeshima, N. Mori, K.
6-Hz seizure test. Epilepsia 49:334339. Nakashima, K. Clarke, and R. L. Veech. 2000.
Hebert, A. S., K. E. Dittenhafer-Reed, W. Yu, D. Dbeta-hydroxybutyrate protects neurons in
J.Bailey, E. S.Selen, M. D.Boersma, J. J.Carson, models of Alzheimers and Parkinsons disease.
M. Tonelli, A. J. Balloon, A. J. Higbee, M. Proc Natl Acad Sci USA 97:54405444.
S. Westphall, D. J. Pagliarini, T. A. Prolla, F. Keith, H. M. 1933. Factors influencing experimen-
Assadi-Porter, S. Roy, J. M.Denu, and J. J.Coon. tally produced convulsions. Arch Neurol Psychiat
2013. Calorie restriction and SIRT3 trigger 29:148154.
global reprogramming of the mitochondrial pro- Kennedy, A. R., P. Pissios, H. Otu, R. Roberson, B.
tein acetylome. Mol Cell 49:186199. Xue, K. Asakura, N. Furukawa, F. E. Marino,
Henderson, S. T., J. L. Vogel, L. J. Barr, F. Garvin, F. F. Liu, B. B. Kahn, T. A. Libermann, and E.
J. J. Jones, and L. C. Costantini. 2009. Study of Maratos-Flier. 2007. A high-fat, ketogenic diet
the ketogenic agent AC-1202 in mild to mod- induces a unique metabolic state in mice. Am J
erate Alzheimers disease: A randomized, Physiol Endocrinol Metab 292:E1724E1739.
double-blind, placebo-controlled, multicenter Kim do, Y., J. Vallejo, and J. M.Rho. 2010. Ketones
trial. Nutr Metab (Lond)6:31. prevent synaptic dysfunction induced by
Houtkooper, R.H., E. Pirinen, and J. Auwerx. 2012. mitochondrial respiratory complex inhibitors. J
Sirtuins as regulators of metabolism and health- Neurochem 114: 130141.
span. Nat Rev Mol Cell Biol 13:225238. Kim do, Y., L. M.Davis, P. G.Sullivan, M. Maalouf,
Houtkooper, R.H., R. W.Williams, and J. Auwerx. T. A.Simeone, J.van Brederode, and J. M.Rho.
2010. Metabolic networks of longevity. Cell 2007. Ketone bodies are protective against oxida-
142:914. tive stress in neocortical neurons. J Neurochem
Hu, Z. G., H. D. Wang, L. Qiao, W. Yan, Q. 101:13161326.
F. Tan, and H. X. Yin. 2009. The protective Klein, P., J. Janousek, A. Barber, and R.
effect of the ketogenic diet on traumatic brain Weissberger. 2010. Ketogenic diet treatment in
injury-induced cell death in juvenile rats. Brain adults with refractory epilepsy. Epilepsy Behav
Inj 23:459465. 19:575579.
Huttenlocher, P. R. 1976. Ketonemia and sei- Kossoff, E.H., and J. L.Dorward. 2008. The modified
zures: Metabolic and anticonvulsant effects Atkins diet. Epilepsia 49 Suppl 8:3741.
of two ketogenic diets in childhood epilepsy. Kossoff, E. H., and J. M. Rho. 2009. Ketogenic
Pediatr Res 10:536540. diets:Evidence for short- and long-term efficacy.
Huttenlocher, P.R., A. J.Wilbourn, and J. M.Signore. Neurotherapeutics 6:406414.
1971. Medium-chain triglycerides as a therapy Kossoff, E. H., B. A. Zupec-Kania, and J. M. Rho.
for intractable childhood epilepsy. Neurology 2009. Ketogenic diets:An update for child neu-
21:10971103. rologists. J Child Neurol 24:979988.
Ketogenic Diets and Mitochondria and Neurological Diseases 267

Kossoff, E. H., B. A. Zupec-Kania, P. E. Amark, K. Libert, S., and L. Guarente. 2013. Metabolic and neu-
R. Ballaban-Gil, A. G. Christina Bergqvist, R. ropsychiatric effects of calorie restriction and
Blackford, R. Buchhalter, R. H.Caraballo, J. Helen sirtuins. Annu Rev Physiol 75:669684.
Cross, M. G.Dahlin, E. J.Donner, J. Klepper, R. Likhodii, S. S., I. Serbanescu, M. A. Cortez, P.
S.Jehle, H. D.Kim, Y. M.Christiana Liu, J. Nation, Murphy, O. C.Snead, 3rd, and W. M.Burnham.
D. R. Nordli, Jr., H. H. Pfeifer, J. M. Rho, C. 2003. Anticonvulsant properties of acetone, a
E.Stafstrom, E. A.Thiele, Z. Turner, E. C.Wirrell, brain ketone elevated by the ketogenic diet. Ann
J. W.Wheless, P. Veggiotti, E. P.Vining, Charlie Neurol 54:219226.
Foundation, Practice Committee of the Child Lin, M.T., and M. F.Beal. 2006. Mitochondrial dys-
Neurology Society, and. International Ketogenic function and oxidative stress in neurodegenera-
Diet Study Group. 2009. Optimal clinical man- tive diseases. Nature 443:787795.
agement of children receiving the ketogenic Lutas, A., and G. Yellen. 2013. The ketogenic
diet: Recommendations of the International diet: Metabolic influences on brain excitability
Ketogenic Diet Study Group. Epilepsia 50:304317. and epilepsy. Trends Neurosci 36:3240.
Kossoff, E. H., G. L. Krauss, J. R. McGrogan, and Ma, W., J. Berg, and G. Yellen. 2007. Ketogenic
J. M.Freeman. 2003. Efficacy of the Atkins diet diet metabolites reduce firing in central neu-
as therapy for intractable epilepsy. Neurology rons by opening K(ATP) channels. J Neurosci
61:17891791. 27:36183625.
Kossoff, E. H., H. Rowley, S. R. Sinha, and E. Maalouf, M., and J. M.Rho. 2008. Oxidative impair-
P.Vining. 2008. A prospective study of the modi- ment of hippocampal long-term potentiation
fied Atkins diet for intractable epilepsy in adults. involves activation of protein phosphatase 2A
Epilepsia 49:316319. and is prevented by ketone bodies. J Neurosci Res
Kossoff, E.H., J. Huffman, Z. Turner, and J. Gladstein. 86:33223330.
2010. Use of the modified Atkins diet for adoles- Maalouf, M., P. G. Sullivan, L. Davis, D. Y. Kim,
cents with chronic daily headache. Cephalalgia and J. M.Rho. 2007. Ketones inhibit mitochon-
30:10141016. drial production of reactive oxygen species
Kossoff, E. H., J. R. McGrogan, R. M. Bluml, D. production following glutamate excitotoxicity
J.Pillas, J. E.Rubenstein, and E. P.Vining. 2006. by increasing NADH oxidation. Neuroscience
A modified Atkins diet is effective for the treat- 145:256264.
ment of intractable pediatric epilepsy. Epilepsia Mady, M. A., E. H. Kossoff, A. L. McGregor, J.
47:421424. W.Wheless, P. L.Pyzik, and J. M.Freeman. 2003.
Kossoff, E. H., L. C. Laux, R. Blackford, P. The ketogenic diet: Adolescents can do it, too.
F.Morrison, P. L.Pyzik, R. M.Hamdy, Z. Turner, Epilepsia 44:847851.
and D. R.Nordli, Jr. 2008. When do seizures usu- Mantis, J. G., N. A. Centeno, M. T. Todorova,
ally improve with the ketogenic diet? Epilepsia R. McGowan, and T. N. Seyfried. 2004.
49:329333. Management of multifactorial idiopathic epi-
Kossoff, E.H., M. C.Cervenka, B. J.Henry, C. A.Haney, lepsy in EL mice with caloric restriction and the
and Z. Turner. 2013. A decade of the modified ketogenic diet: role of glucose and ketone bodies.
Atkins diet (20032013): Results, insights, and Nutr Metab (Lond)1:11.
future directions. Epilepsy Behav 29:437442. Masino, S. A., and D. N. Ruskin. 2013. Ketogenic
Kovac, S., A. M. Domijan, M. C. Walker, and A. diets and pain. J Child Neurol 28:9931001.
Y. Abramov. 2012. Prolonged seizure activ- Masino, S. A., and J. M. Rho. 2012. Mechanisms
ity impairs mitochondrial bioenergetics and of ketogenic diet action. In: Jaspers Basic
induces cell death. J Cell Sci 125:17961806. mechanisms of the epilepsies. J. L. Noebels, M.
Kovac, S., A. Y. Abramov, and M. C. Walker. 2013. Avoli, M. A. Rogawski, R. W. Olsen, and A.
Energy depletion in seizures: Anaplerosis as a V.Delgado-Escueta, eds. (Bethesda, MD:National
strategy for future therapies. Neuropharmacology Center for Biotechnology Information).
69:96104. Masino, S. A., M. Kawamura, Jr., D. N. Ruskin, J.
Krikorian, R., M. D.Shidler, K. Dangelo, S. C.Couch, D.Geiger, and D. Boison. 2012. Purines and neu-
S. C.Benoit, and D. J.Clegg. 2012. Dietary keto- ronal excitability: Links to the ketogenic diet.
sis enhances memory in mild cognitive impair- Epilepsy Res 100:229238.
ment. Neurobiol Aging 33:425 e419e427. Masino, S. A., T. Li, P. Theofilas, U. S. Sandau,
Lian, X. Y., F. A. Khan, and J. L. Stringer. 2007. D. N. Ruskin, B. B. Fredholm, J. D. Geiger, E.
Fructose-1,6-bisphosphate has anticonvulsant Aronica, and D. Boison. 2011. A ketogenic diet
activity in models of acute seizures in adult rats. suppresses seizures in mice through adenosine
J Neurosci 27:1200712011. A(1) receptors. J Clin Invest 121:26792683.
268 Part III:Homeostatic Manipulators

Mattson, M. P. 2012. Energy intake and exercise Pathak, D., A. Berthet, and K. Nakamura. 2013.
as determinants of brain health and vulner- Energy failure: Does it contribute to neurode-
ability to injury and disease. Cell Metab generation? Ann Neurol 74:506516.
16:706722. Payne, N. E., J. H. Cross, J. W. Sander, and S.
Maurois, P., S. Rocchi, N. Pages, P. Bac, J. P.Stables, P. M.Sisodiya. 2011. The ketogenic and related diets
Gressens, and J. Vamecq. 2008. The PPARgamma in adolescents and adults--a review. Epilepsia
agonist FMOC-L-leucine protects both mature 52:19411948.
and immature brain. Biomed Pharmacother Pfeifer, H. H., and E. A. Thiele. 2005. Low-
62:259263. glycemic-index treatment: A liberalized keto-
Mazzuferi, M., G. Kumar, J. van Eyll, B. Danis, P. genic diet for treatment of intractable epilepsy.
Foerch, and R. M.Kaminski. 2013. Nrf2 defense Neurology 65:18101812.
pathway: Experimental evidence for its protec- Pfeifer, H.H., D. A.Lyczkowski, and E. A.Thiele. 2008.
tive role in epilepsy. Ann Neurol 74:560568. Low glycemic index treatment: Implementation
McDaniel, S. S., N. R. Rensing, L. L. Thio, K. and new insights into efficacy. Epilepsia 49 Suppl
A. Yamada, and M. Wong. 2011. The ketogenic 8:4245.
diet inhibits the mammalian target of rapamycin Pierre, K., and L. Pellerin. 2005. Monocarboxylate
(mTOR) pathway. Epilepsia 52:e7e11. transporters in the central nervous sys-
McKay, J.A., and J. C.Mathers. 2011. Diet induced tem: Distribution, regulation and function. J
epigenetic changes and their implications for Neurochem 94:114.
health. Acta Physiol 202:103118. Porta, N., L. Vallee, C. Lecointe, E. Bouchaert,
Melo, T. M., A. Nehlig, and U. Sonnewald. 2006. B. Staels, R. Bordet, and S. Auvin. 2009.
Neuronal-glial interactions in rats fed a keto- Fenofibrate, a peroxisome proliferator-activated
genic diet. Neurochem Int 48:498507. receptor-alpha agonist, exerts anticonvulsive
Michael-Titus, A. T., and J. V. Priestley. 2013. properties. Epilepsia 50:943948.
Omega-3 fatty acids and traumatic neurological Prins, M. L. 2008. Cerebral metabolic adaptation
injury:From neuroprotection to neuroplasticity? and ketone metabolism after brain injury. J Cereb
Trends Neurosci 37:3038. Blood Flow Metab 28:116.
Milder, J.B., L. P.Liang, and M. Patel. 2010. Acute Prins, M. L., L. S. Fujima, and D. A. Hovda. 2005.
oxidative stress and systemic Nrf2 activation by Age-dependent reduction of cortical contusion
the ketogenic diet. Neurobiol Dis 40:238244. volume by ketones after traumatic brain injury.
Milder, J., and M. Patel. 2012. Modulation of oxida- J Neurosci Res 82:413420.58
tive stress and mitochondrial function by the Prins, M.L., S. M.Lee, L. S.Fujima, and D. A.Hovda.
ketogenic diet. Epilepsy Res 100:295303. 2004. Increased cerebral uptake and oxidation
Muzykewicz, D. A., D. A. Lyczkowski, N. Memon, of exogenous betaHB improves ATP following
K. D. Conant, H. H. Pfeifer, and E. A. Thiele. traumatic brain injury in adult rats. J Neurochem
2009. Efficacy, safety, and tolerability of the low 90:666672.
glycemic index treatment in pediatric epilepsy. Rardin, M.J., J. C.Newman, J. M.Held, M. P.Cusack,
Epilepsia 50:11181126. D. J.Sorensen, B. Li, B. Schilling, S. D.Mooney,
Neal, E.G., H. Chaffe, R. H.Schwartz, M. S.Lawson, C. R.Kahn, E. Verdin, and B. W.Gibson. 2013.
N. Edwards, G. Fitzsimmons, A. Whitney, and J. Label-free quantitative proteomics of the lysine
H.Cross. 2008. The ketogenic diet for the treat- acetylome in mitochondria identifies substrates
ment of childhood epilepsy:Arandomised con- of SIRT3 in metabolic pathways. Proc Natl Acad
trolled trial. Lancet Neurol 7:500506. Sci USA 110:66016606.
Neal, E.G., H. Chaffe, R. H.Schwartz, M. S.Lawson, Rardin, M. J., W. He, Y. Nishida, J. C. Newman,
N. Edwards, G. Fitzsimmons, A. Whitney, and J. C. Carrico, S. R. Danielson, A. Guo, P. Gut, A.
H. Cross. 2009. A randomized trial of classical K. Sahu, B. Li, R. Uppala, M. Fitch, T. Riiff, L.
and medium-chain triglyceride ketogenic diets Zhu, J. Zhou, D. Mulhern, R. D. Stevens, O.
in the treatment of childhood epilepsy. Epilepsia R. Ilkayeva, C. B. Newgard, M. P. Jacobson, M.
50:11091117. Hellerstein, E. S.Goetzman, B. W.Gibson, and E.
Noh, H. S., H. P. Lee, D. W. Kim, S. S. Kang, G. Verdin. 2013. SIRT5 regulates the mitochondrial
J.Cho, J. M.Rho, and W. S.Choi. 2004. A cDNA lysine succinylome and metabolic networks. Cell
microarray analysis of gene expression profiles Metab 18:920933.
in rat hippocampus following a ketogenic diet. Reger, M.A., S. T.Henderson, C. Hale, B. Cholerton, L.
Brain Res Mol Brain Res 129:8087. D.Baker, G. S.Watson, K. Hyde, D. Chapman, and
Palop, J. J., and L. Mucke. 2009. Epilepsy and cog- S. Craft. 2004. Effects of beta-hydroxybutyrate on
nitive impairments in Alzheimer disease. Arch cognition in memory-impaired adults. Neurobiol
Neurol 66:435440. Aging 25:311314.
Ketogenic Diets and Mitochondria and Neurological Diseases 269

Rho, J.M., D. W.Kim, C. A.Robbins, G. D.Anderson, Shimazu, T., M. D.Hirschey, J. Newman, W. He, K.
and P. A. Schwartzkroin. 1999. Age-dependent Shirakawa, N. Le Moan, C. A. Grueter, H. Lim,
differences in flurothyl seizure sensitivity in L. R.Saunders, R. D.Stevens, C. B.Newgard, R.
mice treated with a ketogenic diet. Epilepsy Res V.Farese, Jr., R.de Cabo, S. Ulrich, K. Akassoglou,
37:233240. and E. Verdin. 2013. Suppression of oxidative stress
Rho, J. M., G. D. Anderson, S. D. Donevan, by beta-hydroxybutyrate, an endogenous histone
and H. S. White. 2002. Acetoacetate, ace- deacetylase inhibitor. Science 339:211214.
tone, and dibenzylamine (a contaminant in Shimazu, T., M. D. Hirschey, L. Hua, K.
l-(+)-beta-hydroxybutyrate) exhibit direct anti- E.Dittenhafer-Reed, B. Schwer, D. B.Lombard,
convulsant actions in vivo. Epilepsia 43:358361. Y. Li, J. Bunkenborg, F. W. Alt, J. M. Denu, M.
Roopra, A., R. Dingledine, and J. Hsieh. 2012. P.Jacobson, and E. Verdin. 2010. SIRT3 deacety-
Epigenetics and epilepsy. Epilepsia 53 Suppl lates mitochondrial 3-hydroxy-3-methylglutaryl
9:210. CoA synthase 2 and regulates ketone body pro-
Roth, J., A. L. Szulc, and A. Danoff. 2011. Energy, duction. Cell Metab 12:654661.
evolution, and human diseases:An overview. Am Srivastava, S., U. Baxa, G. Niu, X. Chen, and R.
J Clin Nutr 93:875S883. L.Veech. 2013. A ketogenic diet increases brown
Rothstein, J. D. 2009. Current hypotheses for the adipose tissue mitochondrial proteins and UCP1
underlying biology of amyotrophic lateral scle- levels in mice. IUBMB Life 65:5866.
rosis. Ann Neurol 65 Suppl 1:S3S9. Stafford, P., M. G. Abdelwahab, Y. Kim do, M.
Rowley, S., and M. Patel. 2013. Mitochondrial C.Preul, J. M.Rho, and A. C.Scheck. 2010. The
involvement and oxidative stress in temporal ketogenic diet reverses gene expression patterns
lobe epilepsy. Free Radic Biol Med 62:121131. and reduces reactive oxygen species levels when
Ruskin, D. N., J. Svedova, J. L. Cote, U. Sandau, J. used as an adjuvant therapy for glioma. Nutr
M. Rho, M. Kawamura, Jr., D. Boison, and S. Metab (Lond)7:74.
A. Masino. 2013. Ketogenic diet improves core Stafstrom, C.E., and J. M.Rho. 2012. The ketogenic
symptoms of autism in BTBR mice. PLoS One diet as a treatment paradigm for diverse neuro-
8:e65021. logical disorders. Front Pharmacol3:59.
Ruskin, D. N., M. Kawamura, and S. A. Masino. Stafstrom, C. E., J. C. Ockuly, L. Murphree, M.
2009. Reduced pain and inflammation in juve- T. Valley, A. Roopra, and T. P. Sutula. 2009.
nile and adult rats fed a ketogenic diet. PLoS One Anticonvulsant and antiepileptic actions of
4:e8349. 2-deoxy-D-glucose in epilepsy models. Ann
Sampath, H., and J. M.Ntambi. 2005. Polyunsaturated Neurol 65:435447.
fatty acid regulation of genes of lipid metabo- Sullivan, P.G., C. Dube, K. Dorenbos, O. Steward, and
lism. Annu Rev Nutr 25:317340. T. Z. Baram. 2003. Mitochondrial uncoupling
Schiff, M., P. Benit, A. Coulibaly, S. Loublier, R. protein-2 protects the immature brain from exci-
El-Khoury, and P. Rustin. 2011. Mitochondrial totoxic neuronal death. Ann Neurol 53:711717.
response to controlled nutrition in health and Sullivan, P. G., N. A. Rippy, K. Dorenbos, R.
disease. Ntr Rev 69:6575. C. Concepcion, A. K. Agarwal, and J. M. Rho.
Schlanger, S., M. Shinitzky, and D. Yam. 2002. Diet 2004. The ketogenic diet increases mitochon-
enriched with omega-3 fatty acids alleviates con- drial uncoupling protein levels and activity. Ann
vulsion symptoms in epilepsy patients. Epilepsia Neurol 55:576580.
43:103104. Suzuki, M., K. Sato, S. Dohi, T. Sato, A. Matsuura,
Sekhar, K. R., G. Rachakonda, and M. L. Freeman. and A. Hiraide. 2001. Effect of betahydroxybu-
2010. Cysteine-based regulation of the CUL3 tyrate, a cerebral function improving agent, on
adaptor protein Keap1. Toxicol Appl Pharmacol cerebral hypoxia, anoxia and ischemia in mice
244:2126. and rats. Jpn J Pharmacol 87:143150.
Seyfried, T.N., and L. M.Shelton. 2010. Cancer as a Suzuki, T., H. Motohashi, and M. Yamamoto. 2013.
metabolic disease. Nutr Metab (Lond)7:7. Toward clinical application of the Keap1-Nrf2
Seyfried, T.N., M. A.Kiebish, J. Marsh, L. M.Shelton, pathway. Trends Pharmacol Sci 34:340346.
L. C. Huysentruyt, and P. Mukherjee. 2011. Taha, A.Y., M. A.Ryan, and S. C.Cunnane. 2005.
Metabolic management of brain cancer. Biochim Despite transient ketosis, the classic high-fat
Biophys Acta 1807:577594. ketogenic diet induces marked changes in
Seyfried, T.N., T. M.Sanderson, M. M.El-Abbadi, fatty acid metabolism in rats. Metabolism
R. McGowan, and P. Mukherjee. 2003. Role of 54:11271132.
glucose and ketone bodies in the metabolic con- Taha, A. Y., W. M. Burnham, and S. Auvin. 2010.
trol of experimental brain cancer. Br J Cancer Polyunsaturated fatty acids and epilepsy.
89:13751382. Epilepsia 51:13481358.
270 Part III:Homeostatic Manipulators

Tanner, G.R., A. Lutas, J. R.Martinez-Francois, and Wellen, K.E., G. Hatzivassiliou, U. M.Sachdeva, T.


G. Yellen. 2011. Single K ATP channel opening in V. Bui, J. R. Cross, and C. B. Thompson. 2009.
response to action potential firing in mouse den- ATP-citrate lyase links cellular metabolism to
tate granule neurons. J Neurosci 31:86898696. histone acetylation. Science 324:10761080.
Thio, L. L., M. Wong, and K. A. Yamada. 2000. Wheless, J. W. 2008. History of the ketogenic diet.
Ketone bodies do not directly alter excitatory or Epilepsia 49 Suppl 8:35.
inhibitory hippocampal synaptic transmission. Willis, S., J. Stoll, L. Sweetman, and K. Borges. 2010.
Neurology 54:325331. Anticonvulsant effects of a triheptanoin diet in
Tieu, K., C. Perier, C. Caspersen, P. Teismann, two mouse chronic seizure models. Neurobiol
D. C. Wu, S. D. Yan, A. Naini, M. Vila, V. Dis 40:565572.
Jackson-Lewis, R. Ramasamy, and S. Przedborski. Won, Y.J., V. B.Lu, H. L.Puhl III, and S. R.Ikeda.
2003. D-beta-hydroxybutyrate rescues mito- 2013. Beta-hydroxybutyrate modulates N-type
chondrial respiration and mitigates features of calcium channels in Rat sympathetic neurons by
Parkinson disease. J Clin Invest 112:892901. acting as an agonist for the G-protein-coupled
Todorova, M. T., P. Tandon, R. A. Madore, C. receptor FFA3. J Neurosci 33:1931419325.
E. Stafstrom, and T. N. Seyfried. 2000. The Wong, M. 2013. Mammalian target of rapamycin
ketogenic diet inhibits epileptogenesis in EL (mTOR) pathways in neurological diseases.
mice: A genetic model for idiopathic epilepsy. Biomed J 36:4050.
Epilepsia 41:933940. Yamada, K. A., N. Rensing, and L. L. Thio. 2005.
Urdinguio, R.G., J. V.Sanchez-Mut, and M. Esteller. Ketogenic diet reduces hypoglycemiainduced
2009. Epigenetic mechanisms in neurologi- neuronal death in young rats. Neurosci Lett
cal diseases: Genes, syndromes, and therapies. 385:210214.
Lancet Neurol 8:10561072. Yu, X., X. G. Shao, H. Sun, Y. N. Li, J. Yang, Y.
Van der Auwera, I., S. Wera, F. Van Leuven, and S. C. Deng, and Y. G. Huang. 2008. Activation
T. Henderson. 2005. A ketogenic diet reduces of cerebral peroxisome proliferator-activated
amyloid beta 40 and 42 in a mouse model of receptors gamma exerts neuroprotection
Alzheimers disease. Nutr Metab (Lond)2:28. by inhibiting oxidative stress following
Vanitallie, T. B., C. Nonas, A. Di Rocco, K. Boyar, pilocarpine-induced status epilepticus. Brain
K. Hyams, and S. B.Heymsfield. 2005. Treatment Res 1200:146158.
of Parkinson disease with diet-induced hyper- Yudkoff, M., Y. Daikhin, I. Nissim, O. Horyn, A.
ketonemia: A feasibility study. Neurology Lazarow, B. Luhovyy, and S. Wehrli. 2005.
64:728730. Response of brain amino acid metabolism to
Vining, E. P., J. M. Freeman, K. Ballaban-Gil, C. ketosis. Neurochem Int 47:119128.
S.Camfield, P. R.Camfield, G. L.Holmes, S. Shinnar, Yuen, A.W., J. W.Sander, D. Fluegel, P. N.Patsalos,
R. Shuman, E. Trevathan, and J. W.Wheless. 1998. G. S. Bell, T. Johnson, and M. J. Koepp. 2005.
A multicenter study of the efficacy of the ketogenic Omega-3 fatty acid supplementation in patients
diet. Arch Neurol 55:14331437. with chronic epilepsy: A randomized trial.
Wagner, G.R., and R. M.Payne. 2013. Widespread Epilepsy Behav 7:253258.
and enzyme-independent Nepsilonacetylation Zhao, Z., D. J.Lange, A. Voustianiouk, D. MacGrogan,
and Nepsilon-succinylation of proteins in the L. Ho, J. Suh, N. Humala, M. Thiyagarajan, J.
chemical conditions of the mitochondrial Wang, and G. M.Pasinetti. 2006. A ketogenic diet
matrix. J Biol Chem 288:2903629045. as a potential novel therapeutic intervention in
Waldbaum, S., and M. Patel. 2010. Mitochondrial amyotrophic lateral sclerosis. BMC Neurosci7:29.
dysfunction and oxidative stress:Acontributing Ziegler, D.R., L. C.Ribeiro, M. Hagenn, I. R.Siqueira,
link to acquired epilepsy? J Bioenerg Biomembr E. Araujo, I. L.Torres, C. Gottfried, C. A.Netto,
42:449455. and C. A. Goncalves. 2003. Ketogenic diet
Warburg, O. 1956. On the origin of cancer cells. increases glutathione peroxidase activity in rat
Science 123:309314. hippocampus. Neurochem Res 28:17931797.
16
Dietary Manipulations
AT H A N A S I O S E VA N G E L I O U A N D M A R T H A S P I L I O T I

INTRODUCTION of many mitochondrial enzymes involved in the


The brain is the most active organ in the body citric acid cycle, gluconeogenesis, the urea cycle,
in terms of energy consumption and therefore and fatty acid oxidation (Di Lisa et al., 1997;
particularly vulnerable to disruptions of energy Evangeliou,1992).
resources. Energy is available in the form of Carnitine is a nonessential nutrient, first
adenosine triphosphate(ATP), the major source described in the early beginnings of the 20th cen-
of biological free energy in higher organisms. tury. It was initially considered to be an essen-
Consequently, impaired ATP production threat- tial vitamin; it was later discovered that it could
ens brain homeostasis and integrity. Many be produced in the liver, kidney, and brain from
normal and pathological situations are associ- amino acid precursors (lysine and methionine).
ated with either increased energy demands or Human skeletal muscle, heart, liver, kidney, and
impaired brain energy production. Some of these brain are able to synthesize -butyrobetaine,
situations are depicted in Table 16.1. In all catego- an immediate precursor of carnitine, but only
ries in Table 16.1, one question that has occupied the liver, kidney, and brain have the ability to
scientists for many years is how to restore energy convert this substrate to carnitine (Vaz and
balance when and where deficits exists and how Wanders, 2002). The reaction is catalyzed by
to improve existing normal energy balance in the -butyrobetaine hydroxylase. Due to the very
brain. Accordingly, dietary manipulations that low activity of -butyrobetaine hydroxylase in
improve metabolic function or prop up ATP pro- fetal and neonatal age, carnitine is considered
duction may have a vast number of implications a vitamin for newborn babies (Ling etal., 2012;
in health and disease. In the past years there has Rebouche and Seim,1998).
been accumulating evidence that various nutri- In humans, 75% of carnitine intake comes
ents that can be ingested in food or by themselves from dietary sources (Li etal., 1992; Pekala etal.,
may prove to be effective in improving brain 2011). These include most animal foods; however,
energy balance. In this chapter we provide an due to losses in food cooking and preparation,
overview of the nutrients that can impact and there are few data on the total content ingested
improve brain energy homeostasis. (Rospond and Chopicka, 2013). Carnitine is not
metabolized and is excreted, mostly as free carni-
L-CARNITINE AND tine, in theurine.
CARNITINEESTERS Homeostatic mechanisms conserve the
Carnitine (-hydroxy--trimethylaminobutyric amount of L-carnitine in the body and under
acid) is an essential cofactor for the transport of normal circumstances regulate blood and tis-
long-chain fatty acids across the inner mitochon- sue levels within relatively narrow limits. The
drial membrane into the mitochondrial matrix, uptake of L-carnitine into most tissues of the
where they are broken down via -oxidation body, including muscles and neurons, involves
(Evangeliou and Vlassopoulos, 2003; Foster, carrier-mediated transport systems, which
2004; Pekala etal., 2011; Virmani and Binienda, maintain high tissue-to-plasma concentration
2004; Figure 16.1). In addition to its principal ratios. Especially in the brain, carnitine is trans-
function, carnitine also buffers potentially toxic ported from the plasma into neurons by Na+- and
acyl-CoA metabolites and modulates the ratio of energy-dependent processes and may involve the
acyl-CoA/CoA (Di Lisa etal., 1997; Stumpf etal., cation transporter OCTN2 (Berezowski et al.,
1985; Figure 16.2). The later regulates the activity 2004; Naecz etal.,2004).
272 Part III:Homeostatic Manipulators

TABLE16.1 SIT UAT IONS W IT H I M PA IR ED OR I NCR E ASED


EN ERGY DEM A N DS
Normal Situations Pathological Situations

Impaired Energy Increased Energy Impaired/Increased Energy


Demands Demands Demands
Aging Pregnancy Inborn errors of metabolism
Infancy Autism
Childhood Parkinsons disease
Puberty Huntingtons disease
Other stress situations Alzheimers disease
Amyotrophic lateral sclerosis
Depression
Schizophrenia
Bipolar disorder
Cancer
Epilepsy

In addition to effects on lipid metabolism, pyruvate dehydrogenase with subsequent reduc-


carnitine influences carbohydrate and protein tion of carbohydrate breakdown (Holness and
metabolism. This is achieved different ways. As Sugden, 2003). L-Carnitine supplementation
already mentioned, carnitine affects the ratio of reduces the acyl-CoA/CoA ratio, thus increas-
acyl-CoA/CoA. This relation is crucial in mito- ing the amount of nonesterified CoA essential
chondrial metabolism, modulating the activity of for lipid and carbohydrate utilization (Chapela
many mitochondrial enzymes, including, among etal., 2009; Ramsay, 2000; Ramsay and Arduini,
others, pyruvate dehydrogenase, a very impor- 1993). Moreover L-carnitine is an energy car-
tant enzyme in glucose metabolism (Evangeliou rier through the mitochondrial membrane and
and Vlassopoulos, 2003; Ramsay, 2000). Ahigh so modulates the availability of acetyl-CoA
ratio of acyl-CoA to CoA reduces the activity of and consequently controls the activity of

CYTOSOL
Carnitine
Acyl-CoA Acylcarnitine

CPT I Outer Mitochondrial Membrane

Acylcarnitine CoA
Carnitine Acylcarnitine

Carnitine

CT CT Inner Mitochondrial Membrane


CPT II CAT
Acylcarnitine
CoA
Carnitine Carnitine
Acyl-CoA Carnitine
Acylcarnitine
CoA
MITOCHONDRIAL MATRIX
-oxidation Acetyl-CoA

FIGURE16.1: Transport of long-chain fatty acids across the inner mitochondrial membrane into the mitochon-
drial matrix. CAT=carnitine acetyl translocase; CPT I=carnitine palmitoyl transferase I; CPT II=carnitine
palmitoyl transferase II; CAT=carnitine acetyl translocase.
Dietary Manipulations 273

Carnitine Acylcarnitine Urine


Vivo, 1995). These symptoms often are accom-
panied by central nervous system (CNS) signs
such as hepatic encephalopathy in which the
major pathologic finding is swelling of astro-
cyte cytoplasm, while less often expanded
mitochondria and myelin sheath splitting in
AcylCoA CoA the white matter are observed (Kimura and
Amemiya, 1990; Malaguarnera, 2013; Virmani
and Binienda, 2004). Similar symptoms to
primary carnitine deficiency are observed in
Toxic organic acids
secondary carnitine deficiencies occurring
Valproic acid
in some inborn errors of metabolism and in
FIGURE 16.2: Carnitine buffering of toxic acyl-CoAs some acquired conditions such as in patients
by conversion to acylcarnitine and modulation of the under valproate therapy (Morand et al., 2012;
ratio of acyl-CoA/CoA. Valproic acid, used broadly Okumura et al., 2011; Stumpf et al., 1985;
as an antiepileptic, causes an iatrogenic organic aci- Vreken etal.,1999).
demia and is also buffering through carnitine. This Beyond the changes in carnitine metabo-
reaction is a two-way chemical reaction modulating lism observed in inborn errors of metabolism
the acyl-CoA/CoA ratio. The acyl-CoA/CoA ratio is and the role of carnitine in the prevention of
very crucial in mitochondrial metabolism by control- clinical symptoms related to these disorders,
ling enzymes of the Krebs cycle: pyruvate dehydoge- carnitine seems to play an important role as
nase complex, pyruvate carboxylase, citrate synthase, adjunctive treatment in some neurodegen-
acetyl-CoA carboxylase, adenine nucleotide translo- erative disorders and also in the prevention of
case, glutamate dehydrogenase, malate dehydrogenase, consequences associated with aging (Calabrese
and succinyl-CoA ligase. etal.,2010).

Carnitine andAging
the respiratory chain complex (Virmani and Clinical and experimental data in recent
Binienda, 2004). Indeed, the majority of carni- decades has shown the relation of carnitine
tine deficiency syndromes are associated with metabolism to aging and the beneficial effect of
mitochondrial dysfunction and vice versa (Geier carnitine on the amelioration of aging conse-
and Geier, 2013; Kaido et al., 1997; Weinberg quences. Animal experiments have shown that
and Baughman, 2006). In addition, experimen- the aging process is accompanied by carnitine
tal and clinical data provide evidence for ben- deficiency in brain, myocardium, and skeletal
eficial effects of L-carnitine in situations with muscle (Costell et al., 1989; Flanagan et al.,
mitochondrial dysfunction (Binienda, 2003; 2010; Maebashi et al., 1982; Rebouche, 1992).
Binienda etal., 2005; La Guardia etal., 2013). In Carnitine deficiency in these tissues is accompa-
particular, carnitine increases function of the nied by disturbances in mitochondrial genesis,
respiratory chain and antioxidation and lipid morphology, and function. These mitochon-
peroxidation tolerance capacity in skeletal mus- drial alterations are increasingly recognized as
cle mitochondria. L-carnitine supplementation components of the aging process, and experi-
also has protein sparing effects; the activation of mental work provides evidence that L-carnitine
the -oxidation pathway, mediated through car- raises mitochondrial energy-producing capa-
nitine, reduces the breakdown of branched-chain bilities by reversing the age-associated decline
amino acids (BCAAs) via an internal feedback in mitochondrial enzyme activities and thereby
mechanism (inhibition of the branched chain protecting mitochondria from aging (Arockia
keto-dehydrogenase) and promotes protein Rani and Panneerselvam, 2001; Noland et al.,
synthesis resulting in a leaner body (Feller and 2009; Savitha etal., 2005). Consistent with this,
Rudman, 1988; Owen etal.,1994). clinical work done in centenarians indicates
Several primary carnitine deficiency syn- that oral administration of carnitine produces
dromes have been described, and the major- a reduction of total fat mass, increases total
ity of them are characterized by hepatopathy, muscular mass, and facilitates an increased
cardiomyopathy, myopathy, myoglobinuria, capacity for physical and cognitive activity by
and nonketotic hypoglycemia (Magoulas and reducing fatigue and improving cognitive func-
El-Hattab, 2012; Pons etal., 1997; Pons and De tions (Malaguarnera etal.,2007).
274 Part III:Homeostatic Manipulators

Carnitine andAutism Acetyl-L-Carnitine


Autism spectrum disorder (ASD) is highly Acetyl-L-carnitine is an endogenous molecule
genetic and multifactorial, with many interact- synthesized in mitochondria by the enzyme
ing risk factors. Recent studies have implicated acetyl-L-carnitine transferase and is the predom-
abnormalities in mitochondrial metabolism inant acylcarnitine in normal tissues (Hudson
(Guevara-Campos et al., 2013; Legido et al., and Tabet, 2003; Pettegrew etal., 2000; Rebouche,
2013; Rossignol and Frye, 2012). Interestingly, 2004). Acetyl-L-carnitine facilitates the influx
most individuals with ASD and mitochondrial and efflux of acetyl groups across the mitochon-
dysfunction do not have a specific genetic muta- drial inner membrane (Reuter and Evans, 2012;
tion, raising the possibility that mitochondrial Virmani and Binienda, 2004). Some of acetyl-L-
dysfunction may be acquired, at least in a sub- carnitines proposed neuroprotective benefits
group of children with ASD (Dhillon etal., 2011; involve improved mitochondrial energetics and
Hadjixenofontos etal., 2013). Consequently, one function, antioxidant activity, stabilization of
suggested approach to treating patients diag- membranes, protein and gene expression modu-
nosed with an ASD entails the administration lation, and enhancement of cholinergic neuro-
of mitochondrial respiratory chain cofactors to transmission (Ando etal., 2001; Assaf etal., 2012;
enhance mitochondrial function. Such a strat- Haorah et al., 2011; Kathirvel et al., 2013; Patel
egy would employ stimulation of enzyme activ- etal., 2010; Pillich etal., 2005; Traina etal., 2008;
ity by supplying precursors or co-enzyme and Virmani etal.,2001).
alternative substrates (Frye etal., 2013a, 2013b). Multiple clinical studies have shown that
L-carnitine is among the suggested options for acetyl-L-carnitine may be of benefit in treating
treatment, and a recent prospective, double-blind, aging-related consequences, neurodegenerative
placebo-controlled trial demonstrated that disorders, peripheral neuropathies, and cognitive
L-carnitine therapy for 3months among subjects impairment related to various conditions (Beghi
diagnosed with an ASD significantly improved et al., 2013; Bianchetti et al., 2003; De Grandis,
on clinical measurements recorded by trained 2007; Hudson and Tabet, 2003; Malaguarnera
professionals and parents of study subjects (Geier et al., 2008; Montgomery et al., 2003; Zanelli
etal.,2011). etal.,2005).

Carnitine and Valproate C R E AT I N E


Valproate treatment is known to reduce blood Creatine (Cr; -methyl-guanidinoacetic acid)
and muscle carnitine levels (Anil et al., 2009; was discovered and isolated from meat extract
Morand et al., 2012). Reduced carnitine lev- in 1835 by Chevreul. Although studies regard-
els in patients under valproate therapy may ing its physiology have been published, until
be associated with severe hepatotoxicity and recently research for its clinical application was
hyperammonemia (Hamed and Abdella, 2009). lacking. Since Roger Harris and his colleagues
In addition, in some rare cases treatment with have demonstrated that Cr loading is able to
valproate is associated with the development of enhance muscle Cr and phospho-Cr (PCr) con-
coma and life-threatening cerebral edema, even tent, Cr supplementation has been largely used
in the absence of concomitant hepatic failure to improve exercise capacity in healthy individu-
(Triggs etal.,1997). als and athletes (Francaux and Poortmans, 1999;
Numerous studies have demonstrated the Harris etal., 1992; Hoffman etal.,2006).
beneficial effects of carnitine supplementation Work by Stockler et al. (1994, 1996) and
in the treatment of valproate-induced hepa- Salomons et al. (2001) identified the first disor-
totoxicity (Bhmer et al., 2010; Perrott et al., ders of Cr metabolism and their clinical presenta-
2010; Sonik et al., 2011). In November 1996, a tions that involve CNS and respond positively to
panel of pediatric neurologists met to update Cr therapy. Accordingly, researchers are focused
the consensus statement issued in 1989 by a on the possible role of Cr supplementation on
panel of neurologists and metabolic experts brain metabolism. It is well known that Cr treat-
on L-carnitine supplementation in childhood ment provides neuroprotection against various
epilepsy. The panelists agreed that intravenous toxic insults in fetal rat ventral mesencephalic
L-carnitine supplementation is clearly indicated and striatal cultures (Andres etal., 2005). There
for valproate-induced hepatotoxicity and over- is some evidence that Cr treatment can be
dose (De Vivo etal.,1998). applied in neurodegenerative disorders such as
Dietary Manipulations 275

Huntingtons and Parkinsons diseases (Bender the potential benefits of oral Cr supplementa-
etal., 2008; Hass etal., 2007; Hersch etal., 2006; tion to treat brain-related disorders (Allen, 2012;
Tabrizi et al., 2005). In the past few years there Nash etal., 1994; Ohtsuki etal., 2002). Questions
has been growing evidence about Crs neuropro- arise about the rate of Cr crossing the BBB.
tective effect on acquired brain disorders such as Recent research shows that the CRT SLC6A8 is
brain trauma, perinatal brain damage, and epi- expressed by microcapillary endothelial cells at
lepsy, as well as a variety of pathologies in which the BBB, but it is absent from their surrounding
energy metabolism and oxidative stress play an astrocytes. This raised the concept that the BBB
etiological role (Adcock et al., 2002; Evangeliou has a limited permeability for peripheral Cr and
et al., 2009b; Iqbal et al., 2013; Sakellaris that the brain supplies a part of its Cr by endog-
etal.,2006). enous synthesis (Braissant, 2012). Recent data
Cr is distributed throughout the body with on Cr metabolism shows that in physiological
95% found in skeletal muscle. The remaining conditions, Cr is taken up by the CNS from the
5% is located in the brain, liver, kidney, and periphery through SLC6A8 at BBB but in limited
testes (Walker, 1979). Cr content in the body is amounts, and that the CNS also needs its own Cr
maintained by nutritional intake and endog- synthesis (Braissant,2012).
enous biosynthesis roughly equally. The dietary Cr is taken up by neurons and oligoden-
source is restricted to animal sources, such as drocytes through CRTs where, together with
meat. In vegetarians it is likely that the synthe- endogenous Cr, it participates in the regulation
sis route is upregulated. Chronic supplemen- of energy balance. The effect of Cr in energy
tation of Cr results in downregulation of the metabolism is depicted in Figure 16.3. As noted,
Cr transporter (CRT; Guerrero-Ontiveros and recent research on Cr has demonstrated positive
Wallimann,1998). therapeutic results in various acquired clinical
The first step of Cr biosynthesis probably occurs disorders, especially for those with disturbed
mainly in the kidney with L-arginine:glycine energy balance.
amidinotransferase (AGAT), an enzyme that
catalyzes the conversion of arginine and glycine Creatine and BrainTrauma
to form guanidinoacetate and ornithine, whereas The most common cause of death and disability
the liver is likely to be the principal organ accom- after severe trauma in childhood is traumatic
plishing the subsequent methylation of guanidi- brain injury (TBI). Of all trauma deaths, 25% are
noacetate to Cr (Bera etal., 2008; Lage etal., 2013; caused by head injury (Tepas et al., 1990). TBI
Walker, 1979). Cr phospokinase (CPK) catalyses leads to functional deficits caused in primary
the phosphorylation and dephosphorylation of Cr and secondary mechanisms. While for primary
and PCr, respectively (Hespel etal., 2001). Cr and mechanisms the damage occurs at the time of
PCr play an essential role in the storage and trans- impact and results from the mechanical insult
mission of phosphate-bound energy (Balestrino itself, for secondary mechanisms the cellular
etal.,2002). damage occurs not immediately after the trauma
Endogenous synthesis of Cr occurs in the but develops within minutes, hours, or even days,
brain as AGAT and S-adenosyl-L-methionine:g and during this time medical intervention, phar-
uanidinoacetate N-methyltransferase (GAMT) maceutical or mechanical, is usually applied.
are expressed in most cell types, particularly One of the first events caused by the secondary
neurons, oligodendrocytes, and astrocytes, it mechanisms is mitochondrial dysfunction asso-
is generally agreed that the majority of Cr syn- ciated with the disruption in cellular calcium
thesis occurs peripherally in the kidney (via homeostasis. Maintenance of cellular calcium
AGAT), liver, and pancreas (via GAMT; Bard homeostasis is intimately related to ATP use and
and Braissant, 2010; Braissant et al., 2011; synthesis, which are key to proper brain function-
Brosnan etal., 2011; Brosnan etal., 2007; Edison ing. At this point enhanced neuron survival may
et al., 2007). Following biosynthesis or inges- be improved by providing an adequate supply of
tion, Cr is transferred from blood plasma by ATP immediately after trauma, and at this point
specific sodiumand chloride-dependent CRTs Cr could play an important role; recent accu-
located in skeletal muscle, kidney, heart, brain, mulative literature support this idea (Sullivan
and liver (Allen, 2012; Snow and Murphy, 2001). etal., 2000). Specifically, it is well known that Cr
Importantly, Cr enters the brain via these CRTs at supplementation increases intramuscular and
the blood-brain barrier (BBB), which emphasizes cerebral stores with both Cr and PCr (Dechent
276 Part III:Homeostatic Manipulators

Glycolysis b-oxidation ATP dependent processes


at cell membrane, place1
in cytosol, at cytoskeleton
BRAIN at organelles
Acetyl-CoA
ATP
Energy deficit
Endogenous Krebs situations
creatine synthesis cycle ATP
CK Neurodegenerative
CK disorders
NADH place2
PCr pool
Mitochondrial
respiratory chain

Creatine + ATP CK ADP + PCr

creatinine

CRT
SLC6A8 BBB BBB

Dietary creatine
intake
Exogenously
administered
creatine

FIGURE 16.3: Simplified schema of creatine (Cr) metabolism and its relationship to ATP. Cr taken up into the
brain via Cr transporters (CRT SLC6A8) is transformed to the high-energy compound PCr by Cr kinase (CK).
ATP synthesis mainly from carbohydrate (glucose) and less from fat occurs via three series of metabolic path-
ways:glycolysis and -oxidation, the Krebs cycle, and the mitochondrial respiratory chain. When ATP is rapidly
depleted, CK catalyzes the donation of a phosphate group from phosphocreatine (PCr) to ADP, producing more
ATP to buffer energy needs. Conversely, when energy is released, an individual phosphate group is cleaved from
ATP and bound to Cr to rejoin the PCr pool. PCr is used to buffer cytosolic ATP/ADP ratios and for local ATP
consumption, e.g., by cytosolic metabolic enzymes, ATP-requiring contractile processes or cell motility, organelle
transport or ATP-dependent cell signaling or at the cell membrane by ATP-requiring ion pumps, ATP-gated
ion-channels or ATP-regulated receptors (place 1). In addition, PCr plays an important role in managing energy
deficits in neurodegenerative diseases such as Parkinsons and Alzheimers (place2).

etal., 1999). The increase of these stores may offer 6 months at a dose of 0.4 g/kg/day orally)
therapeutic benefits by stimulating protein syn- improved results in several parameters, including
thesis or reducing protein degradation, stabiliz- duration of posttraumatic amnesia, duration of
ing biological membranes, and preventing ATP intubation and intensive care unit stay, disability,
depletion, which occur in patients with TBI. The good recovery, self-care, communication, loco-
experimental work by Lifshitz et al. (2003) and motion, sociability, personality/behavior, and
Hausmann et al. (2002) demonstrated that Cr neurophysical and cognitive function (Sakellaris
reduced cortical damage and protected mito- etal., 2006; Sakellaris etal.,2008).
chondrial energetics in the brain while reducing
the spread of secondary spinal cord injury. Creatine and Epilepsies
Although there is plethora of experimental It is well known that Cr is involved in the patho-
work dealing with the neuroprotective role of Cr physiology of epilepsy and that epileptic seizures
in TBI, few clinical studies have been published are accompanied by increased CPK concentra-
on the same subject. In clinical work done by our tion levels. Thus in patients with a recent loss
team in 39 children and adolescents 1 to18years of consciousness and abnormal movements,
old with TBI, Cr supplementation (given for serum CPK concentration is a useful, practical,
Dietary Manipulations 277

and relatively accurate parameter to assist in the experimental work and in light of the afore-
differentiation of epileptic seizures from either mentioned findings, the investigation of energy
vasovagal syncope or psychogenic nonepileptic metabolites within the hippocampal tissue dem-
seizures (Neufeld et al., 1997; Petramfar et al., onstrated that the ketogenic diet induced an
2009). Early experimental work done from increase in the PCr/Cr energy-store ratio and tis-
Glatzner et al. (1979) suggested that sources sue levels of -hydroxybutyrate, although ATP,
other than skeletal muscle alone contribute to ADP, and AMP levels did not change appreciably.
the increased CPK activity after grand mal sei- Based on these observations, we gave a Cr
zures. A later magnetic resonance spectroscopy supplement to a group of patients for whom
study in humans suggested that an abnormal the ketogenic diet was partially successful
N-acetylaspartate/Cr (or choline) ratio will poten- (Evangeliou A, Asprangathou D, Karanika E,
tially provide a useful indicator of poor seizure Spilioti M., unpublished data). Our rationale
control condition in patients with temporal lobe was to precipitate a further rise in the PCr/Cr
epilepsy (Mendes-Ribeiro et al., 1998). Several energy-store ratio in order to induce an increase
relevant publications suggest that an increase in in energy production in the brain, thus enhanc-
Cr may reflect reactive astrocytosis (mesial tem- ing the efficacy of the diet. Preliminary data gives
poral sclerosis), even when MRI volumetric stud- us some hope that the ketogenic diet in combi-
ies show normal hipoccampi (Cendes etal., 1997; nation with Cr might be more effective in the
Kuzniecki and Jackson, 1995). Crs antiepileptic treatment of refractory epilepsies. Specifically, in
action on immature brains was first introduced 10 children with therapy-resistant epilepsy
by Holzman et al. (1998). Specifically, rat pups receiving the ketogenic diet, Cr supplementation
were injected with Cr for 3days before exposing was added in doses up to 5 g daily. The ketogenic
them to hypoxia on postnatal day 10 or 20. Before diet had a partial effect in all of these children,
and during hypoxia, the electrocortical activ- and when Cr was added, 3 of the children experi-
ity or 31P nuclear magnetic resonance spectra enced a further seizure reduction of 90% to 100%
were measured. At 10 but not 20days, Cr injec- from baseline.
tions increased brain Cr/nucleotide triphosphate
ratios, decreased hypoxia-induced seizures and M E L AT O N I N
deaths, and enhanced brain PCr and ATP recov- Melatonin (N-acetyl-5-methoxytryptamine) is
eries after hypoxia. Thus Cr protected the meta- a signaling molecule released as a hormone of
bolically immature brain from hypoxia-induced the pineal gland predominantly during night.
seizures and, perhaps, from cellular injury. It regulates the sleepwake cycle by chemically
In another study (Royes et al., 2006), Cr causing drowsiness and lowering body tempera-
supplementation protected not only against ture (Al-Omary, 2013; Hardeland, 2012; Rios
methyl-malonic acid-induced convulsions but et al., 2010). Therefore, interventions that target
also against methyl-malonic acidinduced oxi- sleep may improve child health and child and
dative damage. Furthermore, experimental work family distress but may also ameliorate core and
from the same group demonstrated that physi- related symptoms of autism (Gunol etal., 2011;
cal training, Cr supplementation, or their com- Rossignol and Frye, 2011). Most of its biological
bination attenuated pentylenetetrazol-induced effects are mediated by G-protein-coupled recep-
seizures and oxidative damage in vivo, provid- tors MT1 and MT2, with its main action within
ing evidence that combined Cr supplementation the CNS. In mammals, these receptors are
and physical exercise may be a useful strategy in expressed in various tissues and organs, includ-
the treatment of convulsive disorders (Rambo ing the brain (Dubocovich et al., 2010; Levoye
etal.,2009). etal.,2006).
Another interesting finding is the fact that Melatonin secretion decreases during aging.
the ketogenic diet, a metabolic therapy suc- Decreased levels of melatonin, with concomi-
cessful in the treatment of refractory epilep- tant upregulation of MT1 and MT2 receptors
sies, also impacts Cr metabolism. Experimental exceeding that observed during normal aging,
work done by Bough etal. (2006) demonstrated have been repeatedly described in neurodegen-
that the ketogenic diet induced a striking 46% erative disorders, especially Alzheimers disease
increase in the density of mitochondrial profiles and other types of senile dementia, in some
in tissue taken from ketogenic dietfed animals mood disorders, and after prolonged drug treat-
compared to controls. Furthermore, in the same ment (e.g., antidepressants; Boyce and Hopwood,
278 Part III:Homeostatic Manipulators

2013; Bubenik and Konturek, 2011; Carvalho not appear to dissipate with continued mela-
etal., 2009; Mishima etal., 1999; Rosales-Corral tonin use. Although the absolute benefit of
etal.,2012). melatonin compared to placebo is smaller than
Melatonin also acts as a direct free-radical other pharmacological treatments for insomnia,
scavenger and indirect antioxidant by stimulat- melatonin has a role in the treatment of insom-
ing gene expression for antioxidant enzymes nia given its relatively benign side-effect profile
(superoxide dismutase, catalase, glutathione compared to these other agents (Ellis etal., 2012;
peroxidase, glutathione reductase) or to increase Ferracioli-Oda etal.,2013).
their activity (Fischer et al., 2008; Reiter et al.,
2010). Unlike other antioxidants, melatonin can Melatonin andAutism
easily cross all morphophysiological barriers, Sleep difficulties, particularly insomnia, occur in
such as the BBB, and enter cells and subcellu- more than 50% of children with autism spectrum
lar compartments. Based on all these properties disorders (Bain, 2006; Couturier etal., 2005). In
there is accumulating evidence suggesting the addition, there is increasing evidence that sleep
potential of melatonin in neurodegenerative con- disorders in children with autism are associated
ditions such as Parkinsons disease, Alzheimers with disrupted melatonin secretion (Doyen etal.,
disease, Huntingtons disease, amyotrophic 2011; Goldman etal., 2012). Accordingly, numer-
lateral sclerosis, epileptic seizures, and stroke ous studies have suggested the use of melatonin
(Cecon and Markus, 2011; Pandi-Perumal etal., for sleep problems of children and adults with
2013; Polimeni etal.,2014). autism (Cortesi et al., 2012; Malow et al., 2012;
Tordjman etal.,2005).
Melatonin and Insomnia
Insomnia is a common sleep disorder that can Melatonin and Neurodegenerative
make it hard to fall asleep, hard to stay asleep, or Disorders
both, despite the opportunity for adequate sleep. Numerous studies suggest melatonins neuro-
Endogenous melatonin levels decrease with age, protective role in neurodegenerative diseases
probably due to an age-related decline in circa- (Galli-Carminati etal., 2009; Huang etal., 2009;
dian rhythmic functions or the calcification of Srinivasan etal., 2011). Intensive research in the
the pineal gland (Dijk et al., 1999; Ortiz et al., past few years has indicated melatonins benefi-
2008). Since endogenous melatonin has benefi- cial effects in neurodegenerative disorders. Brain
cial effects on sleep in humans and helps stabi- oxidative damage has been implicated as a com-
lize physiological circadian rhythms, including mon link in the pathogenesis of such diseases.
the sleepwake cycle, declining melatonin levels Melatonin crosses the BBB easily and acts as a
may contribute to the common complaint of poor free-radical scavenger, and its broad spectrum of
sleep quality seen among the elderly (Touitou antioxidant activities in many CNS neurodegen-
etal., 1984; Wade etal., 2011; Wade etal., 2010). erative diseases has been well documented and
This has raised the possibility of improving sleep reviewed (Galli-Carminati etal., 2009; Gehrman
in elderly patients with insomnia by appropri- et al., 2009; Sliwinski et al., 2007; Wang, 2009;
ately timed treatment with melatonin (Al-Aama Weishaupt etal.,2006).
et al., 2011; Lyseng-Williamson, 2012; Simn
Padilla et al., 2009). The prevalence of chronic BR ANCHED CHAIN
sleep onset insomnia is common not only in AMINOACIDS
elderly people but in all ages (Blader etal., 1997;
Cunnington etal., 2013; Luthringer etal.,2009). Metabolism
A recent meta-analysis involving 1,683 sub- The BCAAs (leucine, isoleucine, and valine) con-
jects of all ages demonstrated that melatonin stitute an important subgroup of the indispensa-
significantly improved sleep in subjects with ble amino acids. BCAAs are the most hydrophobic
primary sleep disorders compared to placebo. of the amino acids and play crucial roles in deter-
In this study, melatonin reduced sleep-onset mining the structures of globular proteins as well
latency, increased total sleep time, and improved as the interaction of the transmembrane domains
overall sleep quality compared to placebo to a sta- of membranous proteins with phospholipid bilay-
tistically significant degree (Monterrosa-Castro ers (Brosnan and Brosnan, 2006). Despite their
et al., 2013). According to the same study, the similarity, the three BCAAs play subtly different
effects of melatonin on sleep are modest but do roles in proteins. Their side chains differ in size,
Dietary Manipulations 279

shape, and hydrophobicity. Therefore, they have branched chain -ketoacid-dehydrogenase and
different predilections for different secondary can oxidize the branched-chain ketoacids. As
structure motifs. Most proteins have a relatively shown in Figure 16.4 all three BCAAs are con-
high proportion of these amino acids. Indeed, verted to branched-chain ketoacids while their
most dietary proteins consist of ~20% BCAAs. further metabolism employs distinct pathways
They comprise some 35% of the indispensable to end products of glucose and/or ketone bod-
amino acid requirements of mammals (Harper ies. Ketone bodies and glucose can then be oxi-
etal.,1984). dized as a respiratory fuel by extrahepatic tissues,
The three BCAAs support numerous meta- helping the organism in energy-stress situations
bolic processes ranging from the fundamental such as diabetes, epilepsy, neurodegenerative dis-
role as substrates for protein synthesis to meta- ease, and psychiatric disorders (Cole etal., 2010;
bolic roles as energy substrates, precursors for Doisaki et al., 2010; Iwasa et al., 2010; Piscopo
synthesis of alanine and glutamine and the neuro- etal., 2011; Richardson etal., 2004; Scarna etal.,
transmitter glutamate, and modulators of muscle 2003; Sevy etal.,2006).
protein synthesis via the insulin-signaling path-
way (Ahlborg et al., 1974; Anthony et al., 2001; BCAA and Their Role intheCNS
Hutson etal., 2001; Li etal., 2003; Patti etal., 1998; Beyond direct metabolic effects, the BCAAs par-
Ruderman, 1975; Xu etal., 1998). Degradation of ticipate directly and indirectly in a variety of
BCAAs in skeletal muscle is linked to production important biochemical functions in the brain.
of alanine and glutamine and maintenance of BCAAs are crucial for homeostasis of gluta-
glucose homeostasis (Layman, 2003). The catabo- mate, an essential excitatory neurotransmitter.
lism of the BCAAs is shown in Figure16.4. Specifically, BCAAs (especially leucine) provide
Not all tissues can oxidize BCAAs. Whereas neurons with a constant supply of glutamate,
muscle has BCAA aminotransferase activity, the which both neurons and glia oxidize robustly
liver lacks this enzyme. However, the liver has (Bixel et al., 2001; Sweatt et al., 2004). he

Isoleucine Leucine Valine


-KG Branched chain -KG Branched chain -KG Branched chain
aminoacid aminoacid aminoacid
aminotransferase aminotransferase aminotransferase
Glutamate Glutamate Glutamate

-keto--methylvalerate -ketoisovalerate -ketoisocaproate


glucose
CoA Branched chain CoA Branched chain CoA Branched chain
ketoacid- ketoacid- ketoacid-
pyruvate dehydrogenase dehydrogenase dehydrogenase
CO2 CO2 CO2
a-methylbutyryl-CoA Isovaleryl-CoA Isobutyryl-CoA
malate
acetyl-CoA acetyl-CoA
acetoacetate

acetyl-CoA

malate
succinyl-CoA
Krebs
Cycle
fumarate

succinate
succinate

FIGURE 16.4: BCAA metabolism: isoleucine, leucine, and valine and their conversion to ketone bodies and
glucose.
280 Part III:Homeostatic Manipulators

A
BBB
amino acids (tryptophan, phenylalanine, tyros-
Brain
SERUM
ine) at the BBB. BCAAs compete with phenylala-
nine and tyrosine for brain entry, and studies in
healthy volunteers indicate that BCAAs produce
cognitive and endocrine effects consistent with
attenuation of central dopamine neurotransmis-
sion. When plasma BCAA concentrations rise,
which can occur in response to food ingestion
or BCAA administration, brain BCAA concen-
trations rise, and aromatic amino acids concen-
BCAA Aromatic aminoacids
trations decline. Such effects occur acutely and
chronically (Bastone et al., 1995; Berry et al.,
B 1982; Gijsman et al., 2002; Islam et al., 2010;
BBB
Brain Oldendorf, 1971; Sevy etal.,2006).
SERUM
BCAAs enter the brain via a transporter,
Decrease of located at the BBB on CNS capillary endothe-
Aromatic aminoacids
(TRP, PHE, TYR)
lial cells, that is almost fully saturated at normal
plasma amino acid concentrations and com-
petitively shared by a number of large neutral
amino acids, including the aromatic amino acids
(Oldendorf, 1971; Pardridge, 1983; Pardridge and
Decreased levels of
neurotransmitters Choi, 1986; Smith etal., 1987). As a consequence,
5HT, DE, NE
the ingestion of BCAAs causes rapid elevation
of their plasma concentrations, increases their
FIGURE 16.5: BCAAs compete with aromatic amino uptake into the brain, and decreases the brain
acids (phenylalanine, tyrosine and tryptophan) uptakes and levels of the aromatic amino acids
for brain entry. a. Under normal conditions, some (Figure16.5).
amount of BCAAs and aromatic amino acids cross
the blood brain barrier and enter the brain. b. When BCAA Applications inCNS Diseases
plasma BCAA concentrations rise, which can occur in BCAAs have been administered under a num-
response to food ingestion or BCAA administration, ber of circumstances to both healthy humans
brain BCAA concentrations rise, and aromatic amino and individuals with certain neurological dis-
acids decline. eases. A study done in patients with mania
demonstrated that acute administration of
BCAA decreased ratings of mania. The authors
BCAAs readily cross the BBB, with the influx of of this study suggested that the beneficial effect
leucine exceeding that of isoleucine and valine of BCAA to these patients could be explained
(Oldendorf, 1971; Smith et al., 1987; Yudkoff, through the decrease of tyrosine availability to
1997). Astrocytes, which are in close approxima- the brain (Scarna etal., 2003). Elevated tyrosine
tion to brain capillaries, are probably the initial availability to the brain may contribute to the
site of metabolism of leucine (Hannuniemi and development of central/mental fatigue during
Oja, 1981; Yudkoff etal., 1994). Amitochondrial and after sustained exercise. Supplementation
BCAA aminotransferase is very active in these with BCAAs should prevent the exercise-induced
cells. Indeed, from 30 to 50% of all -amino increase in the plasma concentration ratio of
groups of brain glutamate are derived from leu- free tryptophan to other large neutral amino
cine alone (Gijsman et al., 2002; Yudkoff et al., acids (including BCAAs) and thereby prevent
2005b). However, homeostatic mechanisms an elevation in the level of serotonin in the brain
maintain a very low intrasynaptic concentration (Hassmn etal.,1994).
of glutamate, partially accomplished by uptake BCAA supplementation enhances the cog-
and conversion of glutamate to glutamine inside nitive recovery of patients with severe TBI by
astrocytes (Yudkoff etal., 2005b). restoring hippocampal function after a TBI
BCAAs influence brain function not only (Aquilani etal., 2005; Cole etal., 2010). Aseries
by participating in glutamate homeostasis but of studies demonstrated that deficient clear-
also in other aspects. Thus they influence brain ance of the large neutral amino acid phenyla-
metabolism by modifying transport of aromatic lanine was associated with tardive dyskinesia,
Dietary Manipulations 281

that the administration of BCAAs significantly leucine (Kanamori et al., 1998; Sakai et al.,
decreased disease symptoms over placebo, and 2004; Yudkoff et al., 2005b). BCAAs also serve
that the observed symptom reduction was sig- as ammonia donors to glutamate, participating
nificantly correlated with a diminished avail- in the conversion of glutamate to glutamine in
ability of phenylalanine to the brain of adult men the glutamate-glutamine cycle (Evangeliou etal.,
with psychosis (Mosnik etal., 1997; Richardson 2009a; Yudkoff etal., 2005b). Proper function of
et al., 2006; Richardson et al., 1999; Schultz the glutamate-glutamine cycle is important not
etal.,2001). only in glutamate homeostasis but also in the
Older studies suggest a significant benefit prevention of epileptic seizures. An increasing
in terms of maintenance of extremity muscle number of studies have suggested that an abnor-
strength and continued ability to walk in patients mal amplification of glutamatergic activity is
with amyotrophic lateral sclerosis. The posi- involved in the pathophysiology of seizures and
tive effect for these patients could be explained certain types of medically refractory epilepsies
through the BCAAs, glutamate dehydrogenase (Bernard etal., 1997; Eid etal., 2008; Guo etal.,
activation, and, consequently, glutamate decrease 2010; Scimemi etal.,2006).
(Bastone et al., 1995; Plaitakis et al., 1988). However, it appears that for buffering
Evidence is quite mixed however, with some evi- of brain glutamateif levels of this excitatory
dence of toxicity, increased mortality, and lack of and potentially toxic neurotransmitter become
efficacy (Anonymous, 1993; Tandan etal.,1996). elevatedother mechanisms are recruited, such
as the BCAA effect on glutamate dehydrogenase
BCAA and Epilepsy (GDH). GDH is a mitochondrial enzyme link-
As the major excitatory neurotransmitter in the ing the Krebs cycle to the multifunctional amino
CNS of mammals, glutamate is essential for all acid glutamate. GDH catalyzes the reversible
of our behaviors (Meldrum, 2000; Shepherd and interconversion of glutamate to -ketoglutarate
Huganir, 2007; Usherwood et al., 1968). Proper and ammonia using NADP(H) and NAD(H) as
glutamate metabolism is necessary to ensure syn- cofactors, thus interconnecting amino acid and
aptogenesis, neurogenesis, neurite outgrowth, carbohydrate metabolism. Thereby GDH plays
and programmed cell death (Mattson, 2008). a pivotal role between carbohydrate and protein
Glutamate may have detrimental effects on neu- metabolisms, controlling production and con-
rons when present in excessive amounts and has sumption of the messenger molecule glutamate
been implicated in the pathogenesis of a number in neuroendocrine cells (Karaca et al., 2011;
of neurological disorders. Excessive sustained Spanaki etal.,2010).
activation of glutamate receptors can kill neu- In the past few years there has been increased
rons, particularly under conditions of reduced evidence that GDH dysfunction participates in
energy availability and increased oxidative stress, the pathophysiology of epileptogenesis. Clinical
and is one of the major mechanisms of epilep- and experimental data demonstrated that GDH
togenesis (Hampson and Manalo, 1998; Hazell, activity was significantly decreased in the tempo-
2007; Mattson, 2003; Zeng etal., 2007). Thus glu- ral cortex in patients with temporal lobe epilepsy,
tamate homeostasis is very important for proper and it is proposed that significant alterations in
neurotransmission. The brain must provide neu- the enzyme activities may contribute to decreased
rons with a constant supply of glutamate while at metabolism of glutamate, leading to its accumu-
the same time the intrasynaptic glutamate level lation (Kang et al., 2006; Malthankar-Phatak
must be kept low to maximize the signal-to-noise et al., 2006). In this point of view, BCAA may
ratio upon the release of glutamate from nerve help in glutamate buffering by activating gluta-
terminals to minimize the risk of excitotoxicity mate dehydrogenase and in this way converting
consequent to excessive glutamatergic stimula- excessive glutamate amounts to -ketoglutarate
tion of susceptible neurons (Sakai et al., 2004; (Plaitakis, 1989; Plaitakis etal., 1988). Our inte-
Yudkoff etal., 2005b). grated proposal of BCAA action on glutamate
Glutamate metabolism is accomplished metabolism is illustrated in Figure16.6.
through complicated biochemical pathways. Experimental evidence also shows that
BCAAs influence glutamate metabolism in BCAAs alone have antiepileptic properties. An
multiple ways. First, BCAAs (especially leu- infusion of a BCAA solution (300 mg/kg) has been
cine) provide neurons with a constant sup- found to increase the seizure threshold in rats to
ply of glutamate: 30% to 50% of all -amino the proconvulsant drug picrotoxin, an antagonist
groups of brain glutamine are derived from on the GABA-benzodiazepine receptor complex.
282 Part III:Homeostatic Manipulators

Postsynaptic-Neuron
Presynaptic-Neuron
-KG

GDH
glutamate glutamate

glutamine

BCAA
pool
NH3 glutamate Astrocyte

GDH
glutamine -KG

FIGURE 16.6: BCAAs effects on glutamate homeostasis: BCAAs may affect glutamate homeostasis in three
ways:1. BCAAs - especially leucine - provide neurons with a constant supply of glutamate; 2.The intrasynaptic
glutamate level must be kept low to maximize the signal-to-noise ratio upon the release of glutamate from nerve
terminals to minimize the risk of excitotoxicity consequent to excessive glutamatergic stimulation of susceptible
neurons. This is partially achieved by converting glutamate to glutamine inside the astrocytes; 3. BCAA may
help in glutamate buffering by activating glutamate dehydrogenase and thus convert excessive glutamate to
-ketoglutarate.

The effect was similar regardless of whether all despite the change in the fat-to-protein ratio
three BCAAs were given together or separately from 4:1 to2.5:1.
(Skeie etal., 1992; Skeie etal.,1994). Experimental and clinical data indicate
that BCAAs and the ketogenic diet share some
BCAA and KetogenicDiet common properties. First, both could favor
Over the past few decades, the ketogenic diet -aminobutyric acid (GABAergic) neurotrans-
has become one of our valuable weapons in the mission. It is well known that the ketogenic
management of intractable seizures. However, diet may limit the availability of oxaloacetate
not all patients have truly benefited from this to the aspartate aminotransferase reaction, an
therapy. In addition, the application of this diet important route of brain glutamate handling.
is limited by the fact that many patients cannot As a result, more glutamate becomes accessible
tolerate it while others present with side effects to the glutamate decarboxylase reaction to yield
(Freeman etal., 2007; Kang etal., 2004). Recent GABA, the major inhibitory neurotransmitter
efforts have been made to improve its tolerabil- and an important antiseizure (antiepileptic)
ity such as the application of the low glycemic agent (Yudkoff etal., 2004). These experimental
index treatment or the modified Atkins diet data are supported by a clinical trial in 26 chil-
(Kossoff et al., 2003; Pfeifer and Thiele, 2005). dren suffering from refractory epilepsy where
The addition of BCAA to the classic ketogenic the ketogenic diet increased the GABA lev-
diet appears to be a new promising advance- els in cerebrospinal fluid (Dahlin et al., 2005).
ment toward this effort. Work done in our However, experimental data from another
clinic demonstrated that the use of BCAA could research group demonstrated that BCAAs
increase the effectiveness of the ketogenic diet decreased the concentration of glutamate in
(Evangeliou etal., 2009a). From the same study, the thalamus and cortex without affecting
none of our patients had a remarkable reduc- GABA concentrations. A logical explanation
tion in the level of urine ketosis after the sup- of this phenomenon is the glutamate decrease
plementation of BCAAs. Moreover, in all of the through the BCAA cycle as proposed by Hutson
patients, no reduction in ketosis was recorded et al. (2001). BCAAs enter astrocytes where
Dietary Manipulations 283

transamination catalyzed by the mitochon- leucine as an adjunctive treatment to the


drial branched-chain aminotransferase pro- ketogenic diet is impossible because it is toxic
duces glutamate and branched-chain ketoacids. when consumed out of proportion to valine
Because branched-chain ketoacids are poorly and isoleucine. Downregulation of branched
oxidized in astrocytes, they exit astrocytes chain kinase and inhibition of branched
and can be taken up by neurons (Hutson etal., chain kinase by -ketoisocaproate activates
1998). Inside neurons, they are transaminated branched-chain -ketoacid dehydrogenase.
back to the BCAAs by the neuronal cytosolic Activated branched-chain -ketoacid dehy-
branched-chain aminotransferase, in a pro- drogenase depletes -ketoisovalerate and
cess consuming glutamate. These BCAAs exit -keto-L-methylvalerate and therefore also
neurons and return to astrocyte. Another pos- depletes valine and isoleucine. Lack of valine
sibility for decreasing glutamate is through and isoleucine inhibits protein synthesis. The
BCAA glutamate dehydrogenase activation consequence is that leucine should not be
(Plaitakis, 1989; Plaitakis et al., 1988). This consumed in large amounts without valine
aspect is supported by work in which glutamate and isoleucine, even though only leucine pro-
dehydrogenase was significantly decreased in motes protein synthesis (Scarna et al., 2005).
temporal, neocortical, and hippocampal tissues High dietary BCAAs should be well toler-
obtained from medically intractable epilepsy ated because of the reserve of branched chain
patients (Malthankar-Phatak etal., 2006). Thus -ketoacid dehydrogenase activity present
BCAAs, by decreasing the effects of glutamater- in tissues of the body. However, here also,
gic neurotransmission, could facilitate those theoretically the safety margin of maximally
of GABAergic neurotransmission (Dufour activated branched-chain -ketoacid dehydro-
et al., 2001b). In addition, the ketogenic diet genase may be exceeded at high BCAA intake.
appears to favor the synthesis of glutamine, There is a need, therefore, to know the maxi-
an essential precursor to GABA. This occurs mal capacity for BCAA disposal. Not enough
because ketone body carbon is metabolized to information is currently available to make such
glutamine and also because there is increased calculations (Harris etal.,2005).
consumption of acetate in ketosis, which is con- Another reason to be concerned with BCAA
verted quickly into glutamine in the brain by supplementation is the fact that our study is the
astrocytes. Similarly leucine, one of the main only clinical study on epileptic patients, and we
BCAAs, facilitates the production of glutamine. must be very cautious with their administra-
In particular, it serves as an ammoniac donor tion. Our concerns are supported by the fact that
in the formation of glutamine from glutamate. although the majority of publications discuss
Moreover, leucine is related to many more bio- the antiepileptic action of BCAAs, there is some
chemical reactions that take place in ketosis. experimental research addressing the epilepto-
Specifically, ketosis may favor the release of genic action of BCAAs in genetic absence epi-
glutamine from the brain, which is exchanged leptic rats of Strasbourg (GAERS; Dufour etal.,
for blood leucine at the BBB by specific trans- 2001a). Interestingly, in the same experimental
porters. Because brain glutamine is formed in model seizures are connected with increased
astrocytes from glutamate, the overall effect GDH expression in the thalamus of both young
would be to favor the release of glutamate and adult GAERS and in the cortex of young
from the nervous system (Yudkoff etal., 2005a; GAERS (Dutuit etal., 2000). This is in contrast
Yudkoff et al., 2004). These properties of leu- to what happens in patients with temporal lobe
cine are of paramount importance because a epilepsy, where the convulsions are connected to
delayed removal of glutamate in the brain leads low GDH levels and the BCAA could help by acti-
to increased levels of glutamate in neuronal vating GDH (Malthankar-Phatak etal.,2006).
synapses, which is one of the pathophysiologic Another question that needs to be answered
mechanisms implicated in epileptogenesis is related to the use of BCAAs as monotherapy
(Wong etal.,2002). in patients with epilepsy. There is no doubt that
Although encouraging, clinical data on the antiepileptic action of BCAAs is based only
an antiepileptic action of BCAA are still lim- on experimental data. Further studies, espe-
ited. Many questions need to be answered. cially those on patients with intractable epilepsy
How high can we go with the BCAA dose? receiving two to three antiepileptic regimens and
Could we provide leucine alone as the most not on the ketogenic diet, are required for more
ketotic of BCAAs? Providing exclusively concrete conclusions.
284 Part III:Homeostatic Manipulators

PROBIOTICS Furthermore, many studies have demon-


Complex organisms, including humans, devel- strated the restoration of microbiome intestinal
oped symbiosis as a common way to overcome and mucosa after administration of probiotics, and
survive with genetic and metabolic deficiencies in this sense probiotics can be useful to restore
(Resta, 2009; Tylianakis, 2009). Evolutionarily, the microbial balance in the intestine, to relieve
symbiosis has proven beneficial for all partners gastrointestinal problems, and to attenuate
considered and has become our way of life. As immunological abnormalities (Fond etal., 2015;
a result, Homo sapiens largest organ is the Rook etal., 2014). It is well known that much of
gut microbiota, comprising two major divi- the immune system is located in the intestine
sions of bacteria, Bacteroidetes and Firmicutes and that children with autism disorders suf-
(Neish, 2009; Resta, 2009). The recognition that fer from immune system dysfunction (Samsam
the gut microbiota influences several signaling etal.,2014).
pathways led to the suggestion of the concept Probiotics can effect microbiota composition
of a microbiotagutbrain axis (Montiel-Castro and intestinal barrier function and alter intes-
etal., 2013). The gutbrain interation is the bidi- tinal immune responses, thus improving the
rectional communication between the gut and gutbrainmicrobiota axis (Butel, 2014; Smith
the brain that occurs through multiple pathways etal., 2014). The administration of probiotic bac-
that include hormonal, neural, and immune teria to address changes in the microbiota might,
mediators (De Palma etal.,2014). therefore, be a useful novel therapeutic tool with
Gut microbiota have a very important role which to restore normal gut microbiota, reduce
in stress situations. Exposure to stress results in inflammation, restore epithelial barrier func-
alterations of the braingut axis ultimately lead- tion, and potentially ameliorate behavioral
ing to the development of a broad array of gas- symptoms associated with some children with
trointestinal disorders, including negative effects autism (Critchfield et al., 2011; Hemarajata and
on intestinal microbiota (Konturek et al., 2011). Versalovic, 2013; Szajewska, 2013). Although
Accordingly, one area of flourishing research there are many experimental studies that dem-
involves the relationship between the intesti- onstrate the beneficial effect of probiotics on
nal microbiota (as well as the related functional animal models, there are no randomized clini-
integrity of the gastrointestinal tract) and mental cal trials to prove this in humans (Gilbert etal.,
health (Bested etal., 2013a, 2013b, 2013c).Recent 2013; Hsiao etal., 2013). In an ongoing study at
clinical studies demonstrate that the adminis- our clinic, probiotics were administered to 30
tration of beneficial microbes, through supple- children, ages 4 to 10years, with autism. Sixteen
mentation and/or fecal microbial transplant, can of these children had gastrointestinal symptoms
affect end-points related to mood state (glycemic and 14 did not. Probiotics had beneficial effects in
control, oxidative status, uremic toxins), brain both groups of patients. Significant improvement
function (functional MRI), and mental outlook (>12 units on the Childhood Autism Rating Scale)
(depression, anxiety, autism; Bested etal., 2013c; was recorded in 5 patients (pre-scale: 36.00
Chen etal.,2013). 1.39 [mean SD]) from the group with intestinal
symptoms and in 4 from the group with no intes-
Probiotics andAutism tinal symptoms (pre-scale 34.1 1.21; Evangeliou
Children with autism often exhibit symptoms of A, Asprangathou D, Karanika E, Spilioti M,
the gastrointestinal system (Adams et al., 2011; unpublished data). Although these data are very
McElhanon etal., 2014). These symptoms include preliminary, there is a body of emerging evidence
diarrhea, constipation, vomiting/reflux, abdomi- that probiotics may be used in autistic behavior
nal pain/discomfort, gaseousness, and unusually as an additional or alternative therapy.
foul-smelling stools (Chaidez etal., 2014; Hsiao,
2014; Kang et al., 2014). In the past few years, O M E G A - 3 FAT T Y A C I D S
increasing numbers of studies have implicated Omega-3 fatty acids are polyunsaturated fatty
correlation between not only gastrointestinal acids, and three main types are found in the
but also other autistic symptoms and abnor- human diet:-linolenic acid (ALA), docosahex-
mal intestinal probiota (Kang etal., 2013; Wang aenoic acid (DHA), and eicosapentaenoic acid
et al., 2013). In a very recent study, the partici- (EPA). DHA and EPA are found in seafood,
pants demonstrated strong positive correlation of while ALA is found in nut and plant oils (Bent
autism severity with the severity of gastrointesti- et al., 2009; Zhang et al., 2011). Interestingly,
nal dysfunction (Tomova etal.,2015). fish do not produce EPA and DHA, but the oils
Dietary Manipulations 285

are synthesized by single-cell marine organ- fatty acids may be especially important in
isms that are eaten by fish (Harris, 2004). While patients with psychiatric disorders, due to
the human body can synthesize both DHA and high prevalence rates of smoking and obesity
EPA from ALA, it cannot synthesize any of these and the metabolic side effects of some psycho-
three types of fatty acids ex novo, and, accord- tropic medications (Freeman etal., 2006). Thus
ingly, these substances are dietary essentials while it is not currently possible to recommend
(Bent etal., 2009; Freeman etal., 2006). Omega-3 omega-3 polyunsaturated fatty acids as either
fatty acids are critical to brain development and a mono- or adjunctive therapy in any mental
function (Denis et al., 2013; Innis, 2008; Zhang illness, the available evidence is strong enough
et al., 2011). Lack of these fatty acids has been to justify continued study, especially with
implicated in a number of mental health condi- regard to attentional, anxiety, and mood disor-
tions over the lifespan, from developmental dis- ders (Ross etal., 2007). Moreover, a number of
orders and mental retardation in childhood to clinical studies have shown omega-3 fatty acids
depression, bipolar disorder, schizophrenia, and are essential for normal infant vision devel-
borderline personality disorder; stress, hostil- opment, and adequate amounts of DHA and
ity, and aggression in adulthood; and cognitive other omega-3 fatty acids in the diet of preg-
decline, dementia, and Alzheimers disease in nant women also appear to be important in
late adulthood (Andreeva etal., 2011; Bent etal., normal infant vision development (Dennehy,
2011; Fotuhi et al., 2009; Marano et al., 2013; 2011; Hoffman et al., 2009; SanGiovanni
Rondanelli etal., 2010, 2011). Agrowing body of etal.,2000).
clinical research trials has been conducted with
variable outcomes to all these situations. Thus in CONCLUSION
the past 30years a substantial number of obser- Here we have outlined a diverse array of dietary
vational and epidemiological studies have sug- manipulatons that can impact brain homeo-
gested that mental illness, in particular mood stasis directly or indirectly. We hope and
disorders, is associated with reduced dietary expect much new research is on the horizon
intake and/or cellular abundance of omega-3 due to increased patient interest and along-
polyunsaturated fatty acids (Ross etal.,2007). side the potential for limited toxicity and the
Despite the many studies that have been done, broad implications for practical and affordable
the results are questionable. A meta-analysis treatment.
of randomized, placebo-controlled trials of
References
omega-3 fatty acid treatment of major depres-
Adams, J.B., Johansen, L.J., Powell, L.D., Quig, D.,
sive disorder to determine efficacy and examine
and Rubin, R.A. (2011). Gastrointestinal flora and
sources of heterogeneity between trials suggested
gastrointestinal status in children with autism
a small, nonsignificant benefit of omega-3 fatty
comparisons to typical children and correlation
acids for major depression. The authors of the
with autism severity. BMC Gastroenterol. 11,22.
same study suggested that nearly all of the treat- Adcock, K.H., Nedelcu, J., Loenneker, T., Martin, E.,
ment efficacy observed in the published literature Wallimann, T., and Wagner, B.P. (2002).
may be attributable to publication bias (Bloch Neuroprotection of creatine supplementa-
and Hannestad,2012). tion in neonatal rats with transient cerebral
Moreover, in another review of the published hypoxia-ischemia. Dev. Neurosci. 24, 382388.
literature on the effects of omega-3 fatty acids on Ahlborg, G., Felig, P., Hagenfeldt, L., Hendler, R.,
measures of cognitive function in normal aging, and Wahren, J. (1974). Substrate turnover dur-
incidence and treatment of dementia suggested ing prolonged exercise in man: Splanchnic and
that the available data are insufficient to draw leg metabolism of glucose, free fatty acids, and
strong conclusions about the effects of omega-3 amino acids. J.Clin. Invest. 53, 10801090.
fatty acids on cognitive function in normal aging Al-Aama, T., Brymer, C., Gutmanis, I.,
or on the incidence or treatment of dementia; Woolmore-Goodwin, S.M., Esbaugh, J., and
however, also from the same study, the authors Dasgupta, M. (2011). Melatonin decreases
proposed that limited evidence suggests a possi- delirium in elderly patients: A random-
ble association between omega-3 fatty acids and ized, placebo-controlled trial. Int. J. Geriatr.
reduced risk of dementia (Issa etal.,2006). Psychiatry 26, 687694.
Despite the aforementioned concerns and Al-Omary, F.A. (2013). Melatonin: Comprehensive
the lack of sufficient randomized double-blind profile. Profiles Drug Subst. Excip. Relat.
studies, the health benefits of omega-3 essential Methodol. 38, 159226.
286 Part III:Homeostatic Manipulators

Allen, P.J. (2012). Creatine metabolism and psychi- Bastone, A., Micheli, A., Beghi, E., and Salmona, M.
atric disorders: Does creatine supplementation (1995). The imbalance of brain large-chain
have therapeutic value? Neurosci. Biobehav. Rev. aminoacid availability in amyotrophic lateral
36, 14421462. sclerosis patients treated with high doses of
Ando, S., Tadenuma, T., Tanaka, Y., Fukui, F., branched-chain aminoacids. Neurochem. Int.
Kobayashi, S., Ohashi, Y., and Kawabata, T. 27, 467472.
(2001). Enhancement of learning capacity and Bard, E., and Braissant, O. (2010). Synthesis and
cholinergic synaptic function by carnitine in transport of creatine in the CNS:Importance for
aging rats. J. Neurosci. Res. 66, 266271. cerebral functions. J.Neurochem. 115, 297313.
Andreeva, V.A., Kesse-Guyot, E., Barberger-Gateau, P., Beghi, E., Pupillo, E., Bonito, V., Buzzi, P.,
Fezeu, L., Hercberg, S., and Galan, P. (2011). Caponnetto, C., Chi, A., Corbo, M., Giannini, F.,
Cognitive function after supplementation Inghilleri, M., Bella, V.L., et al. (2013).
with B vitamins and long-chain omega-3 fatty Randomized double-blind placebo-controlled
acids:Ancillary findings from the SU.FOL.OM3 trial of acetyl-L-carnitine for ALS. Amyotroph.
randomized trial. Am. J.Clin. Nutr. 94, 278286. Lateral Scler. Frontotemporal Degener. 14,
Andres, R.H., Huber, A.W., Schlattner, U., 397405.
Prez-Bouza, A., Krebs, S.H., Seiler, R.W., Bender, A., Samtleben, W., Elstner, M., and
Wallimann, T., and Widmer, H.R. (2005). Effects Klopstock, T. (2008). Long-term creatine supple-
of creatine treatment on the survival of dopami- mentation is safe in aged patients with Parkinson
nergic neurons in cultured fetal ventral mesence- disease. Nutr. Res. (NewYork) 28, 172178.
phalic tissue. Neuroscience 133, 701713. Bent, S., Bertoglio, K., Ashwood, P., Bostrom, A.,
Anil, M., Helvaci, M., Ozbal, E., Kalenderer, O., Anil, and Hendren, R.L. (2011). A pilot randomized
A.B., and Dilek, M. (2009). Serum and muscle controlled trial of omega-3 fatty acids for autism
carnitine levels in epileptic children receiving spectrum disorder. J. Autism Dev. Disord. 41,
sodium valproate. J. Child Neurol. 24,8086. 545554.
Anonymous (1993). Branched-chain amino acids Bent, S., Bertoglio, K., and Hendren, R.L. (2009).
and amyotrophic lateral sclerosis: A treatment Omega-3 fatty acids for autistic spectrum disor-
failure? The Italian ALS Study Group. Neurology der:Asystematic review. J.Autism Dev. Disord.
43, 24662470. 39, 11451154.
Anthony, J.C., Anthony, T.G., Kimball, S.R., and Bera, S., Wallimann, T., Ray, S., and Ray, M. (2008).
Jefferson, L.S. (2001). Signaling pathways involved Enzymes of creatine biosynthesis, arginine and
in translational control of protein synthesis in methionine metabolism in normal and malig-
skeletal muscle by leucine. J. Nutr. 131, 856S-860S. nant cells. FEBS J. 275, 58995909.
Aquilani, R., Iadarola, P., Contardi, A., Boselli, M., Berezowski, V., Miecz, D., Marszaek, M., Brer, A.,
Verri, M., Pastoris, O., Boschi, F., Arcidiaco, P., Brer, S., Cecchelli, R., and Naecz, K.A. (2004).
and Viglio, S. (2005). Branched-chain amino Involvement of OCTN2 and B0,+ in the transport
acids enhance the cognitive recovery of patients of carnitine through an in vitro model of the
with severe traumatic brain injury. Arch. Phys. blood-brain barrier. J.Neurochem. 91, 860872.
Med. Rehabil. 86, 17291735. Bernard, C., Hirsch, J., and Ben-Ari, Y. (1997).
Arockia Rani, P.J., and Panneerselvam, C. (2001). Glutamatergic excitatory receptors and temporal
Carnitine as a free radical scavenger in aging. lobe epilepsy. Rev. Neurol. 153 Suppl 1, S1424.
Exp. Gerontol. 36, 17131726. Berry, H.K., Bofinger, M.K., Hunt, M.M., Phillips, P.J.,
Assaf, N., Shalby, A.B., Khalil, W.K., and Ahmed, H.H. and Guilfoile, M.B. (1982). Reduction of cerebro-
(2012). Biochemical and genetic alterations of spinal fluid phenylalanine after oral administra-
oxidant/antioxidant status of the brain in rats tion of valine, isoleucine, and leucine. Pediatr.
treated with dexamethasone: Protective roles Res. 16, 751755.
of melatonin and acetyl-L-carnitine. J. Physiol. Bested, A.C., Logan, A.C., and Selhub, E.M.
Biochem. 68,7790. (2013a). Intestinal microbiota, probiotics and
Bain, K.T. (2006). Management of chronic insomnia mental health: From Metchnikoff to modern
in elderly persons. Am. J.Geriatr. Pharmacother. advances:Part Iautointoxication revisited. Gut
4, 168192. Pathog5,5.
Balestrino, M., Lensman, M., Parodi, M., Perasso, L., Bested, A.C., Logan, A.C., and Selhub, E.M.
Rebaudo, R., Melani, R., Polenov, S., and Cupello, A. (2013b). Intestinal microbiota, probiotics and
(2002). Role of creatine and phosphocreatine in mental health: From Metchnikoff to modern
neuronal protection from anoxic and ischemic advances: Part IIcontemporary contextual
damage. Amino Acids 23, 221229 research. Gut Pathog.5,3.
Dietary Manipulations 287

Bested, A.C., Logan, A.C., and Selhub, E.M. Brosnan, M.E., Edison, E.E., da Silva, R., and
(2013c). Intestinal microbiota, probiotics and Brosnan, J.T. (2007). New insights into creatine
mental health: From Metchnikoff to modern function and synthesis. Adv. Enzyme Regul. 47,
advances:Part IIIconvergence toward clinical 252260.
trials. Gut Pathog.5,4. Bubenik, G.A., and Konturek, S.J. (2011). Melatonin
Bianchetti, A., Rozzini, R., and Trabucchi, M. (2003). and aging: Prospects for human treatment.
Effects of acetyl-L-carnitine in Alzheimers dis- J.Physiol. Pharmacol. 62,1319.
ease patients unresponsive to acetylcholinester- Butel, M.J. (2014). Probiotics, gut microbiota and
ase inhibitors. Curr. Med. Res. Opin. 19, 350353. health. Med. Mal. Infect. 44,18.
Binienda, Z., Przybyla-Zawislak, B., Virmani, A., Calabrese, V., Cornelius, C., Stella, A.M., and
and Schmued, L. (2005). L-carnitine and neuro- Calabrese, E.J. (2010). Cellular stress responses,
protection in the animal model of mitochondrial mitostress and carnitine insufficiencies as criti-
dysfunction. Ann. N.Y. Acad. Sci. 1053, 174182. cal determinants in aging and neurodegenera-
Binienda, Z.K. (2003). Neuroprotective effects of tive disorders: Role of hormesis and vitagenes.
L-carnitine in induced mitochondrial dysfunc- Neurochem. Res. 35, 18801915.
tion. Ann. N.Y. Acad. Sci.993, 289295. Carvalho, L.A., Gorenstein, C., Moreno, R., Pariante, C.,
Bixel, M., Shimomura, Y., Hutson, S., and Hamprecht, B. and Markus, R.P. (2009). Effect of antidepressants
(2001). Distribution of key enzymes of on melatonin metabolite in depressed patients.
branched-chain amino acid metabolism in glial J.Psychopharmacol. (Oxford) 23, 315321.
and neuronal cells in culture. J. Histochem. Cecon, E., and Markus, R.P. (2011). Relevance of
Cytochem. 49, 407418. the chronobiological and non-chronobiological
Blader, J.C., Koplewicz, H.S., Abikoff, H., and Foley, C. actions of melatonin for enhancing therapeutic
(1997). Sleep problems of elementary school efficacy in neurodegenerative disorders. Recent
children. A community survey. Arch. Pediatr. Pat. Endocr. Metab. Immune Drug Discov.
Adolesc. Med. 151, 473480. 5,9199.
Bloch, M.H., and Hannestad, J. (2012). Omega-3 fatty Cendes, F., Caramanos, Z., Andermann, F., Dubeau, F.,
acids for the treatment of depression:Systematic and Arnold, D.L. (1997). Proton magnetic reso-
review and meta-analysis. Mol. Psychiatry 17, nance spectroscopic imaging and magnetic reso-
12721282. nance imaging volumetry in the lateralization of
Bhmer, T., Ben, A., and Hymork, S.C. (2010). temporal lobe epilepsy:Aseries of 100 patients.
Valproate-induced hyperammonemic encepha- Ann. Neurol. 42, 737746.
lopathy, rapidly improved by i.v. carnitine and Chaidez, V., Hansen, R.L., and Hertz-Picciotto, I.
glucose/thiamine. Scand. J. Gastroenterol. 45, (2014). Gastrointestinal problems in children
762763. with autism, developmental delays or typi-
Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F., cal development. J. Autism Dev. Disord. 44,
Gawryluk, J.W., Greene, J.G., Shaw, R., Smith, Y., 11171127.
Geiger, J.D., and Dingledine, R.J. (2006). Chapela, S.P., Kriguer, N., Fernndez, E.H., and
Mitochondrial biogenesis in the anticonvulsant Stella, C.A. (2009). Involvement of L-carnitine
mechanism of the ketogenic diet. Ann. Neurol. in cellular metabolism:Beyond Acyl-CoA trans-
60, 223235. port. Mini Rev. Med. Chem. 19, 15181526.
Boyce, P., and Hopwood, M. (2013). Manipulating Chen, X., DSouza, R., and Hong, S.T. (2013). The role
melatonin in managing mood. Acta Psychiatr. of gut microbiota in the gut-brain axis:Current
Scand.,1623. challenges and perspectives. Protein Cell 4,
Braissant, O. (2012). Creatine and guanidinoacetate 403414.
transport at blood-brain and blood-cerebrospinal Cole, J.T., Mitala, C.M., Kundu, S., Verma, A.,
fluid barriers. J.Inherit. Metab. Dis. 35, 655664. Elkind, J.A., Nissim, I., and Cohen, A.S. (2010).
Braissant, O., Henry, H., Bard, E., and Uldry, J. Dietary branched chain amino acids amelio-
(2011). Creatine deficiency syndromes and the rate injury-induced cognitive impairment. Proc.
importance of creatine synthesis in the brain. Natl. Acad. Sci. U.S.A. 107, 366371.
Amino Acids 40, 13151324. Cortesi, F., Giannotti, F., Sebastiani, T., Panunzi,
Brosnan, J.T., and Brosnan, M.E. (2006). S., and Valente, D. (2012). Controlled-release
Branched-chain amino acids:Enzyme and sub- melatonin, singly and combined with cognitive
strate regulation. J.Nutr. 136, 207S-211S. behavioural therapy, for persistent insomnia in
Brosnan, J.T., da Silva, R.P., and Brosnan, M.E. children with autism spectrum disorders:Aran-
(2011). The metabolic burden of creatine synthe- domized placebo-controlled trial. J.Sleep Res. 21,
sis. Amino Acids 40, 13251331. 700709.
288 Part III:Homeostatic Manipulators

Costell, M., OConnor, J.E., and Grisolia, S. (1989). In: Carnitine today, C. De Simone, and G.
Age-dependent decrease of carnitine content in Famularo, eds. (NewYork:Chapman &Hall).
muscle of mice and humans. Biochem. Biophys. Dijk, D.J., Duffy, J.F., Riel, E., Shanahan, T.L., and
Res. Commun. 161, 11351143. Czeisler, C.A. (1999). Ageing and the circadian
Couturier, J.L., Speechley, K.N., Steele, M., Norman, R., and homeostatic regulation of human sleep dur-
Stringer, B., and Nicolson, R. (2005). Parental ing forced desynchrony of rest, melatonin and
perception of sleep problems in children of temperature rhythms. J.Physiol. 516, 611627.
normal intelligence with pervasive develop- Doisaki, M., Katano, Y., Nakano, I., Hirooka, Y.,
mental disorders: Prevalence, severity, and pat- Itoh, A., Ishigami, M., Hayashi, K., Goto, H.,
tern. J.Am. Acad. Child Adolesc. Psychiatry 44, Fujita, Y., Kadota, Y., et al. (2010). Regulation
815822. of hepatic branched-chain a-keto acid dehy-
Critchfield, J.W., van Hemert, S., Ash, M., Mulder, L., drogenase kinase in a rat model for type 2 dia-
and Ashwood, P. (2011). The potential role of pro- betes mellitus at different stages of the disease.
biotics in the management of childhood autism Biochem. Biophys. Res. Commun. 393, 303307.
spectrum disorders. Gastroenterol. Res. Pract. Doyen, C., Mighiu, D., Kaye, K., Colineaux, C.,
2011, 161358. Beaumanoir, C., Mouraeff, Y., Rieu, C., Paubel, P.,
Cunnington, D., Junge, M.F., and Fernando, A.T. and Contejean, Y. (2011). Melatonin in children
(2013). Insomnia:Prevalence, consequences and with autistic spectrum disorders: Recent and
effective treatment. Med. J.Aust. 199, S3640. practical data. Eur. Child Adolesc. Psychiatry 20,
Dahlin, M., Elfving, A., Ungerstedt, U., and mark, P. 231239.
(2005). The ketogenic diet influences the levels of Dubocovich, M.L., Delagrange, P., Krause, D.N.,
excitatory and inhibitory amino acids in the CSF Sugden, D., Cardinali, D.P., and Olcese, J.
in children with refractory epilepsy. Epilepsy (2010). International Union of Basic and Clinical
Res. 64, 115125. Pharmacology. LXXV. Nomenclature, classifica-
De Grandis, D. (2007). Acetyl-L-carnitine for the tion, and pharmacology of G protein-coupled
treatment of chemotherapy-induced peripheral melatonin receptors. Pharmacol. Rev. 62,
neuropathy:Ashort review. CNS Drugs 21 Suppl 343380.
1,3943. Dufour, F., Nalecz, K.A., Nalecz, M.J., and Nehlig, A.
De Palma, G., Collins, S.M., Bercik, P., and Verdu, (2001a). Metabolic approach of absence sei-
E.F. (2014). The microbiota-gut-brain axis in gas- zures in a genetic model of absence epilepsy, the
trointestinal disorders: Stressed bugs, stressed GAERS: Study of the leucine-glutamate cycle.
brain or both? J. Physiol. 592, 29892997. J.Neurosci. Res. 66, 923930.
De Vivo, D.C., Bohan, T.P., Coulter, D.L., Dreifuss, F.E., Dufour, F., Nalecz, K.A., Nalecz, M.J., and Nehlig, A.
Greenwood, R.S., Nordli, D.R., Jr., Shields, W.D., (2001b). Modulation of absence seizures by
Stafstrom, C.E., and Tein, I. (1998). L-carnitine branched-chain amino acids: Correlation with
supplementation in childhood epilepsy: Current brain amino acid concentrations. Neurosci. Res.
perspectives. Epilepsia 39, 12161225. 40, 255263.
Dechent, P., Pouwels, P.J., Wilken, B., Hanefeld, F., Dutuit, M., Didier-Bazs, M., Vergnes, M., Mutin,
and Frahm, J. (1999). Increase of total creatine M., Conjard, A., Akaoka, H., Belin, M.F., and
in human brain after oral supplementation Touret, M. (2000). Specific alteration in the
of creatine-monohydrate. Am. J. Physiol. 277, expression of glial fibrillary acidic protein, glu-
R698704. tamate dehydrogenase, and glutamine synthe-
Denis, I., Potier, B., Vancassel, S., Heberden, C., and tase in rats with genetic absence epilepsy. Glia
Lavialle, M. (2013). Omega-3 fatty acids and 32,1524.
brain resistance to ageing and stress: Body of Edison, E.E., Brosnan, M.E., Meyer, C., and Brosnan,
evidence and possible mechanisms. Ageing Res. J.T. (2007). Creatine synthesis: Production of
Rev. 12, 579594. guanidinoacetate by the rat and human kidney
Dennehy, C. (2011). Omega-3 fatty acids and ginger in vivo. Am. J.Physiol. 293, F17991804.
in maternal health:Pharmacology, efficacy, and Eid, T., Williamson, A., Lee, T.S., Petroff, O.A.,
safety. J.Midwifery Womens Health 56, 584590. and de Lanerolle, N.C. (2008). Glutamate and
Dhillon, S., Hellings, J.A., and Butler, M.G. (2011). astrocyteskey players in human mesial tem-
Genetics and mitochondrial abnormalities in poral lobe epilepsy? Epilepsia 49 Suppl 2,4252.
autism spectrum disorders: A review. Curr. Ellis, J.G., Perlis, M.L., Neale, L.F., Espie, C.A., and
Genomics 12, 322332. Bastien, C.H. (2012). The natural history of
Di Lisa, F., Barbato, L., Menabo, R., and Siliprandi, N. insomnia:Focus on prevalence and incidence of
(1997). Carnitine and mitochondrial dysfunction. acute insomnia. J.Psychiatr. Res. 46, 12781285.
Dietary Manipulations 289

Evangeliou, A., Spilioti, M., Doulioglou, V., Freeman, M.P., Hibbeln, J.R., Wisner, K.L., Davis, J.M.,
Kalaidopoulou, P., Ilias, A., Skarpalezou, A., Mischoulon, D., Peet, M., Keck, P.E., Jr.,
Katsanika, I., Kalamitsou, S., Vasilaki, K., Marangell, L.B., Richardson, A.J., Lake, J., etal.
Chatziioanidis, I., etal. (2009a). Branched chain (2006). Omega-3 fatty acids: Evidence basis for
amino acids as adjunctive therapy to ketogenic treatment and future research in psychiatry.
diet in epilepsy: Pilot study and hypothesis. J.Clin. Psychiatry 67, 19541967.
J.Child Neurol. 24, 12681272. Frye, R.E., Delatorre, R., Taylor, H., Slattery, J.,
Evangeliou, A., Vasilaki, K., Karagianni, P., and Melnyk, S., Chowdhury, N., and James, S.J.
Nikolaidis, N. (2009b). Clinical applications of (2013a). Redox metabolism abnormalities in
creatine supplementation on paediatrics. Curr. autistic children associated with mitochondrial
Pharm. Biotechnol. 10, 683690. disease. Transl. Psychiatry 3,e273.
Evangeliou, A., and Vlassopoulos, D. (2003). Frye, R.E., Rossignol, D., Casanova, M.F., Brown,
Carnitine metabolism and deficitwhen G.L., Martin, V., Edelson, S., Coben, R., Lewine, J.,
supplementation is necessary? Curr. Pharm. Slattery, J.C., Lau, C., etal. (2013b). A review of
Biotechnol. 4, 211219. traditional and novel treatments for seizures in
Evangeliou, A.E. (1992). Entwicklung einer einfachen autism spectrum disorder:Findings from a sys-
dunnschichtchromatographischen Methode zur tematic review and expert panel. Front. Public
Bestimmung der Carnitinesterausscheidung im Health1,31.
Urin in der Diagnostic angeborener und rewor- Galli-Carminati, G., Deriaz, N., and Bertschy, G.
bener Storungen der Intermediarstoffwechsels. (2009). Melatonin in treatment of chronic sleep
PhD diss., Johann Wolfgang Goethe-Universitt disorders in adults with autism:Aretrospective
Frankfurt amMain. study. Swiss Med. Wkly 139, 293296.
Feller, A.G., and Rudman, D. (1988). Role of carni- Gehrman, P.R., Connor, D.J., Martin, J.L., Shochat, T.,
tine in human nutrition. J.Nutr. 118, 541547. Corey-Bloom, J., and Ancoli-Israel, S. (2009).
Ferracioli-Oda, E., Qawasmi, A., and Bloch, M.H. Melatonin fails to improve sleep or agitation in
(2013). Meta-analysis: Melatonin for the treat- double-blind randomized placebo-controlled
ment of primary sleep disorders. PLOS One 8, trial of institutionalized patients with Alzheimer
e63773. disease. Am. J.Geriatr. Psychiatry 17, 166169.
Fischer, T.W., Slominski, A., Zmijewski, M.A., Reiter, Geier, D.A., and Geier, M.R. (2013). L-carnitine
R.J., and Paus, R. (2008). Melatonin as a major exposure and mitochondrial function in human
skin protectant:From free radical scavenging to neuronal cells. Neurochem. Res. 38, 23362341.
DNA damage repair. Exp. Dermatol. 17, 713730. Geier, D.A., Kern, J.K., Davis, G., King, P.G., Adams, J.B.,
Flanagan, J.L., Simmons, P.A., Vehige, J., Willcox, Young, J.L., and Geier, M.R. (2011). A prospec-
M.D., and Garrett, Q. (2010). Role of carnitine in tive double-blind, randomized clinical trial of
disease. Nutr. Metabol.7,30. levocarnitine to treat autism spectrum disorders.
Fond, G., Boukouaci, W., Chevalier, G., Regnault, A., Med. Sci. Monit. 17, PI1523.
Eberl, G., Hamdani, N., Dickerson, F., Macgregor, Gijsman, H.J., Scarna, A., Harmer, C.J., McTavish,
A., Boyer, L., Dargel, A., etal. (2015). The psy- S.B., Odontiadis, J., Cowen, P.J., and Goodwin,
chomicrobiotic: Targeting microbiota in major G.M. (2002). A dose-finding study on the
psychiatric disorders: A systematic review. effects of branch chain amino acids on sur-
Pathol. Biol. (Paris) 63(1):35-42 rogate markers of brain dopamine function.
Foster, D.W. (2004). The role of the carnitine sys- Psychopharmacology 160, 192197.
tem in human metabolism. Ann. N.Y. Acad. Sci. Gilbert, J.A., Krajmalnik-Brown, R., Porazinska, D.L.,
1033,116. Weiss, S.J., and Knight, R. (2013). Toward effec-
Fotuhi, M., Mohassel, P., and Yaffe, K. (2009). Fish tive probiotics for autism and other neurodevel-
consumption, long-chain omega-3 fatty acids opmental disorders. Cell 155, 14461448.
and risk of cognitive decline or Alzheimer dis- Glotzner, F.L., Planner, M., and Gaab, M. (1979).
ease:Acomplex association. Nat. Clin. Pract. 5, Creatine kinase in serum after grand mal sei-
140152. zures. Eur. Neurol. 18, 399404.
Francaux, M., and Poortmans, J.R. (1999). Effects Goldman, S.E., Richdale, A.L., Clemons, T., and
of training and creatine supplement on muscle Malow, B.A. (2012). Parental sleep concerns in
strength and body mass. Eur. J.Applied Physiol. autism spectrum disorders: Variations from
Occup. Physiol. 80, 165168. childhood to adolescence. J.Autism Dev. Disord.
Freeman, J.M., Kossoff, E.H., and Hartman, A.L. 42, 531538.
(2007). The ketogenic diet: One decade later. Gunol, F., Godbout, R., Nicolas, A., Franco, P.,
Pediatrics 119, 535543. Claustrat, B., and Baleyte, J.M. (2011). Melatonin
290 Part III:Homeostatic Manipulators

for disordered sleep in individuals with autism muscle of normal subjects by creatine supple-
spectrum disorders: Systematic review and dis- mentation. Clin. Sci. (Lond.) 83, 367374.
cussion. Sleep Med. Rev. 15, 379387. Harris, W.S. (2004). Fish oil supplementa-
Guerrero-Ontiveros, M.L., and Wallimann, T. tion: Evidence for health benefits. Cleve. Clin.
(1998). Creatine supplementation in health and J.Med. 71, 208210, 212, 215208 passim.
disease. Effects of chronic creatine ingestion in Hass, C.J., Collins, M.A., and Juncos, J.L. (2007).
vivo:Down-regulation of the expression of cre- Resistance training with creatine monohy-
atine transporter isoforms in skeletal muscle. drate improves upper-body strength in patients
Mol. Cell. Biochem. 184, 427437. with Parkinson disease: A randomized trial.
Guevara-Campos, J., Gonzlez-Guevara, L., Neurorehabil. Neural Repair 21, 107115.
Puig-Alcaraz, C., and Cauli, O. (2013). Autism Hassmn, P., Blomstrand, E., Ekblom, B., and
spectrum disorders associated to a deficiency of Newsholme, E.A. (1994). Branched-chain amino
the enzymes of the mitochondrial respiratory acid supplementation during 30-km competitive
chain. Metab. Brain Dis. 28, 605612. run:Mood and cognitive performance. Nutrition
Guo, F., Sun, F., Yu, J.L., Wang, Q.H., Tu, D.Y., (Burbank) 10, 405410.
Mao, X.Y., Liu, R., Wu, K.C., Xie, N., Hao, L.Y., Hausmann, O.N., Fouad, K., Wallimann, T., and
etal. (2010). Abnormal expressions of glutamate Schwab, M.E. (2002). Protective effects of oral
transporters and metabotropic glutamate recep- creatine supplementation on spinal cord injury
tor 1 in the spontaneously epileptic rat hippo- in rats. Spinal Cord 40, 449456.
campus. Brain Res. Bull. 81, 510516. Hazell, A.S. (2007). Excitotoxic mechanisms in
Hadjixenofontos, A., Schmidt, M.A., Whitehead, P.L., stroke: An update of concepts and treatment
Konidari, I., Hedges, D.J., Wright, H.H., strategies. Neurochem. Int. 50, 941953.
Abramson, R.K., Menon, R., Williams, S.M., Hemarajata, P., and Versalovic, J. (2013). Effects of
Cuccaro, M.L., et al. (2013). Evaluating mito- probiotics on gut microbiota: Mechanisms of
chondrial DNA variation in autism spectrum intestinal immunomodulation and neuromodu-
disorders. Ann. Hum. Genet. 77,921. lation. Ther. Adv. Gastroenterol. 6,3951.
Hamed, S.A., and Abdella, M.M. (2009). The risk Hersch, S.M., Gevorkian, S., Marder, K., Moskowitz, C.,
of asymptomatic hyperammonemia in chil- Feigin, A., Cox, M., Como, P., Zimmerman, C.,
dren with idiopathic epilepsy treated with val- Lin, M., Zhang, L., et al. (2006). Creatine in
proate: Relationship to blood carnitine status. Huntington disease is safe, tolerable, bioavail-
Epilepsy Res. 86,3241. able in brain and reduces serum 8OH2dG.
Hampson, D.R., and Manalo, J.L. (1998). The activa- Neurology 66, 250252.
tion of glutamate receptors by kainic acid and Hespel, P., Eijnde, B.O., Derave, W., and Richter, E.A.
domoic acid. Nat. Toxins 6, 153158. (2001). Creatine supplementation:Exploring the
Hannuniemi, R., and Oja, S.S. (1981). Uptake of leu- role of the creatine kinase/phosphocreatine sys-
cine, lysine, aspartic acid, and glycine into iso- tem in human muscle. Can. J.Applied Physiol. 26
lated neurons and astrocytes. Neurochem. Res. Suppl, S79102.
6, 873884. Hoffman, D.R., Boettcher, J.A., and Diersen-Schade,
Haorah, J., Floreani, N.A., Knipe, B., and Persidsky, D.A. (2009). Toward optimizing vision and
Y. (2011). Stabilization of superoxide dismutase cognition in term infants by dietary docosa-
by acetyl-L-carnitine in human brain endothe- hexaenoic and arachidonic acid supplementa-
lium during alcohol exposure: Novel protective tion: A review of randomized controlled trials.
approach. Free Radic. Biol. Med. 51, 16011609. Prostaglandins Leukot. Essent. Fatty Acids 81,
Hardeland, R. (2012). Neurobiology, pathophysi- 151158.
ology, and treatment of melatonin deficiency Hoffman, J., Ratamess, N., Kang, J., Mangine, G.,
and dysfunction. ScientificWorldJournal 2012, Faigenbaum, A., and Stout, J. (2006). Effect of
640389. creatine and -alanine supplementation on per-
Harper, A.E., Miller, R.H., and Block, K.P. (1984). formance and endocrine responses in strength/
Branched-chain amino acid metabolism. Annu. power athletes. Int. J.Sport Nutr. Exerc. Metab.
Rev. Nutr. 4, 409454. 16, 430446.
Harris, R.A., Joshi, M., Jeoung, N.H., and Obayashi, M. Holness, M.J., and Sugden, M.C. (2003). Regulation
(2005). Overview of the molecular and biochem- of pyruvate dehydrogenase complex activity by
ical basis of branched-chain amino acid catabo- reversible phosphorylation. Biochem. Soc. Trans.
lism. J.Nutr. 135, 1527S-1530S. 31, 11431151.
Harris, R.C., Soderlund, K., and Hultman, E. (1992). Holtzman, D., Togliatti, A., Khait, I., and Jensen, F.
Elevation of creatine in resting and exercised (1998). Creatine increases survival and
Dietary Manipulations 291

suppresses seizures in the hypoxic immature rat. Kaido, M., Fujimura, H., Ono, A., Toyooka, K.,
Pediatr. Res. 44, 410414. Yoshikawa, H., Nishimura, T., Ozaki, K., Narama,
Hsiao, E.Y. (2014). Gastrointestinal issues in autism I., and Kuwajima, M. (1997). Mitochondrial
spectrum disorder. Harv. Rev. Psychiatry 22, abnormalities in a murine model of primary car-
104111. nitine deficiency:Systemic pathology and trial of
Hsiao, E.Y., McBride, S.W., Hsien, S., Sharon, G., replacement therapy. Eur. Neurol. 38, 302309.
Hyde, E.R., McCue, T., Codelli, J.A., Chow, J., Kanamori, K., Ross, B.D., and Kondrat, R.W.
Reisman, S.E., Petrosino, J.F., et al. (2013). (1998). Rate of glutamate synthesis from leu-
Microbiota modulate behavioral and physiologi- cine in rat brain measured in vivo by 15N NMR.
cal abnormalities associated with neurodevelop- J.Neurochem. 70, 13041315.
mental disorders. Cell 155, 14511463. Kang, D.W., Park, J.G., Ilhan, Z.E., Wallstrom, G.,
Huang, J.Y., Hong, Y.T., and Chuang, J.I. Labaer, J., Adams, J.B., and Krajmalnik-Brown,
(2009). Fibroblast growth factor 9 prevents R. (2013). Reduced incidence of Prevotella and
MPP+-induced death of dopaminergic neurons other fermenters in intestinal microflora of autis-
and is involved in melatonin neuroprotection in tic children. PLoS One 8, e68322.
vivo and in vitro. J.Neurochem. 109, 14001412. Kang, H.C., Chung, D.E., Kim, D.W., and Kim, H.D.
Hudson, S., and Tabet, N. (2003). Acetyl-L-carnitine (2004). Early- and late-onset complications of the
for dementia. Cochrane Database Syst. Rev. 2, ketogenic diet for intractable epilepsy. Epilepsia
CD003158. 45, 11161123.
Hutson, S.M., Berkich, D.A., Drown, P., Xu, B., Kang, T.C., Kim, D.S., Kwak, S.E., Kim, J.E., Won,
Aschner, M., and LaNoue, K.F. (1998). Role of M.H., Kim, D.W., Choi, S.Y., and Kwon, O.S.
branched chain aminotransferase isoenzymes (2006). Epileptogenic roles of astroglial death
and gabapentin in neurotransmitter metabo- and regeneration in the dentate gyrus of experi-
lism. J.Neurochem. 71, 863874. mental temporal lobe epilepsy. Glia 54, 258271.
Hutson, S.M., Lieth, E., and LaNoue, K.F. (2001). Kang, V., Wagner, G.C., and Ming, X. (2014).
Function of leucine in excitatory neurotransmit- Gastrointestinal dysfunction in children with
ter metabolism in the central nervous system. autism spectrum disorders. Autism Res. 7,
J.Nutr. 131, 846S-850S. 501506.
Innis, S.M. (2008). Dietary omega 3 fatty acids and Karaca, M., Frigerio, F., and Maechler, P. (2011).
the developing brain. Brain Res. 1237,3543. From pancreatic islets to central nervous system,
Iqbal, S., Nazir, N., Gillani, Q., Akbar, A., and Iqbal, F. the importance of glutamate dehydrogenase for
(2013). Effect of creatine monohydrate supple- the control of energy homeostasis. Neurochem.
mentation on various hematological and serum Int. 59, 510517.
biochemical parameters of male albino mice fol- Kathirvel, E., Morgan, K., French, S.W., and Morgan,
lowing neonatal hypoxia-ischemia encephalopa- T.R. (2013). Acetyl-L-carnitine and lipoic acid
thy. ScientificWorldJournal 2013, 286075. improve mitochondrial abnormalities and serum
Islam, M.M., Nautiyal, M., Wynn, R.M., Mobley, levels of liver enzymes in a mouse model of non-
J.A., Chuang, D.T., and Hutson, S.M. (2010). alcoholic fatty liver disease. Nutr. Res. (New
Branched-chain amino acid metabolon: York) 33, 932941.
Interaction of glutamate dehydrogenase with the Kimura, S., and Amemiya, F. (1990). Brain and liver
mitochondrial branched-chain aminotransfer- pathology in a patient with carnitine deficiency.
ase (BCATm). J.Biol. Chem. 285, 265276. Brain Dev. 12, 436439.
Issa, A.M., Mojica, W.A., Morton, S.C., Traina, S., Konturek, P.C., Brzozowski, T., and Konturek, S.J.
Newberry, S.J., Hilton, L.G., Garland, R.H., and (2011). Stress and the gut:Pathophysiology, clini-
Maclean, C.H. (2006). The efficacy of omega-3 cal consequences, diagnostic approach and treat-
fatty acids on cognitive function in aging and ment options. J.Physiol. Pharmacol. 62, 591599.
dementia:Asystematic review. Dement. Geriatr. Kossoff, E.H., Krauss, G.L., McGrogan, J.R., and
Cogn. Disord. 21,8896. Freeman, J.M. (2003). Efficacy of the Atkins diet
Iwasa, J., Shimizu, M., Shiraki, M., Shirakami, Y., as therapy for intractable epilepsy. Neurology 61,
Sakai, H., Terakura, Y., Takai, K., Tsurumi, H., 17891791.
Tanaka, T., and Moriwaki, H. (2010). Dietary Kuzniecki, R.I., and Jackson, G.D. (1995). Magnetic
supplementation with branched-chain amino resonance spectroscopy in epilepsy. In:Magnetic
acids suppresses diethylnitrosamine-induced resonance in epilepsy, R.I. Kuzniecki, and G.D.
liver tumorigenesis in obese and diabetic C57BL/ Jackson, eds. (New York: Raven Press), pp.
KsJ-db/db mice. Cancer Sci. 101, 460467. 289214.
292 Part III:Homeostatic Manipulators

La Guardia, P.G., Alberici, L.C., Ravagnani, F.G., Magoulas, P.L., and El-Hattab, A.W. (2012). Systemic
Catharino, R.R., and Vercesi, A.E. (2013). primary carnitine deficiency: An overview of
Protection of rat skeletal muscle fibers by either clinical manifestations, diagnosis, and manage-
L-carnitine or coenzyme Q10 against statins tox- ment. Orphanet J.Rare Dis.7,68.
icity mediated by mitochondrial reactive oxygen Malaguarnera, M. (2013). Acetyl-L-carnitine in
generation. Front. Physiol. 4,103. hepatic encephalopathy. Metab. Brain Dis. 28,
Lage, S., Andrade, F., Prieto, J.A., Asla, I., Rodrguez, A., 193199.
Ruiz, N., Echeverria, J., Luz Couce, M., Malaguarnera, M., Cammalleri, L., Gargante, M.P.,
Sanjurjo, P., and Aldmiz-Echevarra, L. (2013). Vacante, M., Colonna, V., and Motta, M. (2007).
Arginine-guanidinoacetate-creatine pathway in L-Carnitine treatment reduces severity of physical
preterm newborns:Creatine biosynthesis in new- and mental fatigue and increases cognitive func-
borns. J.Pediatr. Endocrinol. Metab. 26,5360. tions in centenarians:Arandomized and controlled
Layman, D.K. (2003). The role of leucine in weight clinical trial. Am. J.Clin. Nutr. 86, 17381744.
loss diets and glucose homeostasis. J.Nutr. 133, Malaguarnera, M., Gargante, M.P., Cristaldi, E.,
261S-267S. Colonna, V., Messano, M., Koverech, A., Neri, S.,
Legido, A., Jethva, R., and Goldenthal, M.J. (2013). Vacante, M., Cammalleri, L., and Motta, M.
Mitochondrial dysfunction in autism. Semin. (2008). Acetyl L-carnitine (ALC) treatment in
Pediatr. Neurol. 20, 163175. elderly patients with fatigue. Arch. Gerontol.
Levoye, A., Jockers, R., Ayoub, M.A., Delagrange, P., Geriatr. 46, 181190.
Savaskan, E., and Guillaume, J.L. (2006). Are G Malow, B., Adkins, K.W., McGrew, S.G., Wang, L.,
protein-coupled receptor heterodimers of physi- Goldman, S.E., Fawkes, D., and Burnette, C.
ological relevance? Focus on melatonin recep- (2012). Melatonin for sleep in children with
tors. Chronobiol. Int. 23, 419426. autism:Acontrolled trial examining dose, toler-
Li, B., Lloyd, M.L., Gudjonsson, H., Shug, A.L., and ability, and outcomes. J.Autism Dev. Disords 42,
Olsen, W.A. (1992). The effect of enteral carnitine 17291737.
administration in humans. Am. J.Clin. Nutr. 55, Malthankar-Phatak, G.H., de Lanerolle, N., Eid, T.,
838845. Spencer, D.D., Behar, K.L., Spencer, S.S., Kim, J.H.,
Li, C., Najafi, H., Daikhin, Y., Nissim, I.B., and Lai, J.C. (2006). Differential glutamate
Collins, H.W., Yudkoff, M., Matschinsky, F.M., dehydrogenase (GDH) activity profile in
and Stanley, C.A. (2003). Regulation of patients with temporal lobe epilepsy. Epilepsia
leucine-stimulated insulin secretion and glu- 47, 12921299.
tamine metabolism in isolated rat islets. J.Biol. Marano, G., Traversi, G., Nannarelli, C., Mazza, S.,
Chem. 278, 28532858. and Mazza, M. (2013). Omega-3 fatty acids and
Lifshitz, J., Friberg, H., Neumar, R.W., Raghupathi, R., schizophrenia: Evidences and recommenda-
Welsh, F.A., Janmey, P., Saatman, K.E., Wieloch, T., tions. Clin. Ter. 164, e529e537.
Grady, M.S., and McIntosh, T.K. (2003). Mattson, M.P. (2003). Excitotoxic and excitopro-
Structural and functional damage sustained by tective mechanisms: Abundant targets for the
mitochondria after traumatic brain injury in the prevention and treatment of neurodegenerative
rat:Evidence for differentially sensitive popula- disorders. Neuromol. Med. 3,6594.
tions in the cortex and hippocampus. J. Cereb. Mattson, M.P. (2008). Glutamate and neurotrophic
Blood Flow Metab. 23, 219231. factors in neuronal plasticity and disease. Ann.
Ling, B., Aziz, C., and Alcorn, J. (2012). Systematic N.Y. Acad. Sci. 1144, 97112.
evaluation of key L-carnitine homeostasis mech- McElhanon, B.O., McCracken, C., Karpen, S., and
anisms during postnatal development in rat. Sharp, W.G. (2014). Gastrointestinal symptoms
Nutr. Metab.9,66. in autism spectrum disorder: A meta-analysis.
Luthringer, R., Muzet, M., Zisapel, N., and Staner, L. Pediatrics 133, 872883.
(2009). The effect of prolonged-release melato- Meldrum, B.S. (2000). Glutamate as a neurotrans-
nin on sleep measures and psychomotor per- mitter in the brain: Review of physiology and
formance in elderly patients with insomnia. Int. pathology. J.Nutr. 130, 1007S1015S.
Clin. Psychopharmacol. 24, 239249. Mendes-Ribeiro, J.A., Soares, R., Simes-Ribeiro, F.,
Lyseng-Williamson, K.A. (2012). Melatonin prolonged and Guimares, M.L. (1998). Reduction in tem-
release:In the treatment of insomnia in patients poral N-acetylaspartate and creatine (or cho-
aged >/=55years. Drugs Aging 29, 911923. line) ratio in temporal lobe epilepsy: Does this
Maebashi, M., Imamura, A., and Yoshinaga, K. 1
H-magnetic resonance spectroscopy finding
(1982). Effect of aging on lipid and carnitine mean poor seizure control? J. Neurol. Neurosurg.
metabolism. Tohoku J.Exp. Med. 138, 231236. Psychiatry 65, 518522.
Dietary Manipulations 293

Mishima, K., Tozawa, T., Satoh, K., Matsumoto, Y., (2002). The blood-brain barrier creatine trans-
Hishikawa, Y., and Okawa, M. (1999). Melatonin porter is a major pathway for supplying creatine
secretion rhythm disorders in patients with senile to the brain. J. Cereb. Blood Flow Metab. 22,
dementia of Alzheimers type with disturbed 13271335.
sleep-waking. Biol. Psychiatry 45, 417421. Okumura, A., Morita, M., Ikeno, M., Abe, S., and
Monterrosa-Castro, A., Marrugo-Flrez, M., Shimizu, T. (2011). Acute encephalopathy in a
Romero-Prez, I., Chedraui, P., Fernndez-Alonso, child with secondary carnitine deficiency due to
A.M., and Prez-Lpez, F.R. (2013). Prevalence of pivalate-conjugated antibiotics. Pediatr. Infect.
insomnia and related factors in a large mid-aged Dis. J. 30,92.
female Colombian sample. Maturitas 74, 346351. Oldendorf, W.H. (1971). Brain uptake of radiolabeled
Montgomery, S.A., Thal, L.J., and Amrein, R. (2003). amino acids, amines, and hexoses after arterial
Meta-analysis of double blind randomized con- injection. Am. J.Physiol. 221, 16291639.
trolled clinical trials of acetyl-L-carnitine ver- Ortiz, G.G., Bentez-King, G.A., Rosales-Corral, S.A.,
sus placebo in the treatment of mild cognitive Pacheco-Moiss, F.P., and Velzquez-Brizuela, I.E.
impairment and mild Alzheimers disease. Int. (2008). Cellular and biochemical actions of
Clin. Psychopharmacol. 18,6171. melatonin which protect against free radi-
Montiel-Castro, A.J., Gonzlez-Cervantes, R.M., cals:Role in neurodegenerative disorders. Curr.
Bravo-Ruiseco, G., and Pacheco-Lpez, G. (2013). Neuropharmacol. 6, 203214.
The microbiota-gut-brain axis:Neurobehavioral Owen, K.Q., Smith, J.W., II, Nelssen, J.L., Goodband,
correlates, health and sociality. Front. Integr. R.D., Tokach, M.D., Friesen, K.G., and Blum, S.A.
Neurosci.7,70. (1994). The effect of L-carnitine on growth
Morand, R., Todesco, L., Donzelli, M., Fischer-Barnicol, performance and carcass characteristics of
D., Mullen, P.J., and Krhenbhl, S. (2012). Effect growing-finishing pigs. Swine Day, 161164.
of short- and long-term treatment with valproate Pandi-Perumal, S.R., BaHammam, A.S., Brown,
on carnitine homeostasis in humans. Ther. Drug G.M., Spence, D.W., Bharti, V.K., Kaur, C.,
Monit. 34, 406414. Hardeland, R., and Cardinali, D.P. (2013).
Mosnik, D.M., Spring, B., Rogers, K., and Baruah, S. Melatonin antioxidative defense: Therapeutical
(1997). Tardive dyskinesia exacerbated after inges- implications for aging and neurodegenerative
tion of phenylalanine by schizophrenic patients. processes. Neurotox. Res. 23, 267300.
Neuropsychopharmacology 16, 136146. Pardridge, W.M. (1983). Brain metabolism: A per-
Naecz, K.A., Miecz, D., Berezowski, V., and spective from the blood-brain barrier. Physiol.
Cecchelli, R. (2004). Carnitine: Transport Rev. 63, 14811535.
and physiological functions in the brain. Mol. Pardridge, W.M., and Choi, T.B. (1986). Neutral
Aspects Med. 25, 551567. amino acid transport at the human blood-brain
Nash, S.R., Giros, B., Kingsmore, S.F., Rochelle, J.M., barrier. Fed. Proc. 45, 20732078.
Suter, S.T., Gregor, P., Seldin, M.F., and Caron, Patel, S.P., Sullivan, P.G., Lyttle, T.S., and Rabchevsky,
M.G. (1994). Cloning, pharmacological char- A.G. (2010). Acetyl-L-carnitine ameliorates
acterization, and genomic localization of the mitochondrial dysfunction following contusion
human creatine transporter. Receptors Channels spinal cord injury. J.Neurochem. 114, 291301.
2, 165174. Patti, M.E., Brambilla, E., Luzi, L., Landaker, E.J.,
Neish, A.S. (2009). Microbes in gastrointesti- and Kahn, C.R. (1998). Bidirectional modulation
nal health and disease. Gastroenterology of insulin action by amino acids. J.Clin. Invest.
136,6580. 101, 15191529.
Neufeld, M.Y., Treves, T.A., Chistik, V., and Korczyn, Pekala, J., Patkowska-Sokoa, B., Bodkowski, R.,
A.D. (1997). Sequential serum creatine kinase Jamroz, D., Nowakowski, P., Lochyski, S., and
determination differentiates vaso-vagal syncope Librowski, T. (2011). L-carnitine-metabolic func-
from generalized tonic-clonic seizures. Acta tions and meaning in humans life. Curr. Drug
Neurol. Scand. 95, 137139. Metab. 12, 667678.
Noland, R.C., Koves, T.R., Seiler, S.E., Lum, H., Lust, Perrott, J., Murphy, N.G., and Zed, P.J. (2010).
R.M., Ilkayeva, O., Stevens, R.D., Hegardt, F.G., L-carnitine for acute valproic acid over-
and Muoio, D.M. (2009). Carnitine insufficiency dose: A systematic review of published cases.
caused by aging and overnutrition compromises Ann. Pharmacother. 44, 12871293.
mitochondrial performance and metabolic con- Petramfar, P., Yaghoobi, E., Nemati, R., and
trol. J.Biol. Chem. 284, 2284022852. Asadi-Pooya, A.A. (2009). Serum creatine phos-
Ohtsuki, S., Tachikawa, M., Takanaga, H., Shimizu, H., phokinase is helpful in distinguishing gener-
Watanabe, M., Hosoya, K., and Terasaki, T. alized tonic-clonic seizures from psychogenic
294 Part III:Homeostatic Manipulators

nonepileptic seizures and vasovagal syncope. Rebouche, C.J. (1992). Carnitine function and
Epilepsy Behav. 15, 330332. requirements during the life cycle. FASEB J. 6,
Pettegrew, J.W., Levine, J., and McClure, R.J. (2000). 33793386.
Acetyl-L-carnitine physical-chemical, meta- Rebouche, C.J. (2004). Kinetics, pharmacokinet-
bolic, and therapeutic properties: Relevance for ics, and regulation of L-carnitine and acetyl-L-
its mode of action in Alzheimers disease and carnitine metabolism. Ann. N.Y. Acad. Sci.
geriatric depression. Mol. Psychiatry 5, 616632. 1033,3041.
Pfeifer, H.H., and Thiele, E.A. (2005). Rebouche, C.J., and Seim, H. (1998). Carnitine
Low-glycemic-index treatment: A liberalized metabolism and its regulation in microorgan-
ketogenic diet for treatment of intractable epi- isms and mammals. Annu. Rev. Nutr. 18,3961.
lepsy. Neurology 65, 18101812. Reiter, R.J., Manchester, L.C., and Tan, D.X. (2010).
Pillich, R.T., Scarsella, G., and Risuleo, G. (2005). Neurotoxins: Free radical mechanisms and
Reduction of apoptosis through the mitochon- melatonin protection. Curr. Neuropharmacol. 8,
drial pathway by the administration of acetyl-L- 194210.
carnitine to mouse fibroblasts in culture. Exp. Resta, S.C. (2009). Effects of probiotics and com-
Cell Res. 306,18. mensals on intestinal epithelial physiol-
Piscopo, P., Crestini, A., Adduci, A., Ferrante, ogy: Implications for nutrient handling.
A., Massari, M., Popoli, P., Vanacore, N., and J.Physiol. 587, 41694174.
Confaloni, A. (2011). Altered oxidative stress Reuter, S.E., and Evans, A.M. (2012). Carnitine and
profile in the cortex of mice fed an enriched acylcarnitines:Pharmacokinetic, pharmacologi-
branched-chain amino acids diet: Possible link cal and clinical aspects. Clin. Pharmacokinet. 51,
with amyotrophic lateral sclerosis? J. Neurosci. 553572.
Res. 89, 12761283. Richardson, M.A., Chao, H.M., Read, L.L., Clelland,
Plaitakis, A. (1989). Abnormal glutamatergic mecha- J.D., and Suckow, R.F. (2006). Investigation of the
nisms and branched-chain aminoacids in amyo- phenylalanine hydroxylase gene and tardive dys-
trophic lateral sclerosis. Lancet 1,157. kinesia. Am. J. Med. Genet. B Neuropsychiatr.
Plaitakis, A., Smith, J., Mandeli, J., and Yahr, M.D. Genet. 141B, 195197.
(1988). Pilot trial of branched-chain aminoac- Richardson, M.A., Reilly, M.A., Read, L.L., Flynn,
ids in amyotrophic lateral sclerosis. Lancet 1, C.J., Suckow, R.F., Maher, T.J., and Sziraki, I.
10151018. (1999). Phenylalanine kinetics are associated
Polimeni, G., Esposito, E., Bevelacqua, V., Guarneri, C., with tardive dyskinesia in men but not in women.
and Cuzzocrea, S. (2014). Role of melatonin sup- Psychopharmacology 143, 347357.
plementation in neurodegenerative disorders. Richardson, M.A., Small, A.M., Read, L.L., Chao,
Front. Biosci. (Landmark) 19, 429446. H.M., and Clelland, J.D. (2004). Branched chain
Pons, R., Carrozzo, R., Tein, I., Walker, W.F., amino acid treatment of tardive dyskinesia in
Addonizio, L.J., Rhead, W., Miranda, A.F., children and adolescents. J. Clin. Psychiatry
Dimauro, S., and De Vivo, D.C. (1997). Deficient 65,9296.
muscle carnitine transport in primary carnitine Rios, E.R., Venncio, E.T., Rocha, N.F.,
deficiency. Pediatr. Res. 42, 583587. Woods, D.J., Vasconcelos, S., Macedo, D.,
Pons, R., and De Vivo, D.C. (1995). Primary and sec- Sousa, F.C., and Fonteles, M.M. (2010).
ondary carnitine deficiency syndromes. J.Child Melatonin:Pharmacological aspects and clinical
Neurol. 10 Suppl 2,S824. trends. Int. J.Neurosci. 120, 583590.
Rambo, L.M., Ribeiro, L.R., Oliveira, M.S., Furian, A.F., Rondanelli, M., Giacosa, A., Opizzi, A., Pelucchi, C.,
Lima, F.D., Souza, M.A., Silva, L.F., Retamoso, La Vecchia, C., Montorfano, G., Negroni, M.,
L.T., Corte, C.L., Puntel, G.O., et al. (2009). Berra, B., Politi, P., and Rizzo, A.M. (2010).
Additive anticonvulsant effects of creatine Effect of omega-3 fatty acids supplementation on
supplementation and physical exercise against depressive symptoms and on health-related qual-
pentylenetetrazol-induced seizures. Neurochem. ity of life in the treatment of elderly women with
Int. 55, 333340. depression: A double-blind, placebo-controlled,
Ramsay, R.R. (2000). The carnitine acyltransfer- randomized clinical trial. J. Am. Coll. Nutr.
ases: Modulators of acyl-CoA-dependent reac- 29,5564.
tions. Biochem. Soc. Trans. 28, 182186. Rondanelli, M., Giacosa, A., Opizzi, A., Pelucchi,
Ramsay, R.R., and Arduini, A. (1993). The carnitine C., La Vecchia, C., Montorfano, G., Negroni,
acyltransferases and their role in modulating M., Berra, B., Politi, P., and Rizzo, A.M. (2011).
acyl-CoA pools. Arch. Biochem. Biophys. 302, Long chain omega 3 polyunsaturated fatty
307314. acids supplementation in the treatment of
Dietary Manipulations 295

elderly depression: Effects on depressive symp- fatigue with creatine administration. A pilot
toms, on phospholipids fatty acids profile and study. Acta Paediatr. 97,3134.
on health-related quality of life. J. Nutr. Health Salomons, G.S., van Dooren, S.J., Verhoeven, N.M.,
Aging 15,3744. Cecil, K.M., Ball, W.S., Degrauw, T.J., and
Rook, G.A., Raison, C.L., and Lowry, C.A. (2014). Jakobs, C. (2001). X-linked creatine-transporter
Microbiota, immunoregulatory old friends and gene (SLC6A8) defect:Anew creatine-deficiency
psychiatric disorders. Adv. Exp. Med. Biol. 817, syndrome. Am. J.Hum. Genet. 68, 14971500.
319356. Samsam, M., Ahangari, R., and Naser, S.A. (2014).
Rosales-Corral, S.A., Acua-Castroviejo, D., Pathophysiology of autism spectrum disor-
Coto-Montes, A., Boga, J.A., Manchester, L.C., ders:Revisiting gastrointestinal involvement and
Fuentes-Broto, L., Korkmaz, A., Ma, S., Tan, immune imbalance. World J.Gastroenterol. 20,
D.X., and Reiter, R.J. (2012). Alzheimers dis- 99429951.
ease: Pathological mechanisms and the benefi- SanGiovanni, J.P., Parra-Cabrera, S., Colditz,
cial role of melatonin. J.Pineal Res. 52, 167202. G.A., Berkey, C.S., and Dwyer, J.T. (2000).
Rospond, B., and Chopicka, J. (2013). The biologi- Meta-analysis of dietary essential fatty acids and
cal function of L-carnitine and its content in the long-chain polyunsaturated fatty acids as they
particular food examples. Przegl. Lek. 70,8591. relate to visual resolution acuity in healthy pre-
Ross, B.M., Seguin, J., and Sieswerda, L.E. (2007). term infants. Pediatrics 105, 12921298.
Omega-3 fatty acids as treatments for mental Savitha, S., Sivarajan, K., Haripriya, D., Kokilavani, V.,
illness: Which disorder and which fatty acid? and Panneerselvam, C. (2005). Efficacy of levo
Lipids Health Dis.6,21. carnitine and alpha lipoic acid in ameliorating
Rossignol, D.A., and Frye, R.E. (2011). Melatonin in the decline in mitochondrial enzymes during
autism spectrum disorders:Asystematic review aging. Clin. Nutr. (Edinburgh) 24, 794800.
and meta-analysis. Dev. Med. Child Neurol. 53, Scarna, A., Gijsman, H.J., McTavish, S.F., Harmer, C.J.,
783792. Cowen, P.J., and Goodwin, G.M. (2003). Effects
Rossignol, D.A., and Frye, R.E. (2012). Substantial of a branched-chain amino acid drink in mania.
problems with measuring brain mitochondrial Br. J.Psychiatry 182, 210213.
dysfunction in autism spectrum disorder using Scarna, A., McTavish, S.F., Cowen, P.J., Goodwin,
magnetic resonance spectroscopy. J.Autism Dev. G.M., and Rogers, R.D. (2005). The effects of
Disord. 42, 640642. a branched chain amino acid mixture sup-
Royes, L.F., Fighera, M.R., Furian, A.F., Oliveira, plemented with tryptophan on biochemi-
M.S., Myskiw Jde, C., Fiorenza, N.G., Petry, J.C., cal indices of neurotransmitter function and
Coelho, R.C., and Mello, C.F. (2006). Effectiveness decision-making. Psychopharmacology 179,
of creatine monohydrate on seizures and oxi- 761768.
dative damage induced by methylmalonate. Schultz, S.K., Ellingrod, V., Fleming, F.W., and
Pharmacol. Biochem. Behav. 83, 136144. Andreasen, N.C. (2001). The association
Ruderman, N.B. (1975). Muscle amino acid metabo- between risk factors for tardive dyskinesia and
lism and gluconeogenesis. Annu. Rev. Med. 26, phenylalanine-induced abnormal movements
245258. in schizophrenia. Hum. Psychopharmacol. 16,
Sakai, R., Cohen, D.M., Henry, J.F., Burrin, D.G., and 273277.
Reeds, P.J. (2004). Leucine-nitrogen metabolism Scimemi, A., Schorge, S., Kullmann, D.M., and
in the brain of conscious rats:Its role as a nitrogen Walker, M.C. (2006). Epileptogenesis is associ-
carrier in glutamate synthesis in glial and neuro- ated with enhanced glutamatergic transmis-
nal metabolic compartments. J.Neurochem. 88, sion in the perforant path. J. Neurophysiol. 95,
612622. 12131220.
Sakellaris, G., Kotsiou, M., Tamiolaki, M., Kalostos, G., Sevy, S., Hassoun, Y., Bechara, A., Yechiam, E.,
Tsapaki, E., Spanaki, M., Spilioti, M., Napolitano, B., Burdick, K., Delman, H.,
Charissis, G., and Evangeliou, A. (2006). and Malhotra, A. (2006). Emotion-based
Prevention of complications related to traumatic decision-making in healthy subjects:Short-term
brain injury in children and adolescents with effects of reducing dopamine levels.
creatine administration:An open label random- Psychopharmacology 188, 228235.
ized pilot study. J.Trauma 61, 322329. Shepherd, J.D., and Huganir, R.L. (2007). The cell
Sakellaris, G., Nasis, G., Kotsiou, M., Tamiolaki, M., biology of synaptic plasticity: AMPA recep-
Charissis, G., and Evangeliou, A. (2008). tor trafficking. Annu. Rev. Cell Dev. Biol. 23,
Prevention of traumatic headache, dizziness and 613643.
296 Part III:Homeostatic Manipulators

Simn Padilla, O.J., De Linares Del Ro, C., Godden, P., J. (1994). Creatine deficiency in the brain:Anew,
Granizo, E., Pinilla, M., and Rodrguez, C. treatable inborn error of metabolism. Pediatr.
(2009). Use of combined treatment with melato- Res. 36, 409413.
nin and clomethiazole in circadian rhythm sleep Stumpf, D.A., Parker, W.D., Jr., and Angelini, C.
disorder in the elderly with dementia. Rev. Esp. (1985). Carnitine deficiency, organic acidemias,
Geriatr. Gerontol. 44, 233234. and Reyes syndrome. Neurology 35, 10411045.
Skeie, B., Petersen, A.J., Manner, T., Askanazi, J., Sullivan, P.G., Geiger, J.D., Mattson, M.P., and
Jellum, E., and Steen, P.A. (1992). Branched-chain Scheff, S.W. (2000). Dietary supplement creatine
amino acids increase the seizure threshold to protects against traumatic brain injury. Ann.
picrotoxin in rats. Pharmacol. Biochem. Behav. Neurol. 48, 723729.
43, 669671. Sweatt, A.J., Garcia-Espinosa, M.A., Wallin, R.,
Skeie, B., Petersen, A.J., Manner, T., Askanazi, J., and Hutson, S.M. (2004). Branched-chain
and Steen, P.A. (1994). Effects of valine, leucine, amino acids and neurotransmitter metabo-
isoleucine, and a balanced amino acid solution lism: Expression of cytosolic branched-chain
on the seizure threshold to picrotoxin in rats. aminotransferase (BCATc) in the cerebellum and
Pharmacol. Biochem. Behav. 48, 101103. hippocampus. J.Comp. Neurol. 477, 360370.
Sliwinski, T., Rozej, W., Morawiec-Bajda, A., Szajewska, H. (2013). Microbiota modulation: Can
Morawiec, Z., Reiter, R., and Blasiak, J. (2007). probiotics prevent/treat disease in pediatrics?
Protective action of melatonin against oxi- Nestle Nutr. Inst. Workshop Ser. 77, 99110.
dative DNA damage: Chemical inactivation Tabrizi, S.J., Blamire, A.M., Manners, D.N.,
versus base-excision repair. Mutat. Res. 634, Rajagopalan, B., Styles, P., Schapira, A.H., and
220227. Warner, T.T. (2005). High-dose creatine therapy
Smith, C.J., Emge, J.R., Berzins, K., Lung, L., for Huntington disease: A 2-year clinical and
Khamishon, R., Shah, P., Rodrigues, D.M., Sousa, MRS study. Neurology 64, 16551656.
A.J., Reardon, C., Sherman, P.M., et al. (2014). Tandan, R., Bromberg, M.B., Forshew, D., Fries, T.J.,
Probiotics normalize the gut-brain-microbiota Badger, G.J., Carpenter, J., Krusinski, P.B., Betts,
axis in immunodeficient mice. Am. J. Physiol. E.F., Arciero, K., and Nau, K. (1996). A controlled
Gastrointest. Liver Physiol. 307, G793802. trial of amino acid therapy in amyotrophic lateral
Smith, Q.R., Momma, S., Aoyagi, M., and Rapoport, S.I. sclerosis: I. Clinical, functional, and maximum
(1987). Kinetics of neutral amino acid transport isometric torque. Neurology 47, 12201226.
across the blood-brain barrier. J.Neurochem. 49, Tepas, J.J., 3rd, DiScala, C., Ramenofsky, M.L., and
16511658. Barlow, B. (1990). Mortality and head injury:The
Snow, R.J., and Murphy, R.M. (2001). Creatine and pediatric perspective. J.Pediatr. Surg. 25,9295.
the creatine transporter: A review. Mol. Cell. Tomova, A., Husarova, V., Lakatosova, S., Bakos, J.,
Biochem. 224, 169181. Vlkova, B., Babinska, K., and Ostatnikova, D.
Sonik, P., Hilty, D.M., Rossaro, L., and Bourgeois, J.A. (2015). Gastrointestinal microbiota in children
(2011). Carnitine supplementation for valproate- with autism in Slovakia. Physiol. Behav. 138,
related hyperammonemia to maintain therapeu- 179187.
tic valproate level. J. Clin. Psychopharmacol. 31, Tordjman, S., Anderson, G.M., Pichard, N., Charbuy,
680682. H., and Touitou, Y. (2005). Nocturnal excretion
Spanaki, C., Zaganas, I., Kleopa, K.A., and Plaitakis, A. of 6-sulphatoxymelatonin in children and ado-
(2010). Human GLUD2 glutamate dehydroge- lescents with autistic disorder. Biol. Psychiatry
nase is expressed in neural and testicular sup- 57, 134138.
porting cells. J.Biol. Chem. 285, 1674816756. Touitou, Y., Fvre, M., Bogdan, A., Reinberg, A.,
Srinivasan, V., Cardinali, D.P., Srinivasan, U.S., De Prins, J., Beck, H., and Touitou, C. (1984).
Kaur, C., Brown, G.M., Spence, D.W., Hardeland, Patterns of plasma melatonin with ageing and
R., and Pandi-Perumal, S.R. (2011). Therapeutic mental condition: Stability of nyctohemeral
potential of melatonin and its analogs in rhythms and differences in seasonal variations.
Parkinsons disease: Focus on sleep and neuro- Acta Endocrinol. 106, 145151.
protection. Ther. Adv. Neurol. Disord. 4, 297317. Traina, G., Bernardi, R., Cataldo, E., Macchi, M.,
Stockler, S., Hanefeld, F., and Frahm, J. (1996). Durante, M., and Brunelli, M. (2008). In the rat
Creatine replacement therapy in guanidinoace- brain acetyl-L-carnitine treatment modulates the
tate methyltransferase deficiency, a novel inborn expression of genes involved in neuronal ceroid
error of metabolism. Lancet 348, 789790. lipofuscinosis. Mol. Neurobiol. 38, 146152.
Stockler, S., Holzbach, U., Hanefeld, F., Marquardt, I., Triggs, W.J., Gilmore, R.L., Millington, D.S.,
Helms, G., Requart, M., Hanicke, W., and Frahm, Cibula, J., Bunch, T.S., and Harman, E. (1997).
Dietary Manipulations 297

Valproate-associated carnitine deficiency and Weishaupt, J.H., Bartels, C., Plking, E., Dietrich, J.,
malignant cerebral edema in the absence of Rohde, G., Poeggeler, B., Mertens, N., Sperling,
hepatic failure. Int. J.Clin. Pharmacol. Ther. 35, S., Bohn, M., Hther, G., etal. (2006). Reduced
353356. oxidative damage in ALS by high-dose enteral
Tylianakis, J.M. (2009). Ecology. Warming up food melatonin treatment. J.Pineal Res. 41, 313323.
webs. Science (NewYork) 323, 13001301. Wong, R.K., Chuang, S.C., and Bianchi, R. (2002).
Usherwood, P.N., Machili, P., and Leaf, G. (1968). Metabotropic glutamate receptors and epilepto-
L-Glutamate at insect excitatory nerve-muscle genesis. Epilepsy Curr. 2,8185.
synapses. Nature 219, 11691172. Xu, G., Kwon, G., Marshall, C.A., Lin, T.A.,
Vaz, F.M., and Wanders, R.J. (2002). Carnitine bio- Lawrence, J.C., Jr., and McDaniel, M.L. (1998).
synthesis in mammals. Biochem. J. 361, 417429. Branched-chain amino acids are essential in the
Virmani, A., and Binienda, Z. (2004). Role of carni- regulation of PHAS-I and p70 S6 kinase by pan-
tine esters in brain neuropathology. Mol. Aspects creatic b-cells. Apossible role in protein transla-
Med. 25, 533549. tion and mitogenic signaling. J.Biol. Chem. 273,
Virmani, M.A., Caso, V., Spadoni, A., Rossi, S., Russo, 2817828184.
F., and Gaetani, F. (2001). The action of acetyl-L- Yudkoff, M. (1997). Brain metabolism of
carnitine on the neurotoxicity evoked by amyloid branched-chain amino acids. Glia 21,9298.
fragments and peroxide on primary rat cortical Yudkoff, M., Daikhin, Y., Lin, Z.P., Nissim, I.,
neurones. Ann. N.Y. Acad. Sci. 939, 162178. Stern, J., Pleasure, D., and Nissim, I. (1994).
Vreken, P., van Lint, A.E., Bootsma, A.H., Overmars, Interrelationships of leucine and gluta-
H., Wanders, R.J., and van Gennip, A.H. (1999). mate metabolism in cultured astrocytes.
Rapid diagnosis of organic acidemias and J.Neurochem. 62, 11921202.
fatty-acid oxidation defects by quantitative elec- Yudkoff, M., Daikhin, Y., Nissim, I., Horyn, O.,
trospray tandem-MS acyl-carnitine analysis in Lazarow, A., Luhovyy, B., Wehrli, S., and
plasma. Adv. Exp. Med. Biol. 466, 327337. Nissim, I. (2005a). Response of brain amino
Wade, A.G., Crawford, G., Ford, I., McConnachie, A., acid metabolism to ketosis. Neurochem. Int.
Nir, T., Laudon, M., and Zisapel, N. (2011). 47, 119128.
Prolonged release melatonin in the treatment of Yudkoff, M., Daikhin, Y., Nissim, I., Horyn, O.,
primary insomnia:Evaluation of the age cut-off Luhovyy, B., Lazarow, A., and Nissim, I.
for short- and long-term response. Curr. Med. (2005b). Brain amino acid requirements and
Res. Opin. 27,8798. toxicity: The example of leucine. J. Nutr. 135,
Wade, A.G., Ford, I., Crawford, G., McConnachie, A., 1531S-1538S.
Nir, T., Laudon, M., and Zisapel, N. (2010). Yudkoff, M., Daikhin, Y., Nissim, I., Lazarow, A.,
Nightly treatment of primary insomnia with pro- and Nissim, I. (2004). Ketogenic diet, brain
longed release melatonin for 6 months: A ran- glutamate metabolism and seizure control.
domized placebo controlled trial on age and Prostaglandins Leukot. Essent. Fatty Acids 70,
endogenous melatonin as predictors of efficacy 277285.
and safety. BMC Med.8,51. Zanelli, S.A., Solenski, N.J., Rosenthal, R.E., and
Walker, J.B. (1979). Creatine: Biosynthesis, regula- Fiskum, G. (2005). Mechanisms of ischemic neu-
tion, and function. Adv. Enzymol. Relat. Areas roprotection by acetyl-L-carnitine. Ann. N.Y.
Mol. Biol. 50, 177242. Acad. Sci. 1053, 153161.
Wang, L., Christophersen, C.T., Sorich, M.J., Gerber, J.P., Zeng, L.H., Ouyang, Y., Gazit, V., Cirrito, J.R.,
Angley, M.T., and Conlon, M.A. (2013). Increased Jansen, L.A., Ess, K.C., Yamada, K.A., Wozniak,
abundance of Sutterella spp. and Ruminococcus D.F., Holtzman, D.M., Gutmann, D.H., et al.
torques in feces of children with autism spectrum (2007). Abnormal glutamate homeostasis and
disorder. Mol. Autism4,42. impaired synaptic plasticity and learning in a
Wang, X. (2009). The antiapoptotic activity of mouse model of tuberous sclerosis complex.
melatonin in neurodegenerative diseases. CNS Neurobiol. Dis. 28, 184196.
Neurosci. Ther. 15, 345357. Zhang, W., Li, P., Hu, X., Zhang, F., Chen, J., and
Weinberg, G.L., and Baughman, V. (2006). Gao, Y. (2011). Omega-3 polyunsaturated
Carnitine deficiency, mitochondrial metabo- fatty acids in the brain: Metabolism and neu-
lism, and abnormal response to anesthetics. roprotection. Front. Biosci. (Landmark) 16,
Anesthesiology 104,1343. 26532670.
17
Exercise
M ARKUSDWOR AK

INTRODUCTION NEUR AL CONTROL


Participation in regular physical activity has OFMOVEMENT
been associated with positive effects on physi- Exercise involves the contraction of muscle tis-
cal and mental performance. Physical exercise sue through an increase in spatial and temporal
affects the cardiovascular system, the pulmo- recruitment of motor units. The neural organiza-
nary system, and whole-body metabolism and is tion of motor control is organized hierarchically,
an important factor in the prevention of cardio- with smaller and simpler elements of movement
vascular diseases and metabolic diseases such integrated into more complex elements at higher
as type 2 diabetes and obesity. Other than the levels of the nervous system. Simple motor com-
positive effects on the peripheral systems, there mands such as reflexes are directly controlled by
is accumulating evidence that physical activ- the spinal cord. Special types of neurons within
ity can also significantly affect brain structures the central nervous system (CNS) play a key
and brain functioning. The exercise-induced role in motor movement. These efferent nerves
effects on the brain are partially mediated by are called motoneurons, since they project their
alterations in endogenous peptides, amino axons outside the CNS to directly or indirectly
acid transport through the blood-brain barrier control muscles. Each motoneuron sends its axon
(BBB), and neurotransmitter alterations as well to one muscle and innervates a limited number
as changes in energy metabolism. Since physical of muscle fibers, a complex called the motor unit.
activity is linked to neuronal activity and neu- Motoneurons are activated by interneurons of
ronal firing itself consumes much of the energy different motor programs or reflex centers as well
in the brain, physical exercise is closely associ- as by descending tracts from the brainstem and
ated with changes in brain energy metabolism. the forebrain. The average motoneuron receives
With ongoing physical activity, the consump- many thousands of synaptic inputs (Brnnstrm,
tion of oxygen (O2) and glucose increases in 1993) that relay instructions from higher neu-
brain regions involved in motor movement. ronal centers and provide signals from sensory
However, with increasing exercise intensity, modalities in the periphery. Motoneurons that
brain activation increases and changes in brain control different muscles are located in the spinal
energy homeostasis occur, since neurons prefer cord and in the brainstem. Motoneurons supply-
lactate to glucose during high energy conditions ing different muscles can be activated with great
as their primary energy substrate, which raises precision by these different neuronal sources,
the production of the high-energy substrate which determine the degree of activation and
adenosine triphosphate (ATP). Furthermore, the exact timing of the motoneurons of a given
high-intensity exercise increases the ratio of muscle. The recruitment and fine control of
metabolic demand to metabolite availability motoneurons to produce voluntary coordinated
with an accompanied change in brain energy movements therefore requires that multiple
status. This chapter gives an overview of the influences interact in the appropriate sequence
effects of physical activity on homeostatic pro- and with the right balance.
cesses in the brain with a special consideration The region of the cerebral cortex most
of brain energy metabolism and its potential involved in the planning, control, and execu-
implications on physical and mental health and tion of voluntary movements is the motor cortex.
performance. The motor cortex is located in the rear portion
Exercise 299

of the frontal lobe and is divided into two main signaling across several cortical and subcortical
areas:Area 4 (primary motor cortex) and Area 6. areas associated with changes in brain neuro-
The primary motor cortex forms a thin band transmission and metabolism (Ide and Secher,
along the central sulcus. Area 6 is wider and is 2000; Figure17.1).
further subdivided into two distinct subareas. Activity in the frontal regions of the brain
The lateral portion of Area 6 (premotor area) has been related to affective and perceptual
guides body movements by integrating sensory responses to acute bouts of exercise (Nybo and
information and controls muscles that are clos- Nielsen, 2001; Petruzzello etal., 2006). Dynamic
est to the bodys main axis, while the medial movements are associated with cortical activa-
part (supplementary motor area) is involved in tion and increases in blood flow to the primary
planning more complex movements. However, sensorimotor area and supplementary motor
besides the motor cortex, other cortical areas area (Orgogozo and Larsen, 1979). Changes in
also play important roles in generating voluntary neuronal activity and the cerebral metabolic rate
movements, such as the prefrontal cortex and the for glucose determined after running indicate
posterior parietal cortex. an involvement of the hypothalamus, the poste-
The basal ganglia, found below the motor rior parietal cortex, the temporoparietal cortex,
cortex, further help to organize motor programs the prefrontal cortex, as well as the premotor
for complex movements. The main basal ganglia and the primary motor cortex (Orgogozo and
are the striatum (caudate nucleus and the puta- Larsen,1979).
men), the globus pallidus, the substantia nigra, Neuroimaging studies of physical activity in
the nucleus accumbens, and the subthalamic humans have shown differences in brain activ-
nucleus, which are tightly interconnected since ity that are closely related to physical activity.
they also receive information from several dif- Examination of baseline spectral frequency
ferent regions of the cerebral cortex. The impor- distributions of electroencephalograms (EEGs)
tance of these subcortical nuclei for normal has revealed increased activation in the theta
brain function and behavior is emphasized by
the numerous and diverse neurological condi-
tions associated with basal ganglia dysfunction, Cortex
Central Command
which include disorders of behavior control such Basal ganglia
as Tourette syndrome and obsessivecompulsive Cerebellum
disorder; dystonia; psychostimulant addiction; Brainstem
Motoric pattern
and movement disorders, the most notable of coordination Spinal Cord
which are Huntingtons disease, which primarily
involves damage to the striatum, and Parkinsons
disease, which is caused by the degeneration of Motoneurons
Sensory feedback

the dopamine-producing cells in the substantia


nigra (Stocco etal., 2010; Fix,2008).
The timing and coordination of motor tasks Effektor organs
Muscle
and the integration of sensory and motor infor-
mation is coordinated in the cerebellum, and its
damage causes loss of coordination. The cerebel- Movement
lum and the basal ganglia receive information
from various parts of the brain and send this
information to the motor cortex via neurons in
the thalamus, forming the cortico-basal ganglia Sensory receptors
loop and the cortico-cerebellar loop. The neu-
rons from the cortex send their axons via the cor-
ticospinal tract to the opposite side of the body
FIGURE 17.1: Neural regulation of movement.
and connect either to the appropriate cranial
Motor movements are planned and refined in the motor
nerves or to the appropriate spinal nerve in the
cortex, basal ganglia, and cerebellum. Interneurons
spinal cord, where the spinal nerves connect to
within the spinal cord and brainstem send its axon to
particular muscles or muscle groups, thereby giv-
the muscles and innervate the muscle fibers to gener-
ing them the potential to directly control move-
ate movement. Sensory feedback initiates, informs, and
ments. Therefore, the conductance of voluntary
modulates the output at any stage of motor control.
movements is generated by changes in neuronal
300 Part III:Homeostatic Manipulators

(48 Hz), alpha (813 Hz), and beta (1320 Hz) arm crank exercise, indicating that specific brain
spectral bands and higher mean frequency in the activation patterns are linked to different kinds
delta (0.254 Hz), theta, and beta bands in more of exercise and participants physical exercise
active or aerobically fit individuals (Hillman preferences (Schneider et al. 2009; Hanakawa
etal., 2008; Bashore, 1989; Dustman etal., 1990; etal.,2003).
Lardon etal., 1996; Mecklinger etal., 1992). These Therefore neuronal activity within differ-
findings suggest that physical activity influences ent brain regions is directly affected by exer-
baseline electrocortical function and thus has the cise, where the magnitude of brain activation
potential to affect cognitive operations. increases with the intensity and type of exercise
In addition to the effects of exercise on base- (Williamson etal., 1999; Schneider etal., 2009),
line cortical functioning, motoric movements and the brain may become maximally stimu-
are directly associated with changes in neuronal lated when exercise is performed at a level near
activity. During and following aerobic and anaer- exhaustion (Kayser,2003).
obic exercise, there is an increase in EEG activ-
ity in the theta, alpha, and/or beta frequencies B R A I N E N E R G Y M E TA B O L I S M
(Crabbe and Dishman, 2004; Kamijo etal., 2004; DURING EXERCISE
Nielsen etal., 2001; Nybo and Nielsen, 2001). The
increase in EEG activity is associated with the Cerebral Blood Flow and O2
workload of exercise. Studies show that brain Consumption
activity increases above resting levels at higher In addition to the changes in peripheral circu-
workloads especially during anaerobic activities lation during in physical exercise also cerebral
and during muscle fatigue during graded exer- blood flow (CBF) increases. One major factor
cise (Bailey et al., 2008; Brmmer et al., 2011). responsible for the increase in CBF during exer-
The increases in EEG activity were seen across cise is the increase in neuronal activity needed
EEG frequencies (theta, alpha 1, alpha 2, beta 1, to co-ordinate motor control (also described as
beta 2) and electrode sites between the frontal the central command), including ventilatory
(F3, F4, F7, F8), central (C3, C4), and pariental control and control of circulation (Goodwin
(P3, P4) lobes. Furthermore, EEG changes were etal., 1972; Ide and Secher 2000). Furthermore,
not localized to either hemisphere. While older CBF is influenced by the vast sensory input from
studies showed an increase of around 20% in mechanically and chemically sensitive nerves
activity after exercise close to exhaustion with within the muscles (Sato et al., 2009 a, 2009b).
a mean maximum heart rate of 200 beats per Central command and integration of informa-
minute (Beaussart et al., 1959), further stud- tion from activation especially of the mechani-
ies confirmed that the increase in alpha activity cally sensitive nerve fibers work in concert to
was strongest within the first 10 min after exer- elicit the increase in CBF. The relative contri-
cise and returned to pre-exercise baseline after bution of these influences varies depending on
30 min of recovery (Mechau etal., 1998; Boutcher the exercise model, and arm muscles appear to
etal.,1988). require more neural integration from the part
The neuronal activity pattern within the of the brain interrogated by the middle cerebral
brain depends further on the kind of exercise artery than is the case for the leg muscles.
and participants physical exercise preferences An increase in CBF during brain activation
(Schneider et al., 2009). Studies with stand- elicited by exercise has also been shown with
ardized low resolution brain electromagnetic near-infrared spectroscopy (Ide et al., 1999).
tomography to assess EEG activity showed an Besides the changes in whole CBF, regional
increase of alpha and beta activities immedi- changes in CBF (rCBF) and metabolism have been
ately post-exercise widely across the brain. While found during cerebral activation. In humans, CBF
immediately after exercising all three kinds of is transiently increased in the thalamus, ante-
movement led to a significant increase of beta rior cingulate, insula, and sensorimotor cortex
activity in the Brodmann area 7, an area that during static handgrip (Rogers et al., 1990) and
plays an important role in motor coordination, in the thalamus and insula during dynamic
15- and 30-min post-exercise a specific activation cycling exercise (Romer et al., 2006). A distinct
pattern in the beta-frequency range character- increase in both rCBF and regional metabolism
ized by an decrease in frontal brain activity and has become synonymous with cerebral activa-
an increase in occipital regions was recognized tion as visualized by single-photon emission
after bike and treadmill exercises but not after tomography, positron emission tomography, and
Exercise 301

functional magnetic resonance imaging. The organism, as defined by changes in brain oxygen
motor tasks involved in exercise have been asso- and glucose metabolism and the associated pro-
ciated with an increase in rCBF and confirmed duction on ATP, the energy currency of brain cells
in numerous follow-up studies (Gonzalez-Alonso (Magistretti, Pellerin, Rothman, and Shulman,
etal., 2004; Madsen etal., 1993; Ide and Secher, 1999). This association is based on the direct rela-
2000; Sato etal.,2009). tionship between neuronal activity and energy
Early studies showed that cerebral oxygenation consumption as suggested as early as 1890 by Roy
increases during activation by exercise (Astrand and Sherrington. Energy metabolism is highly
etal., 1964). This is in contrast to the progressive organized within cells and tightly coupled to neu-
reduction in muscle oxygenation with increas- ronal activity, where postsynaptic potentials and
ing workload. Also, brain function deteriorates associated ATP-consuming ion pumping domi-
when its average oxygenation becomes reduced nate the energy requirements for neuronal sign-
more than 10% (Gonzalez-Alonso et al., 2004; aling (Ames, 2000; Dhar and Wong-Riley, 2009;
Madsen et al., 1999; Van Lieshout et al., 2001; Magistretti 1999; Alle et al., 2009; Magistretti
Van Lieshout etal., 2003), whereas skeletal mus- et al., 1999; Buzski et al., 2007). Eighty-seven
cles maintain their activity despite O2 saturation percent of brain energy consumption is propor-
below 10% (Astrand etal., 1964). The capillaries tional to neuronal firing rates, most of which is
within skeletal muscle are positioned in direct consumed in the restoration of membrane poten-
contact to the muscle cells. However, within the tial after postsynaptic potentials, whereas only
brain the capillaries are protected by extension of 13% of the energy is used to maintain the rest-
the astrocytes covering the entire capillary net- ing potential (Attwell and Gibb, 2005, 179; Alle
work, thereby creating the BBB, which directly etal., 2009, 165; Magistretti, 2009). This postsyn-
builds an obstacle between capillaries and neu- aptic potentialslinked increase in brain energy
rons. During brain activation, O2 consumption consumption is a function of elevated activity of
increases and the diffusion distance for O2 can the sodium-potassium pump (Na+K+-ATPase),
become critical. To satisfy the enhanced neuronal which is triggered by the influx of sodium ions
metabolism, an increase in rCBF is required to and efflux of potassium ions accompanying post-
generate an elevated O2 gradient. Maximal exer- synaptic potentials or action potential depolari-
cise is associated with a reduced mitochondrial zations. Therefore, increases in neuronal activity
O2 tension as CBF is influenced by the reduction are tightly correlated with elevated concentra-
in partial pressure of carbon dioxide in the blood tions of intracellular Na+ and extracellular K+.
in addition to a decline in arterial O2 content These changes lead to increased Na+K+-ATPase
(Nielsen et al., 1998; Nielsen et al., 2002). The activity to restore ionic gradients across the mem-
reduced cerebral oxygenation in turn affects the brane to their resting levels. Supporting evidence
recruitment of motor units, and supplemental O2 was provided by a recent study that demonstrated
enhances cerebral oxygenation and work capac- a coupling between transcriptional regulation of
ity without effects on muscle oxygenation (Secher Na+K+-ATPase and that of the glutamate trans-
etal.,2008). porters involved in glutamate reuptake as an
The exercise-related changes in brain activ- outcome of increased neuronal activity (Rose
ity are closely connected to changes in CBF and et al., 2009). Therefore, the increase in excita-
oxygen consumption and may occur primary or tory neurotransmission and associated neuronal
even secondary to the metabolic changes during activity are closely linked with increases in brain
prolonged exercise (Dalsgaard and Secher, 2007; energy usage. At high exercise intensity and
Davis and Bailey,1997). hence intense regional neuronal activity, energy
Physical activity leads to an increased meta- demand may exceed energy supply and an imbal-
bolic rate in the active tissues, foremost in the ance may occur in brain regions activated during
active limb locomotor muscles but obviously exhaustive exercise associated with changes in
also in the heart and the respiratory muscles and brain energetics (Dworak etal.,2007).
in activated brain structures associated with a
given physical activity. Exercise-related brain Glucose and Lactate Metabolism
activation is associated with an increase in brain Glucose represents the main fuel for the human
energy usage, since the brain is one of the most brain and can support its energy demands.
energy-demanding organs in the body. Although Glucose can produce metabolic intermediates,
constituting only 2% of body mass, brain energy such as lactate and pyruvate, which do not neces-
utilization account for ~20% of those of the whole sarily enter the tricarboxylic acid cycle but rather
302 Part III:Homeostatic Manipulators

can be released via circulation. Glucose can also through nicotinamide adenine dinucleotide
be incorporated into lipids, proteins, and glyco- (NADH) and flavin adenine dinucleotide for-
gen or serve as the substrate for neurotransmit- mation. Alternatively, pyruvate is reduced to
ters such as -aminobutyric acid, glutamate, and lactate while reoxidizing cytosolic NADH to
acetylcholine (Hoyer, 1990). Moreover, glucose NAD+ as a prerequisite for substrate flow in
is not the only source of ATP at a given time in anaerobic glycolysis. The near-equilibrium state
the brain: other molecules can also contribute of the responsible enzyme, lactate dehydroge-
to ATP production, although in smaller quanti- nase, implies that the ratio of lactate to pyruvate
ties. For example, glycogen, located primarily in cytoplasm reflects that of NAD+ to NADH.
in astrocytes, is an important reserve of glucose Thus a high ratio of lactate/pyruvate in cerebral
equivalents and contributes ~1% of the cerebral arterial-venous differences or cerebrospinal fluid
metabolic rate of glucose in humans and rats is taken to indicate insufficient oxygen supply.
(Gruetter, 2003). During exercise, the brains Nevertheless, pyruvate seems to be less effective
energy demands increase significantly. Transient than lactate as substrate, at least during reperfu-
increases in local cerebral glucose use in the sion/ischemia (Phillis etal.,2001).
somatosensory, auditory, and visual cortices; the During light exercise, a small increase in
dorsal raphe; and specific areas of the hypothala- brain lactate release, relative to lactate uptake,
mus, amygdala, and hippocampus have been is observed that might be related to increased
reported in response to acute strenuous treadmill neural activity. With increasing exercise inten-
running in rats (Vissing etal.,1996). sity, more lactate is produced in skeletal muscles
Besides glucose, other substrates can also be and released to the blood. The increase in arte-
used to provide sufficient energy to the cells (Ide rial lactate concentration during intense exer-
et al. 2000; Schurr, 2006). For example, during cise makes lactate available to the brain, and the
long-term fasting the concentrations of ketone brain switches from a net producer of lactate to
bodies are increased, and they replace glucose as an importer while glucose uptake is reduced (Ide
the primary energy source for the brain (Owen etal., 2000, Kemppainen etal., 2005). In fact, the
etal., 1967). Energy metabolism is highly organ- arterial lactate concentration is highly correlated
ized to provide sufficient energy, mostly in the with the unidirectional brain lactate uptake. The
form of ATP, during periods of high energy lactate taken up from skeletal muscle can act as
demand as well. From rest to high-intensity an intercellular energy shuttle within the brain
exercise, skeletal muscles increase their energy during high-intensity exercise (Dalsgaard et al.,
requirements more than 50-fold, which is asso- 2006). Under situations of a high energy demand,
ciated with ATP consumption and significant neurons prefer lactate to glucose as their primary
increases in lactate production. Under resting energy source, which raises the production of
conditions, the skeletal muscle and the brain ATP (Schurr, 2006; Magistretti etal., 1999), and,
release only small quantities of lactate. However, under exercise conditions, lactate oxidation is
both tissues continuously take up lactate, responsible for approximately 8% of the brain
although in lesser quantities than simultaneously energy requirements (van Hall etal., 2009). The
released, resulting in the small net lactate release lactate taken up by the brain is almost completely
(van Hall etal.,2009). oxidized and does not accumulate in the brain
The monocarboxylic acids such as lactate under the various conditions, which is remark-
and pyruvate are taken up by the brain when the ably different from skeletal muscle in which near
blood level increases. Such transport across the complete oxidation was observed only during
BBB is facilitated by the monocarboxylate trans- exercise (van Hall et al., 2009). The changes in
porter. This transporter is expressed widely blood biochemistry during exercise can directly
throughout the rodent brain, with the high- affect brain activity and cortical functioning,
est concentrations in areas of high neuronal since exercise at high intensities, with blood lac-
activity and metabolism such as the cortex, the tate accumulation, decreases significantly the
hippocampus and the cerebellum (Hertz and EEG beta-2, beta-1, and alpha-1 frequency bands
Dienel, 2005; Pierre and Pellerin, 2005; Pellerin (Bailey etal.,2008).
etal., 1998; Pierre and Pellerin, 2005). Pyruvate, The metabolic response to brain activation in
another monocarboxylic acid and the end prod- exercise can be defined as the cerebral metabolic
uct of glycolysis, is converted to acetylcoenzyme ratio (MR; uptake O2/glucose + 1/2 lactate). At
A, a substrate for the tricarboxylic acid cycle and rest, brain energy is provided by a balanced oxi-
subsequent respiration, albeit only indirectly dation of glucose and the MR is close to 6, but
Exercise 303

further activation provokes an excessive uptake et al., 2005), thus strongly influencing neuronal
of glucose relative to O2. While the MR remains information processing, synaptic strength, gene
stable during light exercise, it is decreased dur- expression, and protein, fatty acid, and glycogen
ing prolonged exhaustive exercise where blood synthesis (Fields and Stevens, 2000; Peters etal.,
lactate remains low, but when vigorous exercise 2004; Pascual etal.,2005).
raises blood lactate, the brain takes up lactate Intracellular ATP levels drop from 4 to 8 mM
in an amount similar to that of glucose, result- to 2 to 3 mM during muscle fatigue (Harris,
ing in a reduced MR by 30% to 40% (Ide and 1996). The energy charge of the adenylate pool
Secher, 2000; Sato et al., 2009). Interestingly, ([ATP]+1/2[ADP]/ ([ATP]+[ADP]+[AMP]) has
studies showed that the MR depends also on the been proposed as a control parameter in the reg-
central command, since a decrease in the MR ulatory interactions by which biological homeo-
is also observed under partial neuromuscular stasis is maintained (Atkinson and Walton,
blockade, when the will to exercise is intense 1967). Decrease in ATP concentration results
(Sato et al., 2009). Intense regional neuronal in increases in ADP and more so in adenosine
activity in cerebral regions during high exer- monophosphate (AMP) and adenosine ADO,
cise intensities cause energy demand to exceed but without drastic changes in the overall energy
production, in turn draining energy reserves, charge. In normal brain tissue, the energy charge
which is hypothesized to result in local energy is closer to 0.85 (Derr and Zieve, 1972) and is
depletion such as brain glycogen and might play regulated by direct participation of adenine
a role in central fatigue (Dalsgaard etal., 2002; nucleotides in energy-converting processes, such
Dworak etal., 2007; see also Central Fatigue as glycolysis and oxidative phosphorylation,
section). and many energy-expending biosynthetic path-
ways, such as those of amino acids, nucleic acids,
Nucleotide and Nucleoside protein, fatty acids, and cholesterol synthesis
Metabolism (Thompson and Atkinson,1971).
At high intensities exhaustion times are short, During intense neuronal activation such
whereas at low intensities exercise can be main- as exhausting physical activity, when the cen-
tained longer. This relationship is determined by tral command and sensory input to the brain is
the maximal use of energy sources available to intense, metabolic demand exceeds metabolic
the organism:alactic anaerobic (Phosphocreatine availability and results in an increased break-
and ATP), lactic anaerobic (anaerobic glyco- down of ATP associated with an increase in the
lysis), and aerobic (sugar and fat oxidation). For nucleotide ADO (Basheer etal.,2004).
short-duration exercises, alactic anaerobic and Previous studies showed significantly
lactic anaerobic metabolism provide sufficient higher ADO concentrations in the rat neostria-
energy to the organism, whereas for lower-power tum and in the hippocampus during the active
outputs aerobic metabolism becomes the main period of rats compared to the sleeping period,
energy source and maximum glucose and fat oxi- a finding that was interpreted as an associa-
dation rates set the limits (Monod and Scherrer, tion of ADO concentrations with motor activ-
1965; Wilkie, 1981). One vital function of the ity (Huston etal., 1996). The increase in brain
metabolic network is to maintain stable concen- ADO concentrations seems to depend on exer-
trations of cellular ATP by adjusting the reac- cise intensity. While moderate exercise did
tion rates to ensure a continuous energy supply not affect brain ADO levels, intense exercise
for sustaining electrophysiological activity and increased the ratio of metabolite demand to
maintaining normal function in the brain. ATP metabolite availability with an accompanied
is the currency of cellular energy and besides its production of ADO from ATP breakdown
role as major energy molecule is also an impor- (Dworak etal.,2007).
tant molecule in cell-to-cell signaling (Burnstock These observations were supported by show-
et al., 2011). Thus anabolic and catabolic pro- ing that minimal exercise did not affect ADO
cesses must be well balanced to maintain con- concentrations in the basal forebrain if meas-
stant levels ofATP. ured via microdialysis (McKenna et al., 2007).
ATP acts as an activity-dependent metabo- The observed findings of increased ADO con-
lite and signaling molecule between neurons and centrations during exhausting exercise might
glia and modifies membrane potentials through reflect a state of bioenergetic stress, possibly as a
ATP-modulated potassium channels (Fields result of an increased breakdown of high-energy
and Stevens, 2000; Peters et al., 2004; Pascual phosphates.
304 Part III:Homeostatic Manipulators

FIGURE17.2: Homeostatic neurometabolic regulation during exercise. The increased excitatory neu-
rotransmission during intense exercise results in a higher activity of Na+-K+-ATPase to restore the concentration
gradient. Na+-K+-ATPase activity is a high energy consuming process, resulting in the breakdown of adenosine
triphosphate (ATP) and the generation of adenosine. Adenosine acts via A1 receptors to inhibit neurotransmit-
ter release and provoke a hyperpolarization, thereby inhibiting neuronal activity. 5-HT=serotonin, LC=locus
coreuleus, NA=noradrenalin.

The changes in brain ATP and ADO concentra- influenced by exercise, since it was shown that
tions during exercise can directly affect behavior. physical activity directly affects the central dopa-
ADO acts as a neuromodulator, and its effects are minergic, glutamatergic, noradrenergic, and ser-
mediated via specific G-protein coupled recep- otonergic systems (Meeusen etal.,1995).
tors, namely A1, A2a, A2b, and A3 (Haas etal.,
2000). The net effect is a presynaptic reduction Dopamine
in transmitter release in wakefulness-promoting In the brain, dopaminergic systems are primar-
networks, including the cholinergic and mono- ily involved in reward-motivated behavior, motor
aminergic systems, as well as a postsynaptical control, and control of the release of several impor-
hyperpolarization and accompanied EEG desyn- tant hormones. Exercise increases dopamine lev-
chronization (Rainnie et al., 1994; Latini et al., els in the brain through a calcium-dependent
2001). The basal forebrain and the mesopontine process that regulates numerous brain functions
cholinergic neurons, whose discharge activity (Sutoo etal., 2003). Dopamine levels were regu-
plays an integral role in EEG arousal and mainte- lated by exercise in epileptic and spontaneously
nance of wakefulness, are especially postsynapti- hypertensive rats, demonstrating the possibility
cally inhibited by endogenous ADO (Latini and that exercise can be used to improve symptoms
Pedata, 2001; Arrigoni etal., 2006; Figure17.2). of Parkinsons disease or dementia (which are
associated with low dopamine levels; Sutoo etal.,
Neurotransmission 2003). Regular aerobic exercise has a protective
During Exercise effect on D2 dopamine receptor levels and inhib-
Associated with the changes in neuronal signaling its modifications in brain dopamine metabolism
throughout the brain regions involved in motor due to the aging process (Uysal etal., 2005). The
control, physical activity directly produces alter- release of dopamine by neurons is necessary for
ations in endogenous peptides, increased amino sustaining neural activity and working memory
acid transport through the BBB, and changes (Surmeier etal., 2007). Dopamine is also a pre-
in the release of various neurotransmitters (Ide cursor to norepinephrine and epinephrine, and
etal., 1999, Herholz etal., 1987; Meeusen etal., levels of dopamine are responsive to levels in
1995). The release of most neurotransmitters is serotonin.
Exercise 305

Synthesized from dopamine, norepinephrine is needed to elevate serotonin (5-HT) synthesis


acts in the central and sympathetic nervous sys- in the brain, whereas anaerobic activities such as
tems by binding to adrenergic receptors. Brain weight lifting and stretching exercises have not
norepinephrine levels are affected after acute been associated with increased brain serotonin
bouts of exercise (running or swimming). Most levels. Hence, it seems that brain 5-HT levels
studies found a decrease or no affect in brain nor- depend on exercise quality (aerobic vs. anaerobic,
epinephrine levels. It seems that acute exercise local vs. whole-body) and quantity (duration).
results in a depletion of brain norepinephrine Chronic activity wheel running or treadmill
levels probably due to an acceleration in nor- running results in small increases in basal levels
epinephrine turnover by activating hydroxylase of 5-HT in the dorsal raphe nucleus (Dishman
activity (Chaouloff, 1989). After chronic exercise et al., 1997) and increased turnover of 5-HT in
training, brain norepinephrine levels increased the brain cortex (Yoo etal; 2000). Chronic wheel
significantly. The effect of norepinephrine neu- running also attenuates stress-induced c-Fos
rotransmission also shows a region-specific pat- induction in serotonergic neurons of the dor-
tern. While norepinephrine levels decreased due sal raphe nucleus and increases levels of 5-HT
to acute exercise in the brainstem, hippocampus, 1A inhibitory autoreceptor mRNA (Greenwood
midbrain, and hypothalamus, increased norepi- etal., 2003; Greenwood etal., 2005). The antide-
nephrine levels were observed in the striatum, pressant and anxiolytic effects of exercise have
cortex, and preoptic area (Meeusen et al., 1995; been clearly demonstrated in different studies,
Dishman et al., 2000). Therefore, brain norepi- and the positive effects might be partially due to
nephrine levels can be directly affected by exer- the exercise-induced change in 5-HT neurotrans-
cise, but the alterations differ between specific mission (Salmon etal., 2001; Young etal.,2007).
brain regions and depend on the type of exercise The interactions between brain neurotrans-
(aerobic vs. anaerobic). mitters, metabolites, and their specific receptors
could also play a central role in the onset of cen-
Glutamate tral fatigue during prolonged exercise.
Glutamate is the most common neurotransmit-
ter in the brain, as well as the major excitatory Central Fatigue
neurotransmitter involved in many aspects of Physical fatigue can originate within the muscle,
brain function, including learning and memory which is known as peripheral fatigue, or within
(Chaddock et al., 2011). Glutamatergic activity the CNS, which is known as central fatigue.
changes as a function of behavioral states, since Several biological mechanisms have been pro-
cortical glutamate levels increase progressively posed to explain CNS fatigue (Davis etal., 1997;
during periods of increased neuronal activity Meeusen etal., 2006), mainly focused on altera-
such as waking and rapid eye movement (REM) tions in several neurotransmitters and metabo-
sleep and decrease progressively in non-rapid lites including 5-HT, dopamine, acetylcholine,
eye movement (NREM) sleep, when neuronal andADO.
activity is decreased significantly (Dash et al., The original central fatigue hypothesis indi-
2009). Animal studies have shown that tread- cates that an exercise-induced increase in extra-
mill running significantly increases glutamate cellular 5-HT concentrations in several brain
levels during and for a short while after exercise regions contribute to the development of fatigue
in rats (Leung etal., 2006)and may influence the during prolonged exercise. 5-HT has been linked
levels of glutamate receptors such as NR2B and to central fatigue because of its established
mGluR5. (Zhang etal., 2010). Moreover, most of effects on sleep, drowsiness, and loss of motiva-
the brain energy consumption is directly associ- tion (Meeusen et al., 2006). Several nutritional
ated with glutamatergic (Attwell and Laughlin, and pharmacological studies have attempted to
2001). These observations are in accordance manipulate central serotonergic activity during
with the increase in neuronal activity within exercise, but this work has yet to provide robust
different brain regions during and shortly after evidence for a significant role of 5-HT in the
exercise. central fatigue process (Davis and Bailey 1997;
Meeusen etal., 2006). Under normal physiologi-
5-Hydroxytryptamine (Serotonin) cal conditions, 5-HT is unable to cross the BBB,
Serotonin is closely regulated by physical activity. therefore cerebral neurons are required to syn-
It has been found that at least 30 min of daily aer- thesize it by themselves and overall brain 5-HT
obic activity such as running, biking, or walking levels arelow.
306 Part III:Homeostatic Manipulators

The precursor of 5-HT is tryptophan, and that during prolonged physical activity, brain
the synthesis as well as the transport through dopamine activity increased (Bailey etal., 1993;
the BBB of 5-HT is driven by the blood supply Gerald etal., 1978), which may influence exercise
of free tryptophan. Tryptophan is transported performance (Freed etal., 1985). Brain serotonin
via the L-system, the amino acid transporter sys- and dopamine content progressively increased
tem, which also transports the other large neutral during exercise, but at the point of exhaustion
amino acids, including the three branched-chain a marked fall in tissue dopamine content was
amino acids (BCAA) and that compete with apparent. Animal studies show that at the point
tryptophan for entry into the brain. Therefore, of fatigue dopamine extracellular concentrations
the amount of tryptophan transported into the are low, possibly due to the interaction with brain
brain depends not only on the concentration of serotonin (Davis et al., 1997) or a depletion of
free tryptophan in the bloodstream but also on central catecholamines (Bailey etal.,1992).
the concentrations of the BCAA, since these Acetylcholine modulates arousal and temper-
make up approximately 75% of the total large ature regulation and might play a role in fatigue.
neutral aminoacids. During exercise, the levels of acetylcholine
The concentration of free tryptophan in the decrease due to a drop in plasma choline levels.
plasma is mainly influenced by the change in However, the results in studies about the effect of
plasma free fatty acids, as these are also trans- acetylcholine on central fatigue are conflicting.
ported bound to albumin in the plasma. An Conlay et al. (1992) found that plasma choline
increased release of fatty acids from the adipose levels decrease by 40% after a marathon, whereas
tissue during exercise results in an increase in some studies have found that supplementation
their concentration in plasma and displaces with choline citrate improved performance in
some of the tryptophan from albumin, thereby long-distance runners and other studies did not
elevating the plasma level of free tryptophan find any improvement in time to exhaustion after
(Bloomstrand,2001). choline supplementation (Spector et al., 1995).
Also, the plasma concentrations of amino The same study also found that plasma choline
acids vary during exercise and are dependent levels had not changed in either the placebo or
on the type of exercise and the duration and the choline-supplemented groups. Therefore
intensity and of the exercise period (Henriksson, more research is needed to investigate acetylcho-
1991). During short-term exercise, there is an lines effects on fatigue.
increase in most amino acids, whereas prolonged Neuromodulators such as cytokines, ammo-
exercise causes a decrease in the concentration of nia, and ADO can also influence central fatigue
most amino acids, such as BCAA (Bloomstrand during exercise. Increases in several cytokines
et al., 2000). The change in the plasma ratio of have been associated with reduced exercise toler-
free tryptophan/BCAA during prolonged exer- ance associated with acute viral or bacterial infec-
cise depends in part on exercise intensity; an tion. Accumulation of ammonia in the blood and
increased ratio was found during the last hours brain during exercise could also negatively affect
of exercise of a 5-hour ergometer exercise at 75% the CNS function and fatigue (Meeusen et al.,
of the maximal oxygen uptake, whereas no sig- 1995; Davis etal.,1997).
nificant change was found during exercise at 50% Recent studies suggest that ADO might play
of the maximal oxygen uptake (Strder et al., a central role in central fatigue (Dworak et al.,
1997). Thus a marked increase in the plasma ratio 2007). ADO is derived from the metabolism of
of free tryptophan/BCAA is found after exercise ATP and is a well-known homeostatic sleep fac-
with longer duration and at higher intensities, tor that links energy metabolism with neuronal
which favors the transport of tryptophan into the activity (Brown etal., 2012). Systemic and central
brain and also the synthesis and release of 5-HT administrations of ADO or ADO A1 receptor ago-
from some neurons that could be responsible for nists induced sleepiness and impaired vigilance
fatigue during and after sustained heavy exercise (Brown etal., 2012). Stimulation of neuronal ADO
(Meeusen etal., 2006; Bloomstrand etal., 2000; receptors mediates presynaptic inhibition in the
Figure17.3). transmitter release, including cholinergic, adren-
Another potential neurotransmitter in cen- ergic, and serotonergic neurons (Rainnie et al.,
tral fatigue is dopamine. Dopamine is an excita- 1994; Arrigoni etal., 2006)as well as postsynaptic
tory neurotransmitter that regulates arousal, hyperpolarization that regulate alertness, sleep,
motivation, endurance performance, and mus- and wakefulness (Basheer et al., 2004; Latini
cular coordination. Animal studies showed and Pedata, 2001). Intense exercise increases
Exercise 307

Blood BBB Brain


intracellular extracellular

Lactate Lactate ATP ATP

Protein
AD AD

FFA LNNA Dopamin


Albumin Acetylcholine
Norephedrine
Tryptophan BCAA
Albumin Tryptophan 5-HT

FATIGUE

FIGURE 17.3: Neuronal mechanisms of central fatigue. Prolonged and intense exercise causes an
increased release of fatty acids from the adipose tissue and elevated concentrations in plasma free fatty acids
(FFA). FFA displace some of the tryptophan from albumin, thereby elevating the plasma level of free tryptophan.
Also, the concentrations of most amino acids, such as BCAA, drop during prolonged exercise thereby increas-
ing the plasma ratio of free tryptophan/BCAA. Free tryptophan is transported via large neutral amino acids
(LNNA) transporter through the blood-brain barrier (BBB) into the brain, thereby increasing the level of brain
tryptophan and 5-HT. Exercise also increases the blood concentrations of lactate. Peripheral lactate can enter the
brain via specific transporters and provide additional energy under intense physiological conditions. Increased
neuronal activity during intense and prolonged exercise is associated with changes in dopamingergic, cholinergic,
and adrenalinergic neurotransmission and an increased cotransmission of adenosine triphosphate (ATP) to the
extracellular space. ATP is metabolized to adenosine ADO. ADO acts via neuronal ADO receptors by mediating
presynaptic inhibition in the transmitter release, including cholinergic, adrenergic, and serotonergic neurons, and
results in a postsynaptic hyperpolarization that can directly affect behavior, performance, and fatigue.

brain ADO concentrations due to an increased effects of caffeine. Caffeine is a potent ADO
energy demand versus limited energy availabil- antagonist and CNS stimulant that easily crosses
ity (Dworak etal., 2007; Brown etal., 2012). The the BBB due to its lipophilic properties (Graham
increase in brain ADO concentrations and release et al., 2001). It has been shown to counteract
to the extracellular space during exercise is a most of the inhibitory effects of ADO on neuro-
potential mediator of fatigue due to the inhibitory excitability (Fredholm etal., 1999; Gervitz etal.,
action of ADO on excitatory neurotransmission 2001), neurotransmitter release (Okada et al.,
(Rainnie etal., 1994; Arrigoni etal., 2006). This 1997), and arousal (Brown etal., 2012). In addi-
relationship is also supported by studies show- tion, blocking CNS ADO receptors may help to
ing that exercise improves sleep quality (Driver explain the fatigue-delaying properties of caf-
etal., 2000; OConnor, Youngstedt, 1995). Intense feine. Ingestion of caffeine has been shown to
(anaerobic) bicycle exercise increases restorative delay fatigue during prolonged intense exercise
NREM sleep (also known as stage 4 sleep), which in both human and animal models (Cole et al.,
correlates closely with brain ADO levels (Dworak 1996; Costill et al., 1978; Jackman et al., 1996;
etal., 2008). During NREM sleep, in turn, resto- Kovacs et al., 1998). Caffeine does not improve
ration of energy stores occurs particularly of ATP maximal oxygen capacity directly but could per-
and glycogen in muscles and in the brain, which mit the athlete to train at a greater power output
is needed for anabolism, such as protein and gly- and/or to train longer. It has also been shown to
cogen synthesis to support recovery and regenera- increase speed and/or power output in simulated
tion processes (Kong et al., 2002; Dworak et al., race conditions (Graham etal.,2001).
2010; Brown etal.,2012). Thus the capabilities of ADO in modulating
Other evidence for an ADO role in central neuronal activity and alertness by linking energy
fatigue is the exercise performance supporting metabolism with neuronal activity provide a
308 Part III:Homeostatic Manipulators

central role for the nucleotide ADO in the reg- neurotrophin levels and neurogenesis, induc-
ulation of central fatigue during intense and/or ing activity-dependent plasticity (reviewed in
long-lasting exercise. Cotman etal.,2007).
The homeostatic control of brain energy
B E H AV I O R A L E F F E C T S metabolism and the associated changes in neu-
OFEXERCISE ronal activity can influence sleep-wake behavior
Regular participation in physical exercise has directly. Epidemiological studies have consist-
positive outcomes on the cardiovascular system, ently supported the view that acute and chronic
the pulmonary system, and whole-body metabo- exercise promotes sleep (Vuori et al., 1988;
lism and is an important factor in the prevention Youngstedt etal., 1997). Exercise at higher inten-
of cardiovascular diseases and metabolic diseases sities in particular reduces sleep onset latency
such as type 2 diabetes and obesity. Besides the and increases total sleep time and the amount of
positive effects on the cardiometabolic systems, homeostatic regulated NREM slow-wave sleep
physical activity significantly affects brain struc- with accompanied reductions in REM sleep
tures and brain functioning, and exercise-related (Driver etal., 2000; Shapiro etal., 1981; Dworak
changes in brain metabolism, neurotransmis- etal.,2008).
sion, and neuronal firing have direct implications The exercise-induced alterations in sleep
on behavioral factors. quality may be related to changes in brain
Emerging evidence suggests that physi- energy metabolism. High-intensity exercise
cal activity may confer health-protective ben- results in an increased brain energy demand,
efits for several neurological diseases, including increased ATP consumption, and coincident
Parkinsons disease, Alzheimers dementia, and increases in brain ADO concentrations (Dworak
ischemic stroke, as well as injuries from falls etal., 2007), reflecting an energy deficit, which
attributable to neuromuscular declines associ- is also observed during prolonged wakeful-
ated with physical inactivity among the elderly ness (Basheer et al., 2004). Due to its inhibi-
(Karp etal., 2006; Wilson etal., 2002; Colcombe tory actions, ADO serves as an sleep-promoting
etal.,2003). factor in the CNS and might be a key player
Physical training has been observed to selec- in mediating the positive effects of exercise on
tively enhance brain health and cognition on sleep quality (Dworak etal.,2007).
the molecular level, including angiogenesis,
synaptogenesis, and neurogenesis, as well as to CONCLUSION AND OUTLOOK
upregulate a number of neurotrophic factors Besides the positive effects of exercise on the
such as brain-derived neurotrophic factor and peripheral system, there is accumulating evi-
insulin-like growth factor 1, which are necessary dence that physical activity influences brain
for neuronal differentiation and synaptic plas- functioning. The exercise-induced effects on
ticity (Uysal et al., 2005; Vaynman et al., 2006; the brain are related to alterations in neuronal
Cotman et al., 2007; Cotman and Berchtold, signaling, neurotransmission, endogenous pep-
2002; Cotman and Engesser-Cesar, 2002). Also, tides, amino acid transport through the BBB,
in animals, voluntary exercise leads to increased and metabolism. The exercise-induced changes
axon regeneration and neuronal growth com- in these systems depend on the quality and
pared to being sedentary (Molteni et al., 2004). quantity of exercise and underlie homeostatic
Exercise increases the expression of genes that factors within the brain. Therefore exercise
are required for rapid neural growth in the has the potential to affect brain homeostasis at
brain and the growth directly correlated with the cellular, molecular, and behavioral levels,
the amount of exercise the animal participated implying that physical activity participation is
in, specifically the total distance the animal had beneficial to brain health, cognition, and mood.
run (Molteni et al., 2004). The exercise-induced Such evidence highlights the importance of
alterations in neurogenesis and brain plasticity promoting physical activity across the lifespan
occurred in different brain regions such as the to reverse recent obesity and cardiovascular
hippocampus, hypothalamus, and dentate gyrus disease trends, as well as to prevent or reverse
(Farmer etal., 2004). Neurogenesis in the hypo- cognitive and neural decline. Accordingly,
thalamus is linked to improvements in learn- physical activity can serve to promote health
ing as well as in memory (Cotman et al., 2007; and performance in individuals while also less-
Ruscheweyh etal., 2011). These and other studies ening the health and economic burden placed
show clearly that voluntary exercise can impact on our society.
Exercise 309

References to pathological brain function. Prog Neurobiol.


Alle H, Roth A, Geiger JR. Energy-efficient action 2011;95(2):229274.
potentials in hippocampal mossy fibers. Science. Buzski G, Kaila K, Raichle M. Inhibition and brain
2009;325(5946):14051408. work. Neuron. 2007;56(5):771783.
Ames A. CNS energy metabolism as related to func- Chaddock L, Hillman CH, Buck SM, Cohen NJ.
tion. Brain Res Brain Res Rev. 2000;34(12):4268. Aerobic fitness and executive control of rela-
Arrigoni E, Chamberlin NL, Saper CB, McCarley RW. tional memory in preadolescent children. Med
Adenosine inhibits basal forebrain cholin- Sci Sports Exerc. 2011;43(2):344349.
ergic and noncholinergic neurons in vitro. Chaouloff F. Physical exercise and brain mono-
Neuroscience. 2006;140:403413. amines: A review. Acta Physiol Scand.
Atkinson DE, Walton GM Adenosine triphos- 1989;137:113.
phate conservation in metabolic regulation. Colcombe S, Kramer AF. Fitness effects on the cog-
Rat liver citrate cleavage enzyme. J Biol Chem. nitive function of older adults: A meta-analytic
1967;242:32393241. study. Psychol Sci. 2003;14:125130.
Attwell D, Gibb A. Neuroenergetics and the kinetic Cole KJ, Costill DL, Starling RD, Goodpaster BH,
design of excitatory synapses. Nat Rev Neurosci. Trappe SW, Fink WJ. Effect of caffeine ingestion
2005;6(11):841849. on perception of effort and subsequent work pro-
Bailey SP, Davis JM, Ahlborn EN. Effect of increased duction. Int J Sport Nutr. 1996;6:1423.
brain serotonergic activity on endurance Conlay LA, Sabournjian LA, Wurtman RJ. Exercise
performance in the rat. Acta Physiol Scand. and neuromodulators: Choline and acetylcho-
1992;145(1):7576. line in marathon runners. Int J Sports Med.
Bailey SP, Davis JM, Ahlborn EN. Serotonergic 1992;13(Suppl. 1):S141142.
agonists and antagonists affect endurance Costill DL, Dalsky GP, Fink WJ. Effects of caffeine
performance in the rat. Int J Sports Med. ingestion on metabolism and exercise perfor-
1993;14(6):330333. mance. Med Sci Sports. 1978;10:155158.
Bailey SP, Hall EE, Folger SE, Miller PC. Changes Cotman CW, Berchtold NC. Exercise: A behav-
in EEG during graded exercise on a recum- ioral intervention to enhance brain health
bent cycle ergometer. J Sport Sci Med. and plasticity. Trends Neurosci. 2002
2008;7:501511. Jun;25(6):295301.
Basheer R, Strecker RE, Thakkar MM, McCarley RW. Cotman CW, Berchtold NC, Christie LA. Exercise
Adenosine and sleep-wake regulation. Prog builds brain health: Key roles of growth factor
Neurobiol. 2004;73:379396. cascades and inflammation. Trends Neurosci.
Bashore, T. R. Age, physical fitness, and mental 2007;30:464472.
processing speed. Ann Rev Gerontol Geriat. Cotman CW, Engesser-Cesar C. Exercise enhances
1989;9:120144. and protects brain function. Exerc Sport Sci Rev.
Beaussart M, Niquet G, Gaudier E, Guislain F. The 2002 Apr;30(2):7579.
EEG of boxers examined immediately after com- Crabbe JB, Dishman RK. Brain electrocortical activ-
bat: Comparative study with the EEG recorded ity during and after exercise: A quantitive syn-
before combat in 52 cases. Rev Obstet Ginecol thesis. Psychophysiology. 2004;41:563574.
Venez. 1959;101:422427. Dalsgaard MK. Fuelling cerebral activity in
Bloomstrand E. Amino acids and central fatigue. exercising man. J Cereb Blood Flow Metab.
Amino Acids. 2001;20(1):2534. Review. 2006;26(6):731750.
Boutcher SH, Landers DM. The effects of vigorous Dalsgaard MK, Secher NH. The brain at work: A
exercise on anxiety, heart rate, and alpha activity cerebral metabolic manifestation of central
of runners and non-runners. Psychophysiology. fatigue? J Neurosci Res. 2007;85:33343339.
1988;25:696702. Davis, JM, Bailey SP. Possible mechanisms of central
Brnnstrm T. Quantitative synaptology of func- nervous system fatigue during exercise. Med Sci
tionally different types of cat medial gastroc- Sports Exerc. 1997;29(1):4557.
nemius alpha-motoneurons. J Comp Neurol. Derr RF, Zieve L. Adenylate energy charge:Relation
1993;330(3):439454. to guanylate energy charge and the adenylate
Brmmer V, Schneider S, Abel T, Vogt T, Strder HK. kinase equilibrium constant. Biochem Biophys
Brain cortical activity is influenced by exer- Res Commun. 1972;49:13851390.
cise mode and intensity. Med Sci Sports Exerc. Dhar SS, Wong-Riley MT. Coupling of energy
2011;43(10):18631872. metabolism and synaptic transmission at the
Burnstock G, Krgel U, Abbracchio MP, Illes P. transcriptional level: role of nuclear respiratory
Purinergic signalling: From normal behaviour factor 1 in regulating both cytochrome c oxidase
310 Part III:Homeostatic Manipulators

and NMDA glutamate receptor subunit genes. hippocampus in vivo. Am J Physiol Regul Integr
J Neurosci. 2009;29(2):483492. Comp Physiol. 2001;280:R639R645.
Dishman RK. Brain monoamines, exercise, and Gonzalez-Alonso J, Dalsgaard MK, Osada T, et al.
behavioral stress: Animal models. Med Sci Brain and central haemodynamics and oxy-
Sports Exerc. 1997;29:6374. genation during maximal exercise in humans. J
Dishman RK, Renner KJ, Youngstedt SD, et al. Physiol. 2004;557:331342.
Activity wheel running reduces escape latency Goodwin GM, McClosky DI, Mitchell JH.
and alters brain monoamine levels after foot- Cardiovascular and respiratory responses to
shock. Brain Res Bull. 1997;42:399406. changes in central command during isometric
Dishman RK, Renner KJ, White-Welkley JE, Burke KA, exercise at constant muscle tension. J Physiol.
Bunnell BN. Treadmill exercise training aug- 1972;226:173190.
ments brain norepinephrine response to Graham, TE. Caffeine and exercise. Sports Med.
familiar and novel stress. Brain Res Bull. 2001;31(11):785807.
2000;52(5):337342. Greenwood BN, Foley TE, Burhans DJ, Maier SF,
Driver HS, Taylor SR. Exercise and sleep. Sleep Med Fleshner M. The consequences of uncontrollable
Rev. 2000;4:387402. stress are sensitive to the duration of prior wheel
Dustman RE, Emmerson RY, Ruhling RO, et al. running, Brain Res. 2005;1033:164178.
Age and fitness effects on EEG, ERPs, visual Greenwood BN, Foley TE, Day HEW, etal. Freewheel
sensitivity, and cognition. Neurobiol. Aging. running prevents learned helplessness/behav-
1990;11:193200. ioral depression:role of dorsal raphe serotoner-
Dworak M, Diel P, Voss S, Hollmann W, Strder HK. gic neurons. J Neurosci. 2003;23:28892898.
Intense exercise increases adenosine concentra- Gruetter R. Glycogen:the forgotten cerebral energy
tions in rat brain: Implications for a homeostatic store. J Neurosci Res. 2003;74(2):179183.
sleep drive. Neuroscience. 2007;150:789795. Haas H, Selbach O. Functions of neuronal adenosine
Dworak M, Wiater A, Alfer D, Stephan E, Hollmann W, receptors. Arch Pharmacol. 2000;362:375381.
Strder HK. Increased slow wave sleep and Hanakawa T, Immisch I, Toma K, Dimyan MA, Van
reduced stage 2 sleep in children depend- Gelderen P, Hallett M. Functional properties of
ing on exercise intensity. Sleep Med. 2008 brain areas associated with motor execution and
Mar;9(3):266272. imagery. J Neurophysiol. 2003;89:9891002.
Farmer J, Zhao X, van Praag H, Wodtke K, Gage FH, Harris DA. Cellular ATP. In:Bittar EE, ed. Principles
Christie BR. Effects of voluntary exercise on syn- of medical biology. Vol 4.Elsevier, 1996:147.
aptic plasticity and gene expression in the den- Henriksson J. Effect of exercise on amino acid con-
tate gyrus of adult male Sprague-Dawley rats in centrations in skeletal muscle and plasma. J Exp
vivo. Neuroscience. 2004;124(1):7179. Biol. 1991;160:149165.
Fields RD, Stevens B. ATP:An extracellular signal- Herholz K, Buskies W, Rist M, Pawlik G, Hollmann W,
ing molecule between neurons and glia. Trends Heiss WD. Regional cerebral blood flow in man at
Neurosci. 2000;23:625633. rest and during exercise. J Neurol. 1987;234(1):913.
Fix JD. Basal ganglia and the striatal motor system. Hertz L, Dienel GA. Lactate transport and trans-
In: Fix, JD, Neuroanatomy. 4th ed. Baltimore: porters: general principles and functional roles
Wulters Kluwer/Lippincott Williams & Wilkins; in brain cells. J Neurosci Res. 2005;79(12):1118.
2008:274281. Hoyer S. Brain glucose and energy metabo-
Fredholm BB, Battig K, Holmen J, Nehlig A, Zvartau EE. lism during normal aging. Aging (Milano).
Actions of caffeine in the brain with special ref- 1990;2(3):245258.
erence to factors that contribute to its widespread Ide K, Horn A, Secher NH. Cerebral metabolic
use. Pharmacol Rev. 1999;51:83133. response to submaximal exercise. J Appl Physiol.
Freed CR, Yamamoto BK. Regional brain dopamine 1999;87:16041608.
metabolism: A marker for the speed, direc- Ide K, Schmalbruch IK, Quistorff B, Horn A, Secher
tion, and posture of moving animals. Science. NH. Lactate, glucose and O2 uptake in human
1985;229(4708):6265. brain during recovery from maximal exercise. J
Gerald MC. Effects of (+)-amphetamine on the Physiol. 2000;522:159164.
treadmill endurance performance of rats. Ide K, Secher NH. Cerebral blood flow and
Neuropharmacology. 1978;17(9):703704. metabolism during exercise. Prog Neurobiol.
Gervitz LM, Lutherer LO, Davies DG, Pirch JH, Fowler 2000;61:397414.
JC. Adenosine induces initial hypoxic-ischemic Jackman MR, Wendling P, Friars D, Graham TE.
depression of synaptic transmission in the rat Metabolic, catecholamine, and endurance
Exercise 311

responses to caffeine during intense exercise. J Meeusen R, Watson P, Hasegawa H, Roelands B,


Appl Physiol. 1996;81:16581663. Piacentini MF. Central fatigue: The serotonin
Kamijo K, Nishihira Y, Hatta, A, et al. Changes in hypothesis and beyond. J Sports Sci. 2006
arousal level by differential exercise intensity. Jul;24(7):773782.
Clin Neurophysiol. 2004;115:26932698. Molteni R, Zheng J-Q, Ying Z, Gmez-Pinilla F,
Karp A, Paillard-Borg S, Wang HX, Silverstein M, Twiss J. Voluntary exercise increases axonal
Winblad B, Fratiglioni L. Mental, physical, and regeneration from sensory neurons. Proc Natl
social components in leisure activities equally Acad Sci USA. 2004;101(22):8473.
contribute to decrease dementia risk. Dement Nielsen B, Hgldig T, Bidstrup F, Gonzalez-Alonso J,
Geriat Cogn Disord. 2006;21:6573. Christoffersen GRJ. Brain activity and fatigue
Kayser B. Exercise starts and ends in the brain. Eur J during prolonged exercise in the heat. Pflugers
Appl Physiol. 2003;90:411419. Arch. 2001;442:4148.
Kemppainen J, Aalto S, Fujimoto T, et al. High Nielsen HB, Bredmose PB, Strmstad M, Volianitis
intensity exercise decreases global brain glucose S, Quistorff B, Secher NH. Bicarbonate attenu-
uptake in humans. J Physiol. 2005;568:323332. ates arterial desaturation during maximal exer-
Kovacs EMR, Stegen JHC, Brouns F. Effect of caf- cise in humans. J Appl Physiol. 2002;93:724731.
feinated drinks on substrate metabolism, caf- Nielsen HB, Madsen P, Svendsen LB, Roach RC,
feine excretion, and performance. J Appl Physiol. Secher NH. The influence of PaCO2, pH and
1998;85:709711. SaCO2 on maximal oxygen uptake. Acta Physiol
Lardon MT, Polich J. EEG changes from long-term Scand. 1988;164:8997.
physical exercise. Biol Psychol. 1996;44:1930. Nybo L, Nielsen B. Perceived exertion is associated
Latini S, Pedata F. Adenosine in the central with an altered brain activity during exercise
nervous system: Release mechanisms and with progressive hyperthermia J Appl Physiol.
extracellular concentrations. J Neurochem. 2001;91:20172023.
2001;79:463484. Nybo L, Rasmussen P. Inadequate oxygen delivery
Leung L, Tong K, Zhang S, Zeng X, Zhang K, Zheng X. to the brain as a factor influencing fatigue dur-
Neurochemical effects of exercise and neuro- ing strenuous exercise. Rev Sports Exerc Sci.
muscular electrical stimulation on brain after 2007;35:110118.
stroke: A microdialysis study using rat model. OConnor PJ, Youngstedt SD. Influence of exer-
Neurosci Lett. 2006;397(12):135139. cise on human sleep. Exerc Sport Sci Rev.
Madsen PL, Secher NH. Near-infrared oximetry of 1995;23:105134.
the brain. Prog Neurobiol. 1999;58:541560. Okada M, Kiryu K, Kawata Y, etal. Determination
Madsen PL, Sperling BK, Warming T, etal. Middle of the effects of caffeine and carbamazepine on
cerebral artery blood velocity and cerebral blood striatal dopamine release by in vivo microdialy-
flow and O2 uptake during dynamic exercise. J sis. Eur J Pharmacol. 1997;324:181188.
Appl Physiol. 1993;74:245250. Orgogozo JM, Larsen B. Activation of the supple-
Magistretti PJ, Pellerin L. Astrocytes Couple mentary motor area during voluntary movement
Synaptic Activity to Glucose Utilization in the in man suggests it works as a supramotor area.
Brain. News Physiol Sci. 1999;14:177182. Science. 1979;206(4420):847850.
Magistretti PJ, Pellerin L, Rothman DL, Owen O, Morgan A, Kemp H, Sullivan J, Herrera
Shulman RG. Energy on demand. Science. M, Cahill GJ. Brain metabolism during fasting.
1999;283(5401):496497. J Clin Invest. 1967;46:15891595.
McKenna JT, Tartar JL, Ward CP, et al. Sleep frag- Pascual O, Casper KB, Kubera C, et al. Astrocytic
mentation elevates behavioral, electrographic purinergic signaling coordinates synaptic net-
and neurochemical measures of sleepiness. works. Science. 2005;310:113116.
Neuroscience. 2007;146:14621473. Pellerin L, Pellegri G, Martin JL, Magistretti PJ.
Mechau D, Mucke S, Weiss M, Liesen H. Effect of Expression of monocarboxylate transporter
increasing running velocity on electroencepha- mRNAs in mouse brain: support for a distinct
logram in a field test. Eur J Appl Physiol Occup role of lactate as an energy substrate for the neo-
Physiol. 1998;78:340345. natal vs. adult brain. Proc Natl Acad Sci U S A.
Mecklinger A, Kramer AF, Strayer DL. Eventrelated 1998;95(7):39903995.
potentials and EEG components in a seman- Peters A, Schweiger U, Pellerin L, et al. The self-
tic memory search task. Psychophysiology. ish brain: Competition for energy resources.
1992;29:104119. Neurosci Biobehav Rev. 2004;28:143180.
Meeusen R, De Meirleir KD. Exercise and brain neu- Petruzzello SJ, Ekkekakis P, Hall EE. Physical activ-
rotransmission. Sports Med. 1995;20(3):160188. ity and affect: EEG studies. In: Acevedo EO,
312 Part III:Homeostatic Manipulators

Ekkekakis P, eds. Psychobiology of Exercise Spector SA, Jackman MR, Sabounjian LA, Sakkas C,
and Sport. Champaign, IL: Human Kinetics; Landers DM, Willis WT. Effects of choline sup-
2006:111128. plementation on fatigue in trained cyclists. Med
Phillis JW, Ren J, O'Regan MH. Studies on the effects Sci Sports Exerc. 1995;27:668673.
of lactate transport inhibition, pyruvate, glucose Stocco A, Lebiere C, Anderson, JR. Conditional
and glutamine on amino acid, lactate and glu- routing of information to the cortex:Amodel of
cose release from the ischemic rat cerebral cor- the basal ganglias role in cognitive coordination.
tex. J Neurochem. 2001;76(1):247257. Psychol Rev. 2010;117(2):541574.
Pierre K, Pellerin L. Monocarboxylate transporters in Strder HK, Hollmann W, Platen P, Wstmann R,
the central nervous system:distribution, regula- Ferrauti A, Weber K. Effect of exercise inten-
tion and function. J Neurochem. 2005;94(1):114. sity on free tryptophan to branched-chain
Rainnie DG, Grunze HC, McCarley RW, Greene amino acids ratio and plasma prolactin dur-
RW. Adenosine inhibition of mesopontine cho- ing endurance exercise. Can J Appl Physiol.
linergic neurons: Implications for EEG arousal. 1997;22:280291.
Science. 1994;263:689692. Surmeier DJ. Dopamine and working memory
Rogers HB, Schroeder T, Secher NH, Mitchell JH. mechanisms in prefrontal cortex. J Physiol.
Cerebral blood flow during static exercise in 2007;885(Pt 3):581.
man. J Appl Physiol. 1990;68:23582361. Sutoo D, Akiyama K. Regulation of brain function
Romer LM, Lovering AT, Haverkamp HC, Pegelow by exercise. Neurobiol Dis. 2003 Jun;13(1):14.
DF, Dempsey JA. Effect of inspiratory mus- Thompson FM, Atkinson DE. Response of nucle-
cle work on peripheral fatigue of locomo- oside diphosphate kinase to the adenylate
tor muscles in healthy humans. J Physiol. energy charge. Biochem Biophys Res Commun.
2006;571:425439. 1971;45:15811585.
Rose EM, Koo JC, Antflick JE, Ahmed SM, Uysal N, Tugyan K, Kayatekin BM, et al. The
Angers S, Hampson DR. Glutamate trans- effects of regular aerobic exercise in adoles-
porter coupling to Na, K-ATPase. J Neurosci. cent period on hippocampal neuron density,
2009;29(25):81438155. apoptosis and spatial memory. Neurosci Lett.
Ruscheweyh R, Willemer C, Krger K, etal. Physical 2005;383:241245.
activity and memory functions:An interventional van Hall G, Strmstad M, Rasmussen P, Jans O, Zaar
study. Neurobiol Aging. 2011;32(7):13041319. M, Gam C, Quistorff B, Secher NH, Nielsen HB.
Salmon P. Effects of physical exercise on anxiety, Blood lactate is an important energy source for
depression, and sensitivity to stress:Aunifying the human brain. J Cereb Blood Flow Metab.
theory. Clin Psychol Rev. 2001;Feb; 21(1):3361. 2009;29(6):11211129.
Sato K, Moriyama M, Sadamoto T. Influence of cen- Van Lieshout JJ, Pott F, Madsen PL, Van Goudoever
tral command on cerebral blood flow at the onset J, Secher NH. Muscle tension during stand-
of exercise. Exp Physiol. 2009a;94:11391146. ing:Effect on cerebral artery blood velocity and
Sato K, Sadamoto T, Ueda-Sasahara C, etal. Central oxygenation. Stroke. 2001;32:15461551.
command and the increase in middle cerebral Van Lieshout JJ, Wieling W, Karemaker JM, Secher
artery blood velocity during static arm exercise NH. Syncope, cerebral perfusion, and oxygen-
in women. Exp Physiol. 2009b;94:11321138. ation. J Appl Physiol. 2003;94:833848.
Schneider S, Brmmer V, Abel T, Askew CD, Strder Vaynman S, Gomez-Pinilla F. Revenge of the
HK. Changes in brain cortical activity measured sit:How lifestyle impacts neuronal and cogni-
by EEG are related to individual exercise prefer- tive health though molecular systems that inter-
ences. Physiol Behav. 2009;98(4):447452. face energy metabolism with neuronal plasticity.
Schurr A. Lactate: The ultimate cerebral oxidative J Neurosci Res. 2006;84:699715.
energy substrate. J Cereb Blood Flow Metab. Vuori I, Urponen H, Hasan J, Partinen M.
2006;26:142152. Epidemiology of exercise effects on sleep. Acta
Secher NH, Seifert T, Van Lieshout JJ. Cerebral blood Physiol Scand. 1988;144(574):1421.
flow and metabolism during exercise: impli- Vissing J, Andersen M, Diemer NH. Exercise-induced
cations for fatigue. J Appl Physiol (1985). changes in local cerebral glucose utilization
2008;104(1):306314. in the rat. J Cereb Blood Flow Metab. 1996
Shapiro CM, Bortz R, Mitchell D. Slow-wave Jul;16(4):729736.
sleep: A recovery period after exercise. Science. Williamson JW, McColl R, Mathews D, Ginsburg M,
1981;214:12531254. Mitchell JH (1999) Activation of the insular
Exercise 313

cortex is affected by the intensity of exercise. J physical activity vs. imipramine: Neonatal clo-
Appl Physiol. 87:12131219. mipramine. Psychobiology. 2000;28:540549.
Wilson RS, Mendes De Leon CF, Barnes LL, et al. Young SN. How to increase serotonin in the human
Participation in cognitively stimulating activi- brain without drugs. J Psychiatry Neurosci. Nov
ties and risk of incident Alzheimer disease. 2007;32(6):394399.
JAMA. 2002;287:742748. Youngstedt SD, OConnor PJ, Dishman RK. The
Yoo HS, Tackett RL, Crabbe JB, Bunnell BN, effect of acute exercise on sleep: A quantitative
Dishman RK. Antidepressant-like effects of synthesis. Sleep. 1997;20(3):203214.
18
Sleep
K R IST INA SIMEONE, CHA Z JOHNSON, K AELI SA MSON,
H A R R I S O N R O U N D T R E E , T I M S I M E O N E , A N D L E I L A TA R O K H

INTRODUCTION and affective disorders. Indeed, insufficient sleep


Proper sleep hygiene can exert beneficial effects and sleep disorders can manifest as comorbid
on several physiological systems. Here we focus conditions of several neurological disorders and
on how sleep influences the central processing affective disorders, including anxiety disorders,
of cognition, stress, behavior, body weight, brain depression, epilepsy, attention deficit hyperactiv-
energy (adenosine triphosphate [ATP]) and waste ity disorder (ADHD), autism, Alzheimers dis-
removal, and immune responses. Research sug- ease, and Parkinsons disease. Third, considering
gests that the neurobiology associated with suf- the effects of insufficient sleep, sleep disorders
ficient sleep interacts with these processes in a can simultaneously be comorbid with cognitive
homeostatic manner to promote optimal day- impairments and/or affective disorders. Finally,
time functioning. sleep neurobiology can be targets of pharmaco-
Sufficient sleep can be achieved with 7 to 9 logical treatments. Tiredness, drowsiness or trou-
hours for adults, 8.5 to 9.5 hours for teenagers, ble sleeping are side effects of many drugs used to
10 to 11 hours for school-aged children, and treat anxiety and depression (including aripipra-
12 to 14 hours for preschool children (Bonnet and zole, serotonin reuptake inhibitors [SSRIs], and
Arand, 2003; Institute of Medicine Committee serotonin norepinephrine reuptake inhibitors
on Sleep Medicine and Research, 2006; National [SNRIs]), epilepsy (ethosuximide, gabapentin,
Sleep Foundation, 2013). Decades of research phenytoin, levetiracetam, zonisamide), autism
have taught us that when the brain is deprived (aripiprazole, risperidal), Alzheimers disease
of sleep, neurobiological instabilities can result. (donepezil, galantamine, rivastigmine), and
Depending on the length of deprivation, the Parkinsons disease (carbidopa with levodopa).
behavioral readouts of these instabilities can In this chapter we first describe the neuro-
be quantified during the deprivation itself and biology of sleep architecture. We discuss the
subsequent days. These behavioral instabili- intersections of neurobiology, sleep, and phar-
ties include impairments of cognitive process- macology in high-incidence disorders, including
ing, reaction time, and vigilance; reduction of anxiety disorders (1:6), depression (1:10), epi-
stress thresholds; promotion of negative affec- lepsy (1:26), ADHD (1:9 children), autism (1:68
tive behaviors (including the inability to focus or children), Alzheimers disease (1:8 over the age of
concentrate, irritability, anxiety, aggression, and 65 and 1:2 over the age of 85), and Parkinsons
depressive-like behaviors); changes in appetite; disease (1:313) (Alzheimers Association, 2012;
and suboptimal immune responses. Institute of Medicine Committee on the Public
Unfortunately, insufficient sleep is a grow- Health Dimensions of the Epilepsies, Board on
ing public health concern for multiple reasons Health Sciences Policy, 2012; National Institute of
(Institute of Medicine Committee on Sleep Mental Health; Parkinsons Disease Foundation,
Medicine and Research, 2006). First, the increase 2014). Next we discuss how intentionally struc-
in societal pace, accessibility of artificial light, turing sleep as a preventative measure or as an
and repeated prolonged periods of wakefulness adjuvant restorative therapy may improve cogni-
can result in chronic insufficient sleep for adults tion and/or behavior in normal individuals and
and children. Second, the neuropeptides and lessen the degree of cognitive/behavioral comor-
neurotransmitters involved in sleep-wake cycles bid afflictions in individuals with neurological or
can be dysregulated in neurological disorders affective disorders. We conclude by highlighting
Sleep 315

how sleep may provide restorative influence deprivation); thus results are discussed in the
on brain regulators of body weight, brain ATP context of the protocol employed.
levels and damage/waste removal systems, and
neuro-immune responses. SLEEP ARCHITECTURE
This chapter is structured to highlight a wide AND NEUROBIOLOGY
array of studies and is not intended to be com- In this section, we discuss the electroencepha-
prehensive. It is important to note that care was lography (EEG), neurobiology and pharmacol-
taken to determine the order in which topics are ogy of non-rapid eye movement sleep (NREMS),
presented based on their interrelationships and rapid eye movement sleep (REMS), wake, and
sections are not concluded with causal specu- wake-sleep transition.
lation. In addition, many sleep studies employ Sleep architecture describes the timing and
sleep deprivation protocols, which vary in terms the predominance of different brain rhythms
of duration (full or partial deprivation), timing during NREMS (stages 1, 2, and 3/4) and REMS
(early or late), and recurrence (acute or chronic (Figure 18.1, left panel). Each NREMSREMS

Adults Sleep Architecture Neurobiology


REM

NREM (80% of sleep) BF VLPO MLPO LH TMN


ACH/A/GABA GABA GABA OX/ACH/A/GABA HA
- S1: Theta, less alpha NERM
- S2: Spindles, k- ViPAG/ SN/VTA/VPAG R LC LDT PPT
complexes SLD
SLEEP (7 hr)

LPT DA 5-HT NE ACH(R)


- S3: Delta, SWA
Motor
neurons
Thalamus
REM (20% of sleep)
- Alpha BF VLPO MLPO LH TMN
- Theta ACH/A/GABA GABA GABA OX/MCH/A/GABA HA
REM
24 hr period

LDT PPT
S1
S2
S3

ViPAG/ SN/VTA/VPAG R LC
SLD ACh(R)
LPT DA 5-HT NE
VM Spinal Motor
GABA/glycine interneurons neurons

WAKE Thalamus
- Beta
BF VLPO MLPO LH TMN
- Alpha ACH/A/GABA GABA GABA OX/MCH/GABA HA
WAKE (17 hr)

- Theta Wake
- Gamma ViPAG/ SN/VTA/VPAG R LC LDT PPT
STD ACh (R)
LPT DA 5-HT NE

Motor
neurons
Frequencies of brain rhythms
- Very slow - Beta (914 Hz)
oscillation (0.11 Hz) - Sigma (1116 Hz)
- Delta SWA (1.24.5 Hz) - Beta (1630 Hz)
- Theta (58 Hz) - Gamma (40100 Hz)

FIGURE 18.1: Brain rhythms and the corresponding neurobiology during each state of vigi-
lance. Left: The schematic depicts a 24-hour period divided into time spent awake and asleep. During sleep,
the non-rapid eye movement sleep (NREM) stages (S) 13 (blue) and the rapid eye movement (REM; purple)
transitions and cycles are depicted. Each state is magnified, and the specific brain rhythms are identified. The
corresponding frequencies that comprise each rhythm are below (grey). Right:Aligned with each state of vigi-
lance is a simplified schematic of some of the known underlying neurobiology during NREM sleep (blue), REM
sleep (purple), and awake stages (green). The boldness of the font indicates the relative changes in different neu-
ronal populations:bold indicates strong signaling; normal font indicates mild/moderate signaling; lighter font in
NREM sleep indicates reduced activity when compared to awake; lighter font in REM indicates reduced activity
when compared to NREM sleep; and light grey indicates minimal or no signaling. We highlighted the activity of
the orexin (OX), adenosine (A), histamine (HA), serotonin (5-HT), dopamine (DA), norepinephrine (NE), ace-
tylcholine (ACh), and GABA systems. BF=basal forebrain, LC=locus coeruleus, LDT=lateral dorsal tegmental
nucleus, LH = lateral hypothalamus, MLPO = medial lateral preoptic area, PPT = pedunculopontine nucleus,
R=raphe nuclei, SLD=sublateral dorsal nucleus, SN=substantia nigra, TMN=tuberomammillary nucleus,
VLPO=ventral lateral preoptic area, VPAG=ventral periaqueductal grey, VTA=ventral tegmentalarea.
316 Part III:Homeostatic Manipulators

cycle transitions from NREMS stages 1, 2, 3/4, and additional projections from the ventrolateral
2, 1 to REMS and is approximately 90 to 120 preoptic nucleus (VLPO) are necessary for main-
minutes in duration in adults. NREMSREMS taining sleep (Chou et al., 2002; Gallopin et al.,
cycles occur several times during sleep, and often 2000; Gaus etal., 2002; Gong etal., 2004; Hassani
waking in the morning occurs following REMS. etal., 2009; Hassani etal., 2010; Lu etal., 2000;
REMS durations range from approximately 10 Manns etal., 2000; McGinty and Sterman, 1968;
minutes to 1 hour and increase with subsequent Saper etal., 2005a; Saper etal., 2010a; Sherin etal.,
episodes. Thus, overall there is a higher amount 1996; Sherin et al., 1998; Suntsova et al., 2002;
of NREMS during early sleep, and a higher inci- Szymusiak et al., 1998; Takahashi et al., 2009).
dence of REMS during later sleep. Analyses of During NREMS, additional GABAergic neuro-
sleep architecture can be used as a tool to identify transmission from the basal forebrain and lateral
problems with specific types or phases ofsleep. hypothalamus (LH) and melanin-concentrating
The neurobiological landscape responsible hormone from the LH contribute to the inhibi-
for promoting sleep and wake states employ the tion of cortical and subcortical activity (Alam
same peptides and transmitters that (i)are dys- etal., 2002; Bittencourt etal., 1992; Hassani etal.,
regulated in neurological disorders and affective 2010; Kilduff and de Lecea, 2001; Koyama etal.,
disorders and (ii) are targets of pharmacothera- 2003; Saito etal., 2001; Verret etal.,2003).
pies. These include gamma-aminobutyric acid
(GABA), serotonin, norepinephrine, acetyl- Pharmacology
choline, adenosine, orexin, histamine, and GABA. Drugs that enhance GABAergic signal-
dopamine. Figure 18.1 (right panel) depicts the ing, such as benzodiazepines (e.g., clonazepam),
relative changes in activity of some of these sys- are used to treat anxiety, insomnia, agitation, and
tems during NREMS and REMS and waking. In seizures in epilepsy and are used as sedatives and
this section we briefly describe the sleep archi- hypnotics. Benzodiazepines are associated with
tecture and how relative levels of these molecules increasing NREMS activity and sleep duration.
play a critical role in the homeostatic balance of However, with sleep-promoting effects on neuro-
NREMS and REMS and wake states (see Figure biology, a potential side effect of benzodiazepines
18.1 throughout this section). is prolonged daytime drowsiness. Other seda-
tives/hypnotics that also increase GABA inhibi-
NREMS tion to promote sleep include zolpidem, zaleplon,
EEG and Neurobiology and eszopiclone.
During stage 1 sleep, there is a reduction in alpha
waves and an increase in theta power (Figure REMS
18.1). During stage 2 sleep, which occupies 45% EEG and Neurobiology
to 55% of our total sleep time, our body tempera- During REMS, there is a reduction of the over-
ture decreases, our heart rate slows, our muscle all GABAergic tone, spindles and slow waves.
tone relaxes, and sleep spindles (sigma 1116 Hz) Increased activation of acetylcholine and glu-
and K-complexes become apparent on the EEG. tamate projections to the thalamus and cortex
During stage 3, which accounts for 15% to 25% of induce phasic activation of the visual and limbic
our total sleep, we are in deep sleep and our brain circuits, neuronal network desychronization, and
rhythms oscillate at high amplitude (75 mV) low-amplitude theta and alpha rhythms (510
and low frequency (i.e., delta frequency range Hz) associated with REMS (Brown et al., 2012;
from 0.5 to 5 Hz also known as slow-wave activity Dang-Vu etal., 2010). Electromyogram is used to
(SWA)). Delta power is associated with a reduc- verify muscle atonia during REMS to distinguish
tion in heart rate and an increase in parasympa- this state from wakefulness.
thetic activity. The amount of delta power and
stage 3/4 sleep declines across the course of the Pharmacology
night and is thus a marker of sleep homeostasis. Acetylcholine. Acetylcholine is a key neurotrans-
During NREMS, GABAergic neuro- mitter during REMS. Increasing acetylcholine
transmission is responsible for inhibiting the levels at the synapse is associated with reducing
wake-promoting regions (discussed later) and the latency to REMS onset and increasing REMS
inducing SWA (Chou etal., 2002; Gallopin etal., duration. One treatment strategy for Alzheimers
2000). GABA projections from the mediolateral disease is to increase levels of acetylcholine at the
preoptic nucleus are necessary for inducing sleep, synapse by inhibiting the enzyme responsible for
Sleep 317

degrading acetylcholine (acetylcholine esterase) tegmental nuclei). These regions then send pro-
with a reversible acetylcholine esterase (AChE) jections to glutamatergic thalamic and cortical
inhibitor, such as donepezil, galantamine, or structures to increase the cortical EEG desyn-
rivastigmine. Indeed, treatment with donepezil chronization and higher frequency rhythms (in
increases REMS in people with Alzheimers dis- the alpha, beta, theta, and gama power), muscle
ease (Moraes Wdos etal.,2006). tone, attention, and alertness.
GABA, serotonin, norepinephrine-enhancing
drugs. REMS can be reduced by drugs that Pharmacology
enhance GABA, serotonin, or norepineph- Alterations of each of the following wake-
rine (Boissard et al., 2002; Curtis et al., 1968; promoting systems, either by disorders or phar-
Fedirchuk and Dai, 2004; Fenik et al., 2004; macology, can negatively influencesleep.
Jelev et al., 2001; Kayama et al., 1992; Lai Orexin receptor blockers.An increase in sleep
et al., 2001; Leonard and Llinas, 1994; Morales fragmentation, or the number of awakenings dur-
et al., 1987; Morrison et al., 2003; Perrier and ing sleep, is a common comorbid sleep-disorder
Delgado-Lezama, 2005; Steriade et al., 1990). symptom in mental retardation, anxiety and
As described, GABA-activating drugs promote stress disorders, depression, Alzheimers,
NREMS and thus reduce time spent in REMS. Parkinsons, and epilepsy (Lee-Chiong, 2008).
Serotonin and norepinephrine are active neu- Orexin-expressing neurons in the LH are a wake
rotransmitters during wake and NREMS and trigger, and dysfunction of the orexin system is
are virtually absent during REMS. Drugs that associated with sleep fragmentation, increased
increase levels of serotonin and/or norepineph- awakenings, narcolepsy, restless leg disorder,
rine, specifically at the synapse, include SSRIs, and epilepsy (Sakurai etal., 2010). Novel strate-
SNRIs, and monoamine oxidase inhibitors. These gies that suppress orexin activity via dual orexin
drug classes are used to treat depression and receptor antagonism improved sleep quality in
ADHD and can result in an increase in NREMS normal mice in clinical trials and in a mouse
rhythms, a reduction in REMS incidence, and model of epilepsy with a comorbid sleep disorder
a delay in the overall drive to induce REMS (Roundtree etal., submitted). This class of drugs
(Wilson and Argyropoulos, 2005). Due to the has a faster half-life than other sedatives, and
fact that serotonin is also involved in promoting thus there is minimal daytime drowsiness.
wakefulness, insomnia is a side effect ofSSRIs. Dopamine, serotonin and norepinephrine
inhibiting agents. Atypical antipsychotics, such
Wake as risperidone, are used to treat schizophrenia
Sleep-disorder symptoms, such as fragmenta- and bipolar disorder and to reduce self-harming,
tion or inappropriate awakenings during sleep, aggressive, and tantrum behaviors in children
involve the neurobiology underlying wakeful- with autism. These compounds antagonize
ness. Symptoms such as longer sleep latency wake-promoting neurotransmission by blocking
involve the neurobiology underlying the drive to dopamine receptors and exerting antiserotonin-
fallsleep. ergic, antinorepinephrinergic, and antihistamin-
ergic activity. Risperidone has been reported to
EEG and Neurobiology improve sleep in autistic children with irritability
Projections from the master circadian pace- and related behaviors (Kent etal.,2013).
maker, the suprachiasmatic nucleus, activate In people with depression, treatment with
orexin (also referred to as hypocretin) neurons olanzapine, another atypical antispyschotic,
in the LH to promote wakefulness. Orexin pro- improved sleep efficiency, total sleep time, and
jections to the ascending reticular activating sleep latency (Lazowski etal., 2014). However, by
system excite several nuclei to increase arousal, blocking wake-promoting neurotransmission,
attention, and muscle tone (Sakurai, 2007; these compounds have fatigue and sedative side
Szymusiak and McGinty, 2008). These efferent effects. Reduced levels of dopamine and seroto-
targets include dopamine neurons (in the sub- nin are associated also with Parkinsons disease
stantia nigra), serotonin neurons (in the dorsal and depression, respectively, both disorders with
raphe nuclei), norepinephrine neurons (in the comorbid daytime drowsiness.
locus coeruleus), histamine neurons (in the tub- Dopamine, serotonin and norepineph-
eromammillary nucleus), and acetylcholine neu- rine enhancing agents. Stimulants including
rons (in the pedunculopontine and laterodorsal amphetamines and cocaine increase synaptic
318 Part III:Homeostatic Manipulators

concentrations of monoamines, primarily dopa- in the tuberomammillary nucleus and basal fore-
mine and norepinephrine (but also serotonin and brain to increase VLPO GABAergic signals to
histamine), via multiple mechanisms of action. enhance process S or the drive to sleep (Basheer
Modafinil, used for the treatment of narcolepsy, etal., 2004; Benington etal., 1995; Huang etal.,
promotes wakefulness primarily by increasing 2007; Huston et al., 1996; Porkka-Heiskanen
dopamine and histamine levels. et al., 1997a; Radulovacki et al., 1984). Caffeine
Antihistamines. Histamine promotes and theophylline, found in tea, are stimulants
wakefulness and the ability to focus. First gen- that antagonize A1R and thus interfere with
eration antihistamines, such as diphenhy- adenosines ability to promotesleep.
dramine (an active ingredient in more than 40 Melatonin and a receptor agonist. The drug
over-the-counter allergy and sleep-aid prod- ramelteon is a selective melatonin receptor ago-
ucts), are able to cross the blood-brain barrier nist that decreases the latency to NREMS (or sleep
(BBB). These antihistamines can promote sleep onset) and increases NREMS duration (Fisher
at night but can also cause daytime drowsiness. et al., 2008; Spadoni et al., 2011). Activation
Second-generation antihistamines, such as lorat- of melatonin receptors in the suprachiasmatic
adine, have minimal BBB permeability and thus nucleus is thought to shift the timing of the cir-
have minimal effects on sleep architecture. cadian system promoting sleep onset directly
and reinforcing regular evening sleepiness and
Wake-Sleep Transition sleep onset (Neubauer, 2008). In preclinical stud-
Two-Process Model ofSleep ies, exogenous melatonin had similar effects as
Regulation ramelteon; however, unfavorable pharmacoki-
The factors that determine when we fall asleep netics has led to variable results in human clini-
and establish our sleep homeostasis are typically cal trials. It does appear that prolonged-release
described with a two-process model of sleep regu- formulations of melatonin do decrease sleep
lation (Daan etal., 1984; Tarokh and Achermann, onset and increase sleep duration (Fisher et al.,
2013). The first is the internal circadian process 2008; Spadoni etal., 2011; Srinivasan etal., 2011).
(process C). Process C determines the ideal In addition, melatonin has potent antioxidant
timing of a sleep episode based on internal fac- activities that may contribute to additional effects
tors and has a 24-hour rhythm governed by the during sleep (discussed later).
suprachiasmatic nucleus. The second is the sleep
homeostatic process (process S). Process S is Pathology
dependent on the amount of time elapsed since Reduction of adenosine tone is associated with
the last sleep episode and is an internal driver to brain pathology that expresses reactive astro-
ensure sleep is attained to re-establish our sleep cytes or astrogliosis (Boison, 2006, 2008).
homeostasis. The actual timing and consolida- Astrogliosis is associated with an increase of
tion of wake and sleep (i.e., when we fall asleep) adenosine kinase, the enzyme responsible for
are regulated by the intersection of process S and converting adenosine into adenosine monophos-
processC. phate. Astrogliosis can occur in neurological
disorders that are associated with sleep comor-
Pharmacology bidities, including epilepsy, Alzheimers disease,
Recent studies suggest that adenosine (which and Parkinsons disease, however, reported loca-
increases with sleep deprivation) is a key neu- tions are typically in non-sleep regulating brain
robiological somnogen that drives process regions. One recent study identified astroglio-
S and can be used as a marker of sleep pres- sis in the sleep-regulating LH of epileptic mice
sure (Porkka-Heiskanen et al., 1997a; (Roundtree etal., submitted).
Porkka-Heiskanen and Kalinchuk, 2011). Other
somnogens that may participate in process S P R E V E N TAT I V E
include melatonin and immune factors (dis- A N D R E S T O R AT I V E
cussed later). OPPORTUNITIES
Adenosine and adenosine receptor block- Proper hygiene to ensure sufficient sleep can
ers. Adenosine is a purine nucleoside somnogen exert preventative and restorative effects on
that acts on its adenosine type 1 receptor (A1R) to many physiological processes. Here we highlight
inhibit wake-promoting basal forebrain and LH the effects of sleep on the central processing of
activity (Liu and Gao, 2007). Adenosine also acts (i)cognitive processing, (ii) stress responses and
Sleep 319

behavior, (iii) brain regulators of body weight, processing of extensive abstract concepts, school
(iv) ATP production and damage/waste removal, curricula, relationships, emotions and behav-
and (v)neuro-immune responses. iors. During development, the brain experiences
approximately 2 to 5 more hours of sleep each
How Does Sleep Influence night (dependent on age), when compared to
Cognition? adults. Chronic partial sleep deprivation in chil-
Cognition andSleep dren and young adults is associated with poorer
Successful processing and consolidation of cognitive performance. In young adults, reduced
different types of memories and next-day SWA is associated with reduced sensory motor
decision-making are strengthened when suf- and cognitive procedural memories (Limoges
ficient sleep is attained. Neuroimaging signals et al., 2013). Further, child-reported sleep prob-
indicate strong activation of the hippocampus lems are a predictor of child-and teacher-reported
and striatum during NREMS SWA. This activity academic problems and teacher-reported inat-
is hypothesized to reflect a replay of coordinated tentiveness (Becker,2014).
hippocampal-striatal activity and contrib-
ute to the selective strengthening of memories Disorders, Cognition, andSleep
(Dang-Vu etal., 2010). When early sleep remains Cognitive impairments occur with Alzheimers
intact, recall of previously learned declarative dementia and can be a comorbid condition of
tasks is enhanced, suggesting that NREMS SWA other disorders and diseases, including autism,
facilitates declarative memory consolidation pro- ADHD, depression, epilepsy, and Parkinsons dis-
cesses (Marshall et al., 2006; Plihal and Born, ease. Sleep disturbances among these populations
1997). When later sleep stages rich in REMS correlate with the degree of cognitive impair-
remain intact, recall of tasks requiring proce- ment. Poor sleep is an indicator of impaired
dural skills is enhanced (Walker etal.,2002). performance on selective attention, declarative
The cognitive processes that are impaired fol- memory, sensory-motor abilitiy, and procedural
lowing different severities of sleep deprivation memories in autistic young adults (Limoges etal.,
depend on the extent of regional or global decline 2013). Likewise, there is a positive correlation
in alertness and attention and whether these pro- between sleep disturbances and impaired cogni-
cesses are able to rely on compensatory support tive functioning in Parkinsons and Alzheimers
(Killgore, 2010). While execution of basic logical, patients, epilepsy patients, and children and
planning, and rule-based tasks is resilient to one adults with ADHD (Fisher etal., 2014; Kim etal.,
night of sleep deprivation, the ability to inno- 2014; Piperidou etal., 2008; Pistacchi etal.,2014).
vate, revise, or effectively handle distractions and
unexpected situations can be impaired (Harrison Sleep Therapy
and Horne, 2000). According to the Centers for Restoration of sleep quality is associated with
Disease Control and Prevention and the National improvements in cognitive performance in peo-
Sleep Foundation, there is a positive correlation ple with autism, epilepsy, ADHD, Parkinsons dis-
between the increased chronicity (or number of ease, and Alzheimers disease (Fisher etal., 2014;
consecutive days) of partial sleep deprivation and Kim etal., 2014; Limoges etal., 2013; Piperidou
impaired cognitive capacities as measured by et al., 2008; Pistacchi et al., 2014). Interestingly,
the number of automobile accidents and medi- increased theta power (of 48 Hz), which is asso-
cal mistakes made by first responders (Institute ciated with normal cognition during wakeful-
of Medicine Committee on Sleep Medicine and ness, and delta power during NREMS SWA is
Research, 2006; Machi et al., 2012; Waggoner associated with improved cognitive performance
etal., 2012). Further, the effect of chronic partial and better recall in people with Alzheimers
sleep deprivation (i.e., 14days of less than 6 hours disease (Hot etal., 2011). Increased theta power
per night) on cognitive function is dose depend- during stage 1 NREMS sleep is associated with
ent (Van Dongen etal.,2003). improved recall in patients with Parkinsons and
Alzheimers diseases (Kempf etal.,2009).
Implications forChildren
and Adolescents Pharmacology
During childhood and adolescence, brain growth Acetylcholine and histamine are wake-promoting
and the generation and pruning of new syn- molecules that are involved in the network pro-
apses occur simultaneously with the cognitive cessing of different types of learning, memory
320 Part III:Homeostatic Manipulators

consolidation, recall, and next-day thinking and hypothalamic-pituitary-adrenal (LHPA) axis


are also targets of neurological conditions and/or and sympathetic nervous system. Key LHPA
pharmacotherapies. molecules are corticotropin releasing hormone
Acetylcholine. Increasing synaptic acetylcho- (CRH) and glucocorticoids (cortisol in humans
line levels with AChE inhibitors enhance cogni- and corticosterone in rodents; Arborelius et al.,
tive abilities in people with Alzheimers disease. 1999; McEwen and Stellar, 1993). Stress sig-
Histamine. The daytime drowsiness side nals from cortical, hippocampal, and amygdala
effect of first-generation antihistamines may regions converge onto the hypothalamic para-
impair cognitive function and induce confusion. ventricular nucleus to elicit the secretion of CRH.
These side effects can be avoided by utilizing CRH acts on the anterior pituitary and ultimately
second-generation antihistamines, which have causes the release of glucocorticoids from the
minimal BBB permeability. adrenal glands. The response is terminated by
glucocorticoid negative feedback. Each transient
How Does Sleep Influence stress response is superimposed on its circadian
Stress and Behavior? rhythm. The circadian rhythm of CRH and corti-
As depicted in Figure 18.2, sleep efficiency, stress, sol expression peaks in mid-morning and reaches
and affective behaviors are cyclically interde- a nadir in the evening.
pendent, thus there are many subsections within Activation of the sympathetic nervous sys-
this section. Maintaining the categorical inde- tem increases release of norepinephrine and
pendence of each subsection was designed to epinephrine. Stressor-induced increases in heart
easily visualize their interrelationships while rate and/or blood pressure are mediated in part
avoiding causality. We begin by discussing the by the enhanced sympathetic activation and
interactions between stress responses and sleep the subsequent actions of norepinephrine and
efficiency followed by affective behaviors and epinephrine. In terms of circadian rhythms,
pharmacology. norepinephrine and epinephrine reach a nadir
approximately 1 hour after sleep onset (Irwin
Stress Responses etal.,1999).
Responses to psychological and physiologi- Implications for children. It is well known
cal stressors are mediated by the limbic- that the duration and severity of the LHPA
response is initially programmed early in life.
The expression ontogeny of CRH and its recep-
tors differ in a brain region specific manner
(Korosi and Baram, 2008), and the epigenetic
Insufficient programming of the LHPA is exquisitely sen-
sleep sitive to early life experiences (Fenoglio et al.,
2004; Fenoglio etal., 2005; Fenoglio etal., 2006a;
Fenoglio etal., 2006b; McEwen and Stellar, 1993).
D C A D Enriched or stressful early life experiences can
shift the programming pendulum to either an
B efficient response or a more severe and prolonged
Negative response, respectively (Biggs etal., 2011; Fenoglio
Stress
behaviors etal., 2006b; Ivy etal., 2008; Spaeth etal., 2013;
Watanabe etal.,2010).

FIGURE18.2: Cyclic interdependencies of insufficient Insufficient Sleep and


sleep, stress, and negative behaviors. (A) Insufficient Stress Responses
sleep can elevate stress molecules. (B) Elevated stress The glucocorticoid response is resilient to
responses are associated with negative behaviors acute and chronic sleep deprivation protocols
including anxiety, reduced threshold for aggression, in rodents and humans (see Figure 18.2; Fadda
irritability, and impulsiveness. (C) Insufficient sleep and Fratta, 1997; Kant etal., 1984; Novati etal.,
can promote negative behaviors including anxiety, 2008; Redwine et al., 2000). However, follow-
hyperactivity, distractibility, aggression, disinhibition, ing acute partial sleep deprivation (sleeping for
oppositional behavior, risk taking, irritability, and approximately 4 hours), increased sympathetic
impulsiveness. (D)Both stress and negative behaviors activity contributes to increased heart rate and
are associated with insufficientsleep. blood pressure variability and levels of serum
Sleep 321

norepinephrine (Dettoni etal., 2012; Irwin etal., sleep efficiency. People with posttraumatic stress
1999). In addition, hypothalamic CRH levels disorder (PTSD) report increased sleep latency,
increase following chronic partial sleep dep- less duration, and increased awakenings (Brown
rivation (5 days of REMS deprivation; Koban et al., 2011; (Cohen et al., 2009). Patients with
etal.,2006). insomnia report more severe symptoms of anxi-
ety and depression (Mason and Harvey, 2014).
LHPA Axis Tone and Behavior Even in young preschoolers, insufficient sleep
Increased CRH activity and LHPA axis tone is asso- is related to psychiatric symptoms, particularly
ciated with behaviors such as anxiety, irritability, anxiety (Steinsbekk etal.,2013).
aggression, and impulsiveness (see Figure 18.2).
Increased LHPA activity is also a predispos- Major Depressive Disorder
ing factor for depression-like psychopathologies In contrast to the effects in bipolar disorder, anxi-
(Roman etal.,2006). ety, stress, and PTSD, there is a well-documented
antidepressant effect of sleep deprivation in peo-
Insufficient Sleep and Behavior ple with major depressive disorder (Gillin, 1983;
Insufficient sleep increases the manifestation Landsness etal.,2011).
of behaviors including anxiety, hyperactivity,
distractibility, aggression, disinhibition, oppo- Pharmacology
sitional behavior, risk taking, irritability, and Norepinephrine. Currently, the most effective
impulsiveness (see Figure 18.2). Children who treatment strategy for PTSD includes cognitive
experience insufficient sleep may express day- behavioral therapy with either prazosin or an
time sleepiness by being overactive and hav- SSRI. Prazosin blocks central and peripheral nor-
ing a difficult time focusing. In children with epinephrine (adrenergic) alpha 1 receptors. This
autism, increased sleep disturbances are a pre- reduces sympathetic responses associated with
dictor of anxiety (May et al., 2013). A recent anxiety (elevated heart rate and/or increased
study reported that inconsistent sleep schedules blood pressure) and can enhance the ability to
and short sleep durations are associated with focus while awake and increase the probability of
behavioral difficulties in children (Biggs et al., REMSsleep.
2011). Further, self-reported sleep problems (by Serotonin. Reduced serotonin levels are asso-
children) predicted child- and teacher-reported ciated with increased depression, aggression,
behavioral problems, including reactive aggres- irritability, and hostility in humans and animals
sion (Becker,2014). (Fuller, 1996; Kamphuis et al., 2012). Reduced
Insufficient sleep is also a predisposing factor sleep efficiency (24-hour deprivation and partial
for depression-like psychopathologies (Roman deprivation) is associated with reducing sero-
et al., 2006). In animal models, chronic partial tonin tone (Figure 18.2; Asikainen et al., 1997;
sleep deprivation (4 hours/day for 1 wk) leads to Elmenhorst etal., 2012). Two advantages of SSRIs
a blunted adrenocorticotropic hormone response (which specifically inhibit serotonin uptake to
(but normal glucocorticoid response as described selectively increase extracellular serotonin lev-
previously), which resembles depression (Novati els), such as fluoxetine, are the associated reduc-
et al., 2008). Self-reports from bipolar patients tion in aggression (Fuller, 1996) and increase
indicate that periods of sleep deprivation can ofNREMS.
trigger depressive episodes (Proudfoot et al., Alcohol. Alcohol consumption reduces REMS.
2012). Child-reported sleep problems is a predic- Imaging analyses during REMS indicates a sig-
tor of child- and teacher-reported social func- nificant increase in amygdala and mesolimbic
tioning, feelings of loneliness, lower friendship dopaminergic activation. Future studies are
satisfaction, and lower self-worth (Becker,2014). required to test the hypothesis that these signals
reflect the processing of events that have emo-
Behavior and InsufficientSleep tional/motivational value (Perogamvros et al.,
Insufficient sleep can be a comorbid condi- 2013).
tion experienced by people with anxiety and Caffeine. Excessive caffeine intake is asso-
stress-related disorders (see Figure 18.2). ciated with insomnia, lower stress thresholds,
Comorbid symptoms in adults with general- increased aggressive behaviors, and increased
ized anxiety disorder and ADHD can have anxiety (see Fredholm etal., 1999, for extensive
increased latency to sleep onset and reduced review). In youth with major depressive disorder,
322 Part III:Homeostatic Manipulators

there is a higher incidence of caffeine intake How Does Sleep Influence


and sleep problems, and caffeine use is associ- Brain ATP Levels and Damage/
ated increased symptoms of anxiety (Whalen Waste Removal Processes?
etal.,2008). Recent studies have examined the role of sleep in
the homeostatic regulation of brain ATP levels
Sleep Therapy and the removal of damage and waste products
Ensuring sufficient sleep may be an effective within the brain milieu. These processes have
therapeutic adjuvant to attenuate stress, anxiety, dynamic interactions among synaptic transmis-
and/or depressive symptoms. Indeed, improve- sion, cognitive processes, adenosine levels, sleep,
ment of sleep quality increases clinical outcomes, reactive species production, and the pathology,
lessens depression, improves quality of life, and which can occur in neurological disorders asso-
increases physical health in individuals with ciated with sleep and cognitive comorbidities
PTSD (Germain et al., 2008). Sufficient sleep is (Figure18.3).
associated with significantly fewer behavioral dif-
ficulties in children (Biggs etal., 2011). Further, ATP
improved sleep is associated with improved social ATP, the brains energy currency, is critically
ability of autistic children (May etal.,2013). important for brain homeostasis. Mitochondria
produce approximately 97% of ATP, and, at the
How Does Sleep Influence cost of 25,000 molecules of ATP to transmit one
Ghrelin and Leptin, Brain bit of information, it is estimated that 41% is
Regulators of Body Weight? expended on synaptic transmission (Hall et al.,
Attainment of sufficient sleep is one mechanism 2012; Harris et al., 2012; Ligon and Steward,
to promote healthy weight and reduce the risk 2000). Recent studies suggest that sleep can pro-
for metabolic disorders. Insufficient sleep is a mote restoration of brain ATP levels. This notion
significant contributing factor of metabolic dis- is supported by data indicating that enzymes that
orders for many reasons; here we focus on two. promote ATP production (including pyruvate
Ghrelin and leptin are two appetite-regulating kinase, adenylate kinase, and malate dehydro-
neuropeptides. Ghrelin is secreted from the genase) peak at the beginning of the rest phase,
stomach and acts at the hypothalamus to pro- both glucose and ATP levels increase during
mote hunger. In contrast, leptin is secreted early NREMS, and deprivation of sleep reduces
from fat cells and acts at the hypothalamus to levels of ATP precursors, glycogen, and glucose
promote satiety. Partial chronic sleep depriva- (Dworak etal., 2010; North etal.,1981).
tion (less than 6 hours per night or late sleep
onset) is associated with increased levels of Damage, Pathology, and
ghrelin and reduced levels of the leptin in the Neurological Disorders
blood. Studies indicate that adults who report In addition to producing ATP, mitochondria can
having less than 5 hours of sleep per night are induce cell damage/injury and trigger cell death.
at higher risk of developing diabetes than those ATP is produced via mitochondrial complexes
who achieve 7 hours. In multiple, large-cohort Ithrough V.Impairment of electron transport at
human studies, partial chronic sleep depriva- complex I results in (i) reduced ATP-producing
tion (less than 6 hours per night or late sleep respiratory rates and (ii) increased production
onset) is associated with increased daily caloric of reactive species by the binding of misshuttled
intake, self-reported increases in hunger, electrons to oxygen and nitrogen (species include
increased BMI, weight gain, and the develop- superoxide, nitric oxide, hydroxyl radical, hydro-
ment of obesity (Spaeth et al., 2013; Spiegel gen peroxide, nitric dioxide, and peroxynitrite)
etal., 2004; Srivastava etal., 2013; Taheri etal., (Fariss etal., 2005; Simeone etal., 2014; Sullivan
2004; Watanabe et al., 2010). Further, glucose etal., 2003; Sullivan etal., 2004). High levels of
tolerance is impaired following restriction of these reactive species cause oxidative damage to
either total sleep time or NREMS SWA (Spiegel DNA, RNA, proteins, lipids, and cell membranes.
et al., 1999; Tasali et al., 2008). Collectively, The cascades of events that can result from mito-
these data suggest that attainment of sufficient chondrial impairment can induce a range of
sleep is one preventative/restorative therapy to pathology from minor cell injury to celldeath.
promote healthy weight and reduce the risk for Normal aging and neurological disor-
metabolic disorders. ders, including epilepsy, Alzheimers disease,
Sleep 323

Mitochondria Impaired mitochondria

Disrupted
Synaptic Low levels High levels
ATP Less ATP synaptic
transmission of RS of RS
transmission
?
Cognitive Less Cognitive Oxidative
Adenosine damage to
Processes adenosine impairments
lipids,
proteins,
nucleic
Disrupted
Sleep acids
sleep

Pathology:
Pathology: Astrogliosis
Cell injury/death

FIGURE 18.3: Dynamic multidirectional crosstalk among mitochondria, neurotransmission, pathology, cogni-
tion, and sleep. Left:Mitochondria play a critical role in regulating adenosine triphosphate (ATP) and reactive
species (RS) homeostasis, both factors upstream of synaptic transmission and cognitive processes. ATP is readily
converted into the somnogen adenosine. Right: Impaired mitochondria, specifically a complex I reduction of
function, is associated with reduced ATP and elevated RS, both implicated in impairing synaptic transmission
and cognitive processing. Impaired mitochondria are upstream of pathological features and are associated with
neurological disorders that have cognitive and sleep disorder comorbidities. The precise nature of the impairment
and the location within the brain will determine which circuitry and behavioral outputs are influenced.

and Parkinsons disease are associated with Damage and Waste Removal
a higher incidence of complex I impairment, The homeostatic mechanisms responsible for
increased levels of reactive species and subse- reducing oxidative damage and neurotoxic waste
quent oxidative damage (Brown et al., 2004; products is critical to prevent overaccumulation.
DiMauro etal., 1999; Fariss etal., 2005; Head Although these processes are not entirely under-
etal., 2009; Joseph etal., 2000; Simeone etal., stood, recent studies suggest that they may occur
2014; Wallace, 1992). Deficient pools of ATP more during sleep. Two waste removal mecha-
and elevated reactive species negatively impact nisms include the antioxidant system and the
synaptic transmission and are associated with glia-lymphatic system.
cognitive impairments (Chouhan et al., 2012; Antioxidants. Reactive species are reduced
Guo etal., 2005; Hall etal., 2012; Harris etal., by many endogenous antioxidants during both
2012; Hongpaisan et al., 2003; Hongpaisan the day and the night. The antioxidant and som-
etal., 2004; Hu etal., 2006; Kamsler and Segal, nogen melatonin is elevated during the night,
2003; Kann et al., 2011; Knapp and Klann, peaking after sleep onset (Redwine etal., 2000).
2002; Lee etal., 2012; Ligon and Steward, 2000; This reactive species clearance mechanism is not
Simeone et al., 2014; Thiels et al., 2000), a sensitive to sleep deprivation, as melatonin levels
hallmark feature of Alzheimers disease and a do not change following acute, partial sleep dep-
potential comorbid condition of epilepsy and rivation (Redwine et al., 2000). However, levels
Parkinsons disease. of the more potent antioxidant glutathione are
Astrogliosis is a pathology that can occur in significantly reduced following sleep deprivation
neurological disorders, including epilepsy and (DAlmeida etal., 2000; Silva etal.,2004).
Alzheimers disease. Astrogliosis and the con- Glia-lymphatic system. Recent reports indi-
comitant increase in adenosine kinase imply a cate that oxidative damage and neurotoxic waste
reduction of adenosine tone. Studies are needed products (i.e., metabolites, soluble proteins
to determine whether reduced adenosine tone including -amyloid, excess fluid, etc.) are cleared
occurs in the brain regions, such as the basal fore- via a glia-lymphatic system. The exchange rate of
brain or lateral hypothalamus, that are known to cerebral spinal fluid with fresh cerebral spinal
regulatesleep. fluid appears to be higher during periods of sleep
324 Part III:Homeostatic Manipulators

(Iliff and Nedergaard, 2013; Nedergaard, 2013; IL-1, IL-6, TNF, NF-
Xie etal.,2013). AA
Insufficient sleep is comorbid to neurologi-
cal disorders that can express pathology, includ- COX
ing astrogliosis, oxidative damage, cell injury,
Protaglandin H2
and cell death. Future studies are required to
determine whether insufficient sleep influences PGI PGE2
the glia-lymphatic system and whether reduced PGF2a
damage/waste clearance during the night pro- PGD2
motes the development of pathology.
- Acts on DP1R
Nutritional Pharmacology - Increases adenosine
Supplements and foods that target mitochondria
and are rich in antioxidants, including ascorbic FIGURE18.4: Immune factors, prostaglandins, aden-
acid and alpha tocopherol (vitamins C and E), can osine and sleep. One mechanism by which immune
be used to minimize accumulation of oxidative factors such as IL-1 beta, IL-6, TNF alpha, or NF kappa
damage. Additional compounds of mono- or pol- beta may promote sleep is by activating the cyclooxy-
ytherapies (including pyruvate and melatonin) genase (COX)-prostaglandin (PGD) pathway. COX
have also been used in preclinical treatments induces the conversion of arachidonic acid to prosta-
for sleep disorders in epilepsy (Simeone et al., glandin (PG) H2, which can be converted into PGD2.
2014).The ketogenic diet (KD), a diet high in fat PGD2 acts on its receptor (DP1R) to increase adenosine,
and low in carbohydrates and proteins, is highly an endogenous somnogen, in sleep-promoting regions.
effective in reducing medically refractory sei-
zures. As a disease-modifying treatment, the KD
is now being explored for its efficacy in autism, space of the ventral forebrain; Huang etal., 2007;
Alzheimers disease, and Parkinsons disease. The Pandey etal., 1995; Ram etal., 1997; Terao etal.,
KD improves mitochondrial function, restores 1998). PGD2 acts on its D prostanoid receptor 1
complex IATP-producing rates, reduces reactive in the basal forebrain, which increases levels of
species, increases endogenous antioxidants, and the somnogen adenosine and activation of the
enhances adenosine tone. Further, recent studies VLPO (Huang et al., 2007; Kristensson et al.,
have found that the KD improves sleep in chil- 2010; Mizoguchi etal., 2001; Saper etal., 2005b;
dren and animal models of epilepsy (Fenoglio Saper etal., 2010; Sherin etal.,1996).
et al., 2009; Hallbook et al., 2007; Masino and
Geiger, 2008; Roundtree et al., submitted; Sleep Can Enhance
Sullivan etal.,2003). Immune Responses
While anti-inflammatory cytokines peak dur-
How Does Sleep Influence ing the day, activity of many proinflammatory
Neuro-Immune Response Factors? factors peak following sleep onset, including lev-
The relationship between sleep and the immune els of cytokines, activation and proliferation of
system is bidirectional and complex. Here we high- proinflammatory APC and T cells, lymphocyte
light two examples:(i)immune factors can act as accumulation in lymph nodes, and phagocyto-
somnogens in specific brain areas to promote sleep, sis (Besedovsky et al., 2012; Lange et al., 2003;
and (ii) sleep can enhance immune responses. Lange et al., 2010). Recent studies suggest that
sleep not only increases the acute number of
Immune Factors Are Somnogenic antigen-specific T helper cells following vacci-
During infection, increased levels of proin- nations (in these studies for hepatitis A) but also
flammatory cytokines, such as TNF-alpha and promotes immunological memory as evidenced
IL-1beta, can act in the brain to promote sleep by by effects persisting one-year postvaccination
inducing NREMS (Figure 18.4). Studies suggest (Bollinger et al., 2011; Lange et al., 2003; Lange
that this is in part mediated via signal convergence etal., 2011; Phillips etal.,2008).
onto the prostaglandin2 (PGD2)-adenosine path-
way (Cao et al., 1996; Terao et al., 1998). PGD2 PROPER SLEEP HYGIENE
can increase NREMS or REMS when injected Routine and proper sleep hygiene is essential.
into different brain regions (i.e., the preoptic Based on the recommended amount of time adults
area, the lateral ventricles, or the subarachnoid and children spend in sleep, approximately 30% of
Sleep 325

brain activity in adults and 44% in children occurs Basheer, R., Strecker, R.E., Thakkar, M.M.,
during sleep. According to the National Sleep and McCarley, R.W. (2004). Adenosine and
Foundation and the National Institute of Health sleep-wake regulation. Prog. Neurobiol. 73,
Center for Sleep Disorders Research, the following 379396.
guidelines can be used to improve sleep hygiene. Becker, S.P. (2014). External validity of childrens
During the day, spend time outside in natural self-reported sleep functioning: Associations
light. Avoid exercising, eating large meals, and with academic, social, and behavioral adjust-
drinking immediately before bed. Avoid caffein- ment. Sleep Med. 15, 10941100.
ated and alcoholic beverages. Benington, J.H., Kodali, S.K., and Heller, H.C.
Maintain a consistent and positive bedtime (1995). Stimulation of A1 adenosine receptors
routine that ensures a sufficient number of hours mimics the electroencephalographic effects of
of sleep is achieved. Develop a bedtime routine sleep deprivation. Brain Res. 692,7985.
that begins 30 min before falling asleep, prefer- Besedovsky, L., Lange, T., and Born, J. (2012). Sleep
ably at the same time each night. This routine and immune function. Pflugers Arch. 463,
121137.
should include relaxing in a cool, quiet room,
Biggs, S.N., Lushington, K., van den Heuvel, C.J.,
which has reduced lighting and lacks electronic
Martin, A.J., and Kennedy, J.D. (2011).
sources of light. Commonly used artificial light
Inconsistent sleep schedules and daytime behav-
has more blue frequencies, which is associated
ioral difficulties in school-aged children. Sleep
with reduced levels of melatonin, an important
Med. 12, 780786.
somnogenic neuropeptide (Lockley etal.,2003).
Bittencourt, J.C., Presse, F., Arias, C., Peto, C.,
Ensuring sufficient sleep offers a preventa- Vaughan, J., Nahon, J.L., Vale, W., and Sawchenko,
tive measure or an adjuvant restorative therapy P.E. (1992). The melanin-concentrating hormone
that may improve cognition and/or behavior in system of the rat brain:An immuno- and hybrid-
normal individuals and lessen the degree of cog- ization histochemical characterization. J. Comp.
nitive/behavioral comorbid afflictions in indi- Neurol. 319, 218245.
viduals with neurological or affective disorders. Boison, D. (2006). Adenosine kinase, epilepsy and
stroke: Mechanisms and therapies. Trends
ACK NOWLEDGMENTS Pharmacol. Sci. 27, 652658.
The authors would like to thank the National Boison, D. (2008). The adenosine kinase hypoth-
Institutes for Healths National Institute of esis of epileptogenesis. Prog. Neurobiol. 84,
Neurological Disorders and Stroke (NS072179) 249262.
for support, E. Staneck for his technical assis- Boissard, R., Gervasoni, D., Schmidt, M.H., Barbagli, B.,
tance, and A. Sachs for his editorial assistance. Fort, P., and Luppi, P.H. (2002). The rat
Please note the first author has published under ponto-medullary network responsible for para-
the names K.A. Dorenbos, K.A. Fenoglio, and doxical sleep onset and maintenance: A combined
K.A. Simeone. microinjection and functional neuroanatomical
study. Eur. J.Neurosci. 16, 19591973.
References Bollinger, T., Leutz, A., Leliavski, A., Skrum, L.,
Alam, M.N., Gong, H., Alam, T., Jaganath, R., Kovac, J., Bonacina, L., Benedict, C., Lange, T.,
McGinty, D., and Szymusiak, R. (2002). Westermann, J., Oster, H., and Solbach, W.
Sleep-waking discharge patterns of neurons (2011). Circadian clocks in mouse and human
recorded in the rat perifornical lateral hypotha- CD4+ T cells. PLoS One 6, e29801.
lamic area. J. Physiol. 538, 619631. Bonnet, M.H., and Arand, D.L. (2003). Clinical
Alzheimers Association. (2012). 2012 Alzheimers effects of sleep fragmentation versus sleep depri-
disease facts and figures (Chicago: Alzheimers vation. Sleep Med. Rev. 7, 297310.
Association). https://www.alz.org/downloads/ Brown, M.R., Geddes, J.W., and Sullivan, P.G. (2004).
facts_figures_2012.pdf Brain region-specific, age-related, alterations in
Arborelius, L., Owens, M.J., Plotsky, P.M., and mitochondrial responses to elevated calcium. J.
Nemeroff, C.B. (1999). The role of corticotropin- Bioenerg. Biomembr. 36, 401406.
releasing factor in depression and anxiety disor- Brown, R.E., Basheer, R., McKenna, J.T., Strecker,
ders. J. Endocrinol. 160, 112. R.E., and McCarley, R.W. (2012). Control of sleep
Asikainen, M., Toppila, J., Alanko, L., Ward, D.J., and wakefulness. Physiol. Rev. 92, 10871187.
Stenberg, D., and Porkka-Heiskanen, T. (1997). Brown, T.H., Mellman, T.A., Alfano, C.A., and
Sleep deprivation increases brain serotonin turn- Weems, C.F. (2011). Sleep fears, sleep distur-
over in the rat. Neuroreport 8, 15771582. bance, and PTSD symptoms in minority youth
326 Part III:Homeostatic Manipulators

exposed to Hurricane Katrina. J. Trauma. Stress serotonin 2A receptor binding in humans. Sleep
24, 575580. 35, 16151623.
Cao, C., Matsumura, K., Yamagata, K., and Fadda, P., and Fratta, W. (1997). Stress-induced sleep
Watanabe, Y. (1996). Endothelial cells of the deprivation modifies corticotropin releasing fac-
rat brain vasculature express cyclooxygenase-2 tor (CRF) levels and CRF binding in rat brain
mRNA in response to systemic interleukin-1 and pituitary. Pharmacol. Res. 35, 443446.
beta: A possible site of prostaglandin synthesis Fariss, M.W., Chan, C.B., Patel, M., Van Houten, B.,
responsible for fever. Brain Res. 733, 263272. and Orrenius, S. (2005). Role of mitochondria in
Chou, T.C., Bjorkum, A.A., Gaus, S.E., Lu, J., toxic oxidative stress. Mol. Interv. 5, 94111.
Scammell, T.E., and Saper, C.B. (2002). Afferents Fedirchuk, B., and Dai, Y. (2004). Monoamines
to the ventrolateral preoptic nucleus. J. Neurosci. increase the excitability of spinal neurones in the
22, 977990. neonatal rat by hyperpolarizing the threshold
Chouhan, A.K., Ivannikov, M.V., Lu, Z., Sugimori, for action potential production. J. Physiol. 557,
M., Llinas, R.R., and Macleod, G.T. (2012). 355361.
Cytosolic calcium coordinates mitochondrial Fenik, V., Davies, R.O., and Kubin, L. (2004).
energy metabolism with presynaptic activity. J. Combined antagonism of aminergic excitatory
Neurosci. 32, 12331243. and amino acid inhibitory receptors in the XII
Cohen, S., Doyle, W.J., Alper, C.M., Janicki-Deverts, nucleus abolishes REM sleep-like depression of
D., and Turner, R.B. (2009). Sleep habits and sus- hypoglossal motoneuronal activity. Arch. Ital.
ceptibility to the common cold. Arch. Intern. Biol. 142, 237249.
Med. 169,6267. Fenoglio, K.A., Brunson, K.L., Avishai-Eliner, S.,
Curtis, D.R., Hosli, L., Johnston, G.A., and Johnston, Chen, Y., and Baram, T.Z. (2004). Region-specific
I.H. (1968). The hyperpolarization of spinal onset of handling-induced changes in
motoneurones by glycine and related amino corticotropin-releasing factor and glucocorti-
acids. Exp. Brain Res. 5, 235258. coid receptor expression. Endocrinology 145,
Daan, S., Beersma, D.G., and Borbely, A.A. (1984). 27022706.
Timing of human sleep:Recovery process gated Fenoglio, K.A., Brunson, K.L., Avishai-Eliner, S.,
by a circadian pacemaker. Am. J. Physiol. 246, Stone, B.A., Kapadia, B.J., and Baram, T.Z.
R161R183. (2005). Enduring, handling-evoked enhance-
DAlmeida, V., Hipolide, D.C., Lobo, L.L., de ment of hippocampal memory function and
Oliveira, A.C., Nobrega, J.N., and Tufik, S. glucocorticoid receptor expression involves acti-
(2000). Melatonin treatment does not prevent vation of the corticotropin-releasing factor type
decreases in brain glutathione levels induced 1 receptor. Endocrinology 146, 40904096.
by sleep deprivation. Eur. J. Pharmacol. 390, Fenoglio, K.A., Brunson, K.L., and Baram, T.Z.
299302. (2006a). Hippocampal neuroplasticity induced
Dang-Vu, T.T., Schabus, M., Desseilles, M., by early-life stress: Functional and molecular
Sterpenich, V., Bonjean, M., and Maquet, P. aspects. Front. Neuroendocrinol. 27, 180192.
(2010). Functional neuroimaging insights Fenoglio, K.A., Chen, Y., and Baram, T.Z.
into the physiology of human sleep. Sleep 33, (2006b). Neuroplasticity of the hypothalamic-
15891603. pituitary-adrenal axis early in life requires
Dettoni, J.L., Consolim-Colombo, F.M., Drager, L.F., recurrent recruitment of stress-regulating brain
Rubira, M.C., Souza, S.B., Irigoyen, M.C., regions. J. Neurosci. 26, 24342442.
Mostarda, C., Borile, S., Krieger, E.M., Fenoglio-Simeone KA, Wilke JC, Milligan HL, Allen
Moreno, H. Jr., and Lorenzi-Filho, G. (2012). CN, Rho JM, Maganti RK. Ketogenic diet treat-
Cardiovascular effects of partial sleep depriva- ment abolishes seizure periodicity and improves
tion in healthy volunteers. J. Appl. Physiol. (1985) diurnal rhythmicity in epileptic Kcna1-null
113, 232236. mice. Epilepsia. 2009;50(9):2027-2034.
DiMauro, S., Kulikova, R., Tanji, K., Bonilla, E., and Fisher, B.C., Garges, D.M., Yoon, S.Y., Maguire, K.,
Hirano, M. (1999). Mitochondrial genes for gen- Zipay, D., Gambino, M., and Shapiro, C.M.
eralized epilepsies. Adv. Neurol. 79, 411419. (2014). Sex differences and the interaction of
Dworak, M., McCarley, R.W., Kim, T., Kalinchuk, A.V., age and sleep issues in neuropsychological test-
and Basheer, R. (2010). Sleep and brain energy ing performance across the lifespan in an ADD/
levels:ATP changes during sleep. J. Neurosci. 30, ADHD sample from the years 1989 to 2009.
90079016. Psychol. Rep. 114, 404438.
Elmenhorst, D., Kroll, T., Matusch, A., and Bauer, A. Fisher, S.P., Davidson, K., Kulla, A., and Sugden, D.
(2012). Sleep deprivation increases cerebral (2008). Acute sleep-promoting action of the
Sleep 327

melatonin agonist, ramelteon, in the rat. J. Pineal lateral hypothalamus discharge maximally dur-
Res. 45, 125132. ing sleep. Eur. J.Neurosci. 32, 448457.
Fredholm, B.B., Battig, K., Holmen, J., Nehlig, A., Hassani, O.K., Lee, M.G., Henny, P., and Jones,
and Zvartau, E.E. (1999). Actions of caffeine in B.E. (2009). Discharge profiles of identified
the brain with special reference to factors that GABAergic in comparison to cholinergic and
contribute to its widespread use. Pharmacol. Rev. putative glutamatergic basal forebrain neurons
51, 83133. across the sleep-wake cycle. J. Neurosci. 29,
Fuller, R.W. (1996). Fluoxetine effects on serotonin 1182811840.
function and aggressive behavior. Ann. N.Y. Head, E., Nukala, V.N., Fenoglio, K.A., Muggenburg,
Acad. Sci. 794,9097. B.A., Cotman, C.W., and Sullivan, P.G. (2009).
Gallopin, T., Fort, P., Eggermann, E., Cauli, B., Luppi, Effects of age, dietary, and behavioral enrich-
P.H., Rossier, J., Audinat, E., Muhlethaler, M., ment on brain mitochondria in a canine model
and Serafin, M. (2000). Identification of of human aging. Exp. Neurol. 220, 171176.
sleep-promoting neurons in vitro. Nature 404, Hongpaisan, J., Winters, C.A., and Andrews, S.B.
992995. (2003). Calcium-dependent mitochondrial
Gaus, S.E., Strecker, R.E., Tate, B.A., Parker, R.A., and superoxide modulates nuclear CREB phos-
Saper, C.B. (2002). Ventrolateral preoptic nucleus phorylation in hippocampal neurons. Mol. Cell.
contains sleep-active, galaninergic neurons in Neurosci. 24, 11031115.
multiple mammalian species. Neuroscience 115, Hongpaisan, J., Winters, C.A., and Andrews, S.B.
285294. (2004). Strong calcium entry activates mitochon-
Germain, A., Buysse, D.J., and Nofzinger, E. (2008). drial superoxide generation, upregulating kinase
Sleep-specific mechanisms underlying post- signaling in hippocampal neurons. J. Neurosci.
traumatic stress disorder:Integrative review and 24, 1087810887.
neurobiological hypotheses. Sleep Med. Rev. 12, Hot, P., Rauchs, G., Bertran, F., Denise, P.,
185195. Desgranges, B., Clochon, P., and Eustache, F.
Gillin, J.C. (1983). The sleep therapies of depression. (2011). Changes in sleep theta rhythm are
Prog. Neuropsychopharmacol. Biol. Psychiatry 7, related to episodic memory impairment in early
351364. Alzheimers disease. Biol. Psychol. 87, 334339.
Gong, H., McGinty, D., Guzman-Marin, R., Chew, K.T., Hu, D., Serrano, F., Oury, T.D., and Klann, E. (2006).
Stewart, D., and Szymusiak, R. (2004). Activation Aging-dependent alterations in synaptic plastic-
of c-fos in GABAergic neurones in the preoptic ity and memory in mice that overexpress extra-
area during sleep and in response to sleep depri- cellular superoxide dismutase. J. Neurosci. 26,
vation. J. Physiol. 556, 935946. 39333941.
Guo, X., Macleod, G.T., Wellington, A., Hu, F., Huang, Z.L., Urade, Y., and Hayaishi, O. (2007).
Panchumarthi, S., Schoenfield, M., Marin, L., Prostaglandins and adenosine in the regula-
Charlton, M.P., Atwood, H.L., and Zinsmaier, tion of sleep and wakefulness. Curr. Opin.
K.E. (2005). The GTPase dMiro is required for Pharmacol. 7,3338.
axonal transport of mitochondria to Drosophila Huston, J.P., Haas, H.L., Boix, F., Pfister, M.,
synapses. Neuron 47, 379393. Decking, U., Schrader, J., and Schwarting, R.K.
Hall, C.N., Klein-Flugge, M.C., Howarth, C., and (1996). Extracellular adenosine levels in neostri-
Attwell, D. (2012). Oxidative phosphorylation, atum and hippocampus during rest and activity
not glycolysis, powers presynaptic and postsyn- periods of rats. Neuroscience 73, 99107.
aptic mechanisms underlying brain information Iliff, J.J., and Nedergaard, M. (2013). A link between
processing. J. Neurosci. 32, 89408951. glial Ca2+ signaling and hypoxia in aging? J.
Hallbook, T., Lundgren, J., and Rosen, I. (2007). Cereb. Blood Flow Metab. 33,170.
Ketogenic diet improves sleep quality in chil- Institute of Medicine Committee on Sleep Medicine
dren with therapy-resistant epilepsy. Epilepsia and Research. (2006). Sleep disorders and sleep
48,5965. deprivation: An unmet public health problem.
Harris, J.J., Jolivet, R., and Attwell, D. (2012). Synaptic HR Colten, BM Altevogt, eds.(Washington,
energy use and supply. Neuron 75, 762777. DC:Institute of Medicine).
Harrison, Y., and Horne, J.A. (2000). The impact of Institute of Medicine, Committee on the Public
sleep deprivation on decision making:Areview. Health Dimensions of the Epilepsies, Board on
J. Exp. Psychol. Appl. 6, 236249. Health Sciences Policy. (2012). Epilepsy across
Hassani, O.K., Henny, P., Lee, M.G., and Jones, the spectrum: Promoting health and under-
B.E. (2010). GABAergic neurons intermin- standing. MJ England et al., eds. (Washington,
gled with orexin and MCH neurons in the DC:National Academies Press).
328 Part III:Homeostatic Manipulators

Irwin, M., Thompson, J., Miller, C., Gillin, J.C., and Killgore, W.D. (2010). Effects of sleep deprivation on
Ziegler, M. (1999). Effects of sleep and sleep cognition. Prog. Brain Res. 185, 105129.
deprivation on catecholamine and interleukin-2 Kim, E.J., Baek, J.H., Shin, D.J., Park, H.M., Lee, Y.B.,
levels in humans: Clinical implications. J. Clin. Park, K.H., Shin, D.H., Noh, Y., and Sung, Y.H.
Endocrinol. Metab. 84, 19791985. (2014). Correlation of sleep disturbance and cog-
Ivy, A.S., Brunson, K.L., Sandman, C., and Baram, T.Z. nitive impairment in patients with Parkinsons
(2008). Dysfunctional nurturing behavior in disease. J. Mov. Disord. 7,1318.
rat dams with limited access to nesting mate- Knapp, L.T., and Klann, E. (2002). Potentiation of
rial: A clinically relevant model for early-life hippocampal synaptic transmission by superox-
stress. Neuroscience 154, 11321142. ide requires the oxidative activation of protein
Jelev, A., Sood, S., Liu, H., Nolan, P., and Horner, R.L. kinase C. J. Neurosci. 22, 674683.
(2001). Microdialysis perfusion of 5-HT into Koban, M., Le, W.W., and Hoffman, G.E. (2006).
hypoglossal motor nucleus differentially mod- Changes in hypothalamic corticotropin-releasing
ulates genioglossus activity across natural hormone, neuropeptide Y, and proopiomela-
sleep-wake states in rats. J. Physiol. 532, 467481. nocortin gene expression during chronic
Joseph, J.A., Denisova, N.A., Bielinski, D., Fisher, D.R., rapid eye movement sleep deprivation of rats.
and Shukitt-Hale, B. (2000). Oxidative stress Endocrinology 147, 421431.
protection and vulnerability in aging: Putative Korosi, A., and Baram, T.Z. (2008). The central cor-
nutritional implications for intervention. Mech. ticotropin releasing factor system during devel-
Ageing Dev. 116, 141153. opment and adulthood. Eur. J. Pharmacol. 583,
Kamphuis, J., Meerlo, P., Koolhaas, J.M., and Lancel, M. 204214.
(2012). Poor sleep as a potential causal factor in Koyama, Y., Takahashi, K., Kodama, T., and Kayama,
aggression and violence. Sleep Med. 13, 327334. Y. (2003). State-dependent activity of neurons in
Kamsler, A., and Segal, M. (2003). Hydrogen perox- the perifornical hypothalamic area during sleep
ide modulation of synaptic plasticity. J. Neurosci. and waking. Neuroscience 119, 12091219.
23, 269276. Kristensson, K., Nygard, M., Bertini, G., and
Kann, O., Huchzermeyer, C., Kovacs, R., Wirtz, S., Bentivoglio, M. (2010). African trypanosome
and Schuelke, M. (2011). Gamma oscillations in infections of the nervous system:Parasite entry
the hippocampus require high complex I gene and effects on sleep and synaptic functions. Prog.
expression and strong functional performance of Neurobiol. 91, 152171.
mitochondria. Brain 134, 345358. Lai, Y.Y., Kodama, T., and Siegel, J.M. (2001).
Kant, G.J., Genser, S.G., Thorne, D.R., Pfalser, J.L., Changes in monoamine release in the ventral
and Mougey, E.H. (1984). Effects of 72 hour sleep horn and hypoglossal nucleus linked to pontine
deprivation on urinary cortisol and indices of inhibition of muscle tone:An in vivo microdialy-
metabolism. Sleep 7, 142146. sis study. J. Neurosci. 21, 73847391.
Kayama, Y., Ohta, M., and Jodo, E. (1992). Firing Landsness, E.C., Goldstein, M.R., Peterson,
of possibly cholinergic neurons in the rat lat- M.J., Tononi, G., and Benca, R.M. (2011).
erodorsal tegmental nucleus during sleep and Antidepressant effects of selective slow wave sleep
wakefulness. Brain Res. 569, 210220. deprivation in major depression:Ahigh-density
Kempf, F., Brucke, C., Salih, F., Trottenberg, T., EEG investigation. J. Psychiatr. Res. 45,
Kupsch, A., Schneider, G.H., Doyle Gaynor, L.M., 10191026.
Hoffmann, K.T., Vesper, J., Wohrle, J., et al. Lange, T., Dimitrov, S., Bollinger, T., Diekelmann, S.,
(2009). Gamma activity and reactivity in human and Born, J. (2011). Sleep after vaccination
thalamic local field potentials. Eur. J. Neurosci. boosts immunological memory. J. Immunol. 187,
29, 943953. 283290.
Kent, J.M., Hough, D., Singh, J., Karcher, K., and Lange, T., Dimitrov, S., and Born, J. (2010).
Pandina, G. (2013). An open-label extension Effects of sleep and circadian rhythm on the
study of the safety and efficacy of risperidone human immune system. Ann. N.Y. Acad. Sci.
in children and adolescents with autistic dis- 1193,4859.
order. J. Child Adolesc. Psychopharmacol. 23, Lange, T., Perras, B., Fehm, H.L., and Born, J. (2003).
676686. Sleep enhances the human antibody response to
Kilduff, T.S., and de Lecea, L. (2001). Mapping hepatitis A vaccination. Psychosom. Med. 65,
of the mRNAs for the hypocretin/orexin 831835.
and melanin-concentrating hormone recep- Lazowski, L.K., Townsend, B., Hawken, E.R., Jokic, R.,
tors:Networks of overlapping peptide systems. J. du Toit, R., and Milev, R. (2014). Sleep architec-
Comp. Neurol. 435,15. ture and cognitive changes in olanzapine-treated
Sleep 329

patients with depression:Adouble blind random- effects of ketogenic diets? Trends Neurosci. 31,
ized placebo controlled trial. BMC Psychiatry 14, 273278.
202-244X-14-202. Mason, E.C., and Harvey, A.G. (2014). Insomnia
Lee, S.H., Kim, K.R., Ryu, S.Y., Son, S., Hong, H.S., before and after treatment for anxiety and
Mook-Jung, I., Lee, S.H., and Ho, W.K. (2012). depression. J. Affect. Disord. 168, 415421.
Impaired short-term plasticity in mossy fiber May, T., Cornish, K., Conduit, R., Rajaratnam,
synapses caused by mitochondrial dysfunction S.M., and Rinehart, N.J. (2013). Sleep in
of dentate granule cells is the earliest synaptic high-functioning children with autism:
deficit in a mouse model of Alzheimers disease. Longitudinal developmental change and associa-
J. Neurosci. 32, 59535963. tions with behavior problems. Behav. Sleep Med.
Lee-Chiong, T.L. (2008). Sleep medicine: Essentials 13,218.
and review (Oxford:Oxford University Press). McEwen, B.S., and Stellar, E. (1993). Stress and the
Leonard, C.S., and Llinas, R. (1994). Serotonergic individual. Mechanisms leading to disease.
and cholinergic inhibition of mesopontine cho- Arch. Intern. Med. 153, 20932101.
linergic neurons controlling REM sleep: An in McGinty, D.J., and Sterman, M.B. (1968). Sleep sup-
vitro electrophysiological study. Neuroscience pression after basal forebrain lesions in the cat.
59, 309330. Science 160, 12531255.
Ligon, L.A., and Steward, O. (2000). Movement of Mizoguchi, A., Eguchi, N., Kimura, K., Kiyohara, Y.,
mitochondria in the axons and dendrites of cul- Qu, W.M., Huang, Z.L., Mochizuki, T., Lazarus,
tured hippocampal neurons. J. Comp. Neurol. M., Kobayashi, T., Kaneko, T., et al. (2001).
427, 340350. Dominant localization of prostaglandin D recep-
Limoges, E., Bolduc, C., Berthiaume, C., Mottron, tors on arachnoid trabecular cells in mouse basal
L., and Godbout, R. (2013). Relationship between forebrain and their involvement in the regula-
poor sleep and daytime cognitive performance tion of non-rapid eye movement sleep. Proc.
in young adults with autism. Res. Dev. Disabil. Natl. Acad. Sci. U.S.A. 98, 1167411679.
34, 13221335. Moraes Wdos, S., Poyares, D.R., Guilleminault, C.,
Liu, Z.W., and Gao, X.B. (2007). Adenosine inhib- Ramos, L.R., Bertolucci, P.H., and Tufik, S.
its activity of hypocretin/orexin neurons by the (2006). The effect of donepezil on sleep and
A1 receptor in the lateral hypothalamus:Apos- REM sleep EEG in patients with Alzheimer dis-
sible sleep-promoting effect. J. Neurophysiol. 97, ease: A double-blind placebo-controlled study.
837848. Sleep 29, 199205.
Lockley, S.W., Brainard, G.C., and Czeisler, C.A. Morales, F.R., Engelhardt, J.K., Soja, P.J., Pereda,
(2003). High sensitivity of the human circadian A.E., and Chase, M.H. (1987). Motoneuron
melatonin rhythm to resetting by short wave- properties during motor inhibition produced
length light. J. Clin. Endocrinol. Metab. 88, by microinjection of carbachol into the pontine
45024505. reticular formation of the decerebrate cat. J.
Lu, J., Greco, M.A., Shiromani, P., and Saper, C.B. Neurophysiol. 57, 11181129.
(2000). Effect of lesions of the ventrolateral Morrison, J.L., Sood, S., Liu, H., Park, E., Liu, X.,
preoptic nucleus on NREMS and REM sleep. J. Nolan, P., and Horner, R.L. (2003). Role of
Neurosci. 20, 38303842. inhibitory amino acids in control of hypoglossal
Machi, M.S., Staum, M., Callaway, C.W., Moore, C., motor outflow to genioglossus muscle in natu-
Jeong, K., Suyama, J., Patterson, P.D., and rally sleeping rats. J. Physiol. 552, 975991.
Hostler, D. (2012). The relationship between shift National Institute of Mental Health. (2013).
work, sleep, and cognition in career emergency Statistics. http://www.nimh.nih.gov/health/sta-
physicians. Acad. Emerg. Med. 19,8591. tistics/index.shtml
Manns, I.D., Alonso, A., and Jones, B.E. (2000). National Sleep Foundation. (2013). How much
Discharge profiles of juxtacellularly labeled and sleep do we really need? http://sleepfoun-
immunohistochemically identified GABAergic d at ion .org / how-s le e p -work s/ how-muc h-
basal forebrain neurons recorded in association sleep-do-we-really-need
with the electroencephalogram in anesthetized Nedergaard, M. (2013). Neuroscience:Garbage truck
rats. J. Neurosci. 20, 92529263. of the brain. Science 340, 15291530.
Marshall, L., Helgadottir, H., Molle, M., and Born, J. Neubauer, D.N. (2008). A review of ramelteon in the
(2006). Boosting slow oscillations during sleep treatment of sleep disorders. Neuropsychiatr.
potentiates memory. Nature 444, 610613. Dis. Treat. 4,6979.
Masino, S.A., and Geiger, J.D. (2008). Are purines North, C., Feuers, R.J., Scheving, L.E., Pauly, J.E.,
mediators of the anticonvulsant/neuroprotective Tsai, T.H., and Casciano, D.A. (1981). Circadian
330 Part III:Homeostatic Manipulators

organization of thirteen liver and six brain and sleep in rats. J. Pharmacol. Exp. Ther. 228,
enzymes of the mouse. Am. J.Anat. 162, 183199. 268274.
Novati, A., Roman, V., Cetin, T., Hagewoud, R., Ram, A., Pandey, H.P., Matsumura, H.,
den Boer, J.A., Luiten, P.G., and Meerlo, P. Kasahara-Orita, K., Nakajima, T., Takahata, R.,
(2008). Chronically restricted sleep leads to Satoh, S., Terao, A., and Hayaishi, O. (1997). CSF
depression-like changes in neurotransmitter levels of prostaglandins, especially the level of
receptor sensitivity and neuroendocrine stress prostaglandin D2, are correlated with increas-
reactivity in rats. Sleep 31, 15791585. ing propensity towards sleep in rats. Brain Res.
Pandey, H.P., Ram, A., Matsumura, H., and Hayaishi, 751,8189.
O. (1995). Concentration of prostaglandin D2 in Redwine, L., Hauger, R.L., Gillin, J.C., and Irwin,
cerebrospinal fluid exhibits a circadian altera- M. (2000). Effects of sleep and sleep deprivation
tion in conscious rats. Biochem. Mol. Biol. Int. on interleukin-6, growth hormone, cortisol, and
37, 431437. melatonin levels in humans. J. Clin. Endocrinol.
Parkinsons Disease Foundation. (2014). Statistics on Metab. 85, 35973603.
Parkinsons. http://www.pdf.org/en/parkinson_ Roman, V., Hagewoud, R., Luiten, P.G., and Meerlo,
statistics P. (2006). Differential effects of chronic partial
Perogamvros, L., Dang-Vu, T.T., Desseilles, M., and sleep deprivation and stress on serotonin-1A and
Schwartz, S. (2013). Sleep and dreaming are for muscarinic acetylcholine receptor sensitivity. J.
important matters. Front. Psychol. 4,474. Sleep Res. 15, 386394.
Perrier, J.F., and Delgado-Lezama, R. (2005). Roundtree, H.M., Samson, K.K., Matthews, S.,
Synaptic release of serotonin induced by stimu- Simeone, T.A., and Simeone, K.A. (2015).
lation of the raphe nucleus promotes plateau Orexinergic activity in the lateral hypothalamus
potentials in spinal motoneurons of the adult may contribute to insufficient sleep in a model
turtle. J. Neurosci. 25, 79937999. epilepsy with a comorbid sleep disorder.
Phillips, A.C., Gallagher, S., Carroll, D., and Drayson, Saito, Y., Cheng, M., Leslie, F.M., and Civelli, O.
M. (2008). Preliminary evidence that morning vac- (2001). Expression of the melanin-concentrating
cination is associated with an enhanced antibody hormone (MCH) receptor mRNA in the rat
response in men. Psychophysiology 45, 663666. brain. J. Comp. Neurol. 435,2640.
Piperidou, C., Karlovasitou, A., Triantafyllou, N., Sakurai, T. (2007). The neural circuit of orexin
Terzoudi, A., Constantinidis, T., Vadikolias, K., (hypocretin): Maintaining sleep and wakeful-
Heliopoulos, I., Vassilopoulos, D., and ness. Nat. Rev. Neurosci. 8, 171181.
Balogiannis, S. (2008). Influence of sleep distur- Sakurai, T., Mieda, M., and Tsujino, N. (2010). The
bance on quality of life of patients with epilepsy. orexin system: Roles in sleep/wake regulation.
Seizure 17, 588594. Ann. N.Y. Acad. Sci. 1200, 149161.
Pistacchi, M., Gioulis, M., Contin, F., Sanson, F., Saper, C.B., Cano, G., and Scammell, T.E. (2005a).
and Marsala, S.Z. (2014). Sleep disturbance and Homeostatic, circadian, and emotional regula-
cognitive disorder:Epidemiological analysis in a tion of sleep. J. Comp. Neurol. 493,9298.
cohort of 263 patients. Neurol. Sci. 35, 1955162. Saper, C.B., Fuller, P.M., Pedersen, N.P., Lu, J., and
Plihal, W., and Born, J. (1997). Effects of early and Scammell, T.E. (2010). Sleep state switching.
late nocturnal sleep on declarative and proce- Neuron 68, 10231042.
dural memory. J. Cogn. Neurosci. 9, 534547. Saper, C.B., Scammell, T.E., and Lu, J. (2005b).
Porkka-Heiskanen, T., and Kalinchuk, A.V. (2011). Hypothalamic regulation of sleep and circadian
Adenosine, energy metabolism and sleep homeo- rhythms. Nature 437, 12571263.
stasis. Sleep Med. Rev. 15, 123135. Sherin, J.E., Elmquist, J.K., Torrealba, F., and Saper,
Porkka-Heiskanen, T., Strecker, R.E., Thakkar, M., C.B. (1998). Innervation of histaminergic tuber-
Bjorkum, A.A., Greene, R.W., and McCarley, R.W. omammillary neurons by GABAergic and
(1997a). Adenosine: A mediator of the galaninergic neurons in the ventrolateral preop-
sleep-inducing effects of prolonged wakefulness. tic nucleus of the rat. J. Neurosci. 18, 47054721.
Science 276, 12651268. Sherin, J.E., Shiromani, P.J., McCarley, R.W., and
Proudfoot, J., Whitton, A., Parker, G., Doran, J., Saper, C.B. (1996). Activation of ventrolateral
Manicavasagar, V., and Delmas, K. (2012). preoptic neurons during sleep. Science 271,
Triggers of mania and depression in young 216219.
adults with bipolar disorder. J. Affect. Disord. Silva, R.H., Abilio, V.C., Takatsu, A.L., Kameda,
143, 196202. S.R., Grassl, C., Chehin, A.B., Medrano, W.A.,
Radulovacki, M., Virus, R.M., Djuricic-Nedelson, Calzavara, M.B., Registro, S., Andersen, M.L.,
M., and Green, R.D. (1984). Adenosine analogs et al. (2004). Role of hippocampal oxidative
Sleep 331

stress in memory deficits induced by sleep Suntsova, N., Szymusiak, R., Alam, M.N.,
deprivation in mice. Neuropharmacology 46, Guzman-Marin, R., and McGinty, D. (2002).
895903. Sleep-waking discharge patterns of median pre-
Simeone, K.A., Matthews, S.A., Samson, K.K., and optic nucleus neurons in rats. J. Physiol. 543,
Simeone, T.A. (2014). Targeting deficiencies in 665677.
mitochondrial respiratory complex I and func- Szymusiak, R., Alam, N., Steininger, T.L., and
tional uncoupling exerts anti-seizure effects in McGinty, D. (1998). Sleep-waking discharge pat-
a genetic model of temporal lobe epilepsy and terns of ventrolateral preoptic/anterior hypotha-
in a model of acute temporal lobe seizures. Exp. lamic neurons in rats. Brain Res. 803, 178188.
Neurol. 251,8490. Szymusiak, R., and McGinty, D. (2008).
Spadoni, G., Bedini, A., Rivara, S., and Mor, M. Hypothalamic regulation of sleep and arousal.
(2011). Melatonin receptor agonists:New options Ann. N.Y. Acad. Sci. 1129, 275286.
for insomnia and depression treatment. CNS Taheri, S., Lin, L., Austin, D., Young, T., and Mignot,
Neurosci. Ther. 17, 733741. E. (2004). Short sleep duration is associated with
Spaeth, A.M., Dinges, D.F., and Goel, N. (2013). reduced leptin, elevated ghrelin, and increased
Effects of experimental sleep restriction on body mass index. PLoS Med. 1,e62.
weight gain, caloric intake, and meal timing in Takahashi, K., Lin, J.S., and Sakai, K. (2009).
healthy adults. Sleep 36, 981990. Characterization and mapping of sleep-waking
Spiegel, K., Leproult, R., and Van Cauter, E. (1999). specific neurons in the basal forebrain and pre-
Impact of sleep debt on metabolic and endocrine optic hypothalamus in mice. Neuroscience 161,
function. Lancet 354, 14351439. 269292.
Spiegel, K., Tasali, E., Penev, P., and Van Cauter, E. Tarokh, L., and Achermann, P. (2013). Sleep homeo-
(2004). Brief communication: Sleep curtail- stasis. In: The encyclopedia of sleep, Kushida
ment in healthy young men is associated with C. ed. (Waltham, MA: Academic Press), pp.
decreased leptin levels, elevated ghrelin levels, 413417.
and increased hunger and appetite. Ann. Intern. Tasali, E., Leproult, R., Ehrmann, D.A., and Van
Med. 141, 846850. Cauter, E. (2008). Slow-wave sleep and the risk of
Srinivasan, V., Brzezinski, A., Pandi-Perumal, S.R., type 2 diabetes in humans. Proc. Natl. Acad. Sci.
Spence, D.W., Cardinali, D.P., and Brown, G.M. U.S.A. 105, 10441049.
(2011). Melatonin agonists in primary insom- Terao, A., Matsumura, H., and Saito, M. (1998).
nia and depression-associated insomnia: Are Interleukin-1 induces slow-wave sleep at the
they superior to sedative-hypnotics? Prog. prostaglandin D2-sensitive sleep-promoting
Neuropsychopharmacol. Biol. Psychiatry 35, zone in the rat brain. J. Neurosci. 18, 65996607.
913923. Thiels, E., Urban, N.N., Gonzalez-Burgos, G.R.,
Srivastava, S., Baxa, U., Niu, G., Chen, X., and Veech, Kanterewicz, B.I., Barrionuevo, G., Chu, C.T.,
R.L. (2013). A ketogenic diet increases brown Oury, T.D., and Klann, E. (2000). Impairment of
adipose tissue mitochondrial proteins and UCP1 long-term potentiation and associative memory
levels in mice. IUBMB Life 65,5866. in mice that overexpress extracellular superoxide
Steinsbekk, S., Berg-Nielsen, T.S., and Wichstrom, L. dismutase. J. Neurosci. 20, 76317639.
(2013). Sleep disorders in preschool- Van Dongen, H.P., Maislin, G., Mullington, J.M.,
ers:Prevalence and comorbidity with psychiatric and Dinges, D.F. (2003). The cumulative cost of
symptoms. J. Dev. Behav. Pediatr. 34, 633641. additional wakefulness: Dose-response effects
Steriade, M., Datta, S., Pare, D., Oakson, G., and on neurobehavioral functions and sleep physiol-
Curro Dossi, R.C. (1990). Neuronal activities in ogy from chronic sleep restriction and total sleep
brain-stem cholinergic nuclei related to tonic deprivation. Sleep 26, 117126.
activation processes in thalamocortical systems. Verret, L., Goutagny, R., Fort, P., Cagnon, L.,
J. Neurosci. 10, 25412559. Salvert, D., Leger, L., Boissard, R., Salin, P.,
Sullivan, P.G., Dube, C., Dorenbos, K., Steward, O., and Peyron, C., and Luppi, P.H. (2003). A role of
Baram, T.Z. (2003). Mitochondrial uncoupling melanin-concentrating hormone producing
protein-2 protects the immature brain from exci- neurons in the central regulation of paradoxical
totoxic neuronal death. Ann. Neurol. 53, 711717. sleep. BMC Neurosci.4,19.
Sullivan, P.G., Rippy, N.A., Dorenbos, K., Waggoner, L.B., Grant, D.A., Van Dongen, H.P.,
Concepcion, R.C., Agarwal, A.K., and Rho, J.M. Belenky, G., and Vila, B. (2012). A combined field
(2004). The ketogenic diet increases mitochon- and laboratory design for assessing the impact
drial uncoupling protein levels and activity. of night shift work on police officer operational
Ann. Neurol. 55, 576580. performance. Sleep 35, 15751577.
332 Part III:Homeostatic Manipulators

Walker, M.P., Brakefield, T., Morgan, A., Hobson, J.A., R.E. (2008). Caffeine consumption, sleep, and
and Stickgold, R. (2002). Practice with sleep makes affect in the natural environments of depressed
perfect: Sleep-dependent motor skill learning. youth and healthy controls. J. Pediatr. Psychol.
Neuron 35, 205211. 33, 358367.
Wallace, D.C. (1992). Mitochondrial genetics:Apar- Wilson, S., and Argyropoulos, S. (2005).
adigm for aging and degenerative diseases? Antidepressants and sleep: A qualitative review
Science 256, 628632. of the literature. Drugs 65, 927947.
Watanabe, M., Kikuchi, H., Tanaka, K., and Takahashi, Xie, L., Kang, H., Xu, Q., Chen, M.J., Liao, Y.,
M. (2010). Association of short sleep duration with Thiyagarajan, M., ODonnell, J., Christensen,
weight gain and obesity at 1-year follow-up: A large- D.J., Nicholson, C., Iliff, J.J., et al. (2013). Sleep
scale prospective study. Sleep 33, 161167. drives metabolite clearance from the adult brain.
Whalen, D.J., Silk, J.S., Semel, M., Forbes, E.E., Ryan, Science 342, 373377.
N.D., Axelson, D.A., Birmaher, B., and Dahl,
19
Botanicals
DA NA EK ST EIN A ND ST E V EN C .SCH ACH T ER

INTRODUCTION Western treatments. For example, according to


Herbal therapy had been the main type of medi- the most recent national survey conducted in
cal treatment worldwide up until the last cen- 2007 (Barnes etal., 2008), nonvitamin/nonmin-
tury. With the development of modern Western eral natural products were the most widely used
medicine, in parallel with the pharmaceutical type of CHA, with 17.7% of Americans reporting
industry and regulatory agencies, the wide use their consumption.
of botanicals was replaced by refined pharma- Interestingly, this return to traditional rem-
ceutical compounds, specifically targeted at edies by consumers coincidentally occurred in
disease processes. This change has been enabled parallel with the development of systems biology,
by each great advance in the understanding of focusing on the complex physical and functional
pathophysiology, extending from the genomic relations between cellular molecules, organs,
and through the proteomic and system levels. and whole organisms (Ideker etal., 2001; Kitano,
Novel pharmaceutical medications were ration- 2002), and the emergence of network pharmacol-
ally designed to act at very specific biologic sites ogy. Network pharmacology, discussed in detail
and reverse the pathologic state. These new treat- in chapter14 - Systems (network) pharmacology
ments widely replaced the traditional herbal rem- for brain functionality restoration, refers to treat-
edies, and their introduction coincided with the ment comprising combinations of compounds to
development of an entire system aimed at prov- maximize the required therapeutic effect and
ing their safety and efficacy, thereby establishing minimize undesired adverse effects of the active
the gold standard of evidence-based medicine. components (Hopkins, 2007; Keith etal., 2005).
Botanicals are now considered part of com- Conceptually, this type of combination poly-
plementary and alternative medicine or comple- therapy is the basis of herbalism, where various
mentary health approaches (CHA)the latter is active compounds are contained in every plant
the preferred term most recently introduced for (herb) derived extract and mixtures of herbs are
these medical practices by the National Center used for treatment of medical conditions. Indeed,
for Complementary and Alternative Medicine several publications have recently attributed net-
(2013). CHA are defined as medical remedies work pharmacology characteristics to herbal
that are not part of the mainstream Western treatment as practiced in TCM (Li and Zhang,
type of medicine. They include natural prod- 2013; Zhang etal.,2013).
ucts, mind and body practices, and practices of The introduction of system biology and net-
whole health systems, such as homeopathy or work pharmacology is particularly relevant to
traditional Chinese medicine (TCM). Herbs (or the treatment of epilepsy, one of the most com-
botanicals), minerals, vitamins, and probiotics mon neurological disorders. Whereas epilepsy
are all considered natural products. Traditional (a chronic neurologic condition characterized
herbal treatments were never completely aban- by recurrent epileptic seizures; Blume et al.,
doned in the developing parts of the world, such 2001)used to be viewed only as a seizure disor-
as China, Korea, India, and South America, der, the current widely used definition acknowl-
and the use of CHA has become increasingly edges that epilepsy is a disorder of the brain
prevalent in Western countries over the past characterized by an enduring predisposition
few decades, especially in attempts to enhance to generate epileptic seizures and by the neu-
wellness or treat the symptoms of chronic dis- robiologic, cognitive, psychological, and social
eases, reflecting disappointment with modern consequences of this condition (Table1, p.471)
334 Part III:Homeostatic Manipulators

(Fisher et al., 2005). This definition reflects the theoretical and empirical aspects of the proper-
broader, network-like character of epilepsy, ties of single herbs and combinations. The con-
implies a complex pathophysiology, and suggests temporary traditional Chinese physician usually
the need for treatments aimed at alleviating both chooses from about 500 classical formulas, each
the occurrence of epileptic seizures as well as the combining 4 to 10 medicinal herbs on average.
related consequences or comorbidities. The prescription formulas are chosen to con-
In addition, a great emphasis has been put form to the jun (ruler, monarch) chen (minis-
on mitigating the adverse effects of antiepileptic ter, official) zuo (assistant) shi (enabler) principle
drugs (AEDs). Accordingly, one of the four 2014 that guides specific combinations. The mon-
National Institute of Neurological Disorders arch is the ingredient directed to act against
and Strokes benchmarks for epilepsy research is the main cause and symptoms of the disease.
Improve treatment options for controlling sei- The ministers attack the underlying causes of
zures and epilepsy-related conditions without the disease and the accompanying symptoms and
side effects. complications. The assistant compounds help
Epilepsy, like many other central nervous the two leading types of drugs to achieve their
system (CNS) disorders, can be viewed as dis- effects by counteracting their potential adverse
equilibrium of normal brain homeostasis, and effects and by treating secondary symptoms of
therefore the goal of treatment should be to the disease. The enablers direct the actions of
restore healthy homeostasis. While using net- the other drugs to ensure that together the other
work pharmacology as a strategy to discover new ingredients do not exceed the patients capacity
AEDs has already been proposed (Margineanu, to cope with their actions (Sucher, 2013). These
2013), we present the case for considering botani- prescription formulations, based on combina-
cals as potential therapies for epilepsy, consistent tions of herbs with various actions, are designed
with the principles of network pharmacology. to achieve homeostasis in systemic conditions, as
well as in brain disorders, and whether they treat
T R A D I T I O N A L T R E AT M E N T disease with a lower risk of undesirable adverse
W I T H B O TA N I C A L S U S E S effects compared to Western medicines requires
MIXTURES OFHERBS furtherstudy.
According to TCM, health is perceived as a bal- Dose-related adverse effects are particularly
anced, homeostatic state. Disease is caused by problematic consequences of Western treatments
unbalance of various defined functional and for chronic CNS disorders. A systematic review
energetic systems, as diagnosed by a complex and network meta-analysis of the tolerability of
of symptoms derived from observation, auscul- AEDs for refractory focal epilepsy included 8,546
tation and olfaction, interrogation, and pulse patients in 43 different trials taking a total of 11
analysis. Therefore, treatment is aimed at restor- AEDs (Bodalia et al., 2013). Overall, the odds
ing the homeostasis of the organism. Chinese ratio for premature drug withdrawal due to devel-
herbal medicine is the most widely used among opment of adverse effects for all AEDs versus pla-
all TCM practices. More than 8,800 different cebo was 3.27. The lack of high-quality trials of
medicinal compounds, extracted mainly from herbal treatments precludes comparison between
plants but also from animals and minerals, are these treatments and modern AEDs, whether for
known. These products are grouped into three efficacy, safety, or tolerability, but recent trials
classes. The lowest class has a low therapeutic of botanicals have suggested better tolerability
index (ratio of toxic dose to effective dose), with compared to phenobarbital (Li etal.,2009).
herbs meant to be taken in small doses to treat
specific disorders for short periods of time. The B O TA N I C A L S C O N TA I N
medicinal herbs in the middle class are safer but MIXTURES OFACTIVE
are also not recommended for prolonged use. The COMPOUNDS
top class includes products that are the safest and Individual plants frequently exert numerous
are recommended for daily consumption to pro- beneficial effects on disease states. Curcumin,
mote general health and longevity (Sucher,2013). for example, has been shown to ameliorate vari-
Combination herbal therapy is a fundamen- ous neurological conditions (Kulkarni and Dhir,
tal principle of TCM, and the herbal combination 2010). The homeostatic effect of curcumin on
formulas have been developed over thousands the brain was nicely demonstrated in an animal
of years by taking into consideration both model of chronic epilepsy induced by chemical
Botanicals 335

kindling. This plant extract not only alleviated may be explained on the basis of the comple-
epileptic seizures and improved depression, mentary activity of different compounds on sev-
learning, and memory but was shown to nor- eral mechanisms, since 9-THC and CBD were
malize epilepsy-induced changes in brain nor- found to inhibit T-type calcium channels (Ross
epinephrine, serotonin, and nitrite level and etal., 2008), CBD regulates intracellular calcium
acetylcholinesterase activity as well (Choudhary concentration (Ryan etal., 2009), and 9-THCV
etal.,2013). modulates inhibitory activity (Ma etal.,2008).
The complexity of botanical treatments can
be at least partially explained by the plethora HER BAL EXTR ACT ED
of compounds with various effects that can C O M P O U N D S E X E R T A VA R I E T Y
be extracted from each individual plant. For O F C O M P L E M E N TA R Y E F F E C T S
example, about 80 different phytocannabinoid The therapeutic effects and restoration of homeo-
compounds can be extracted from the cannabis stasis exerted by botanicals may potentially be
sativa plant. The most psychogenic compound, explained by the combined properties of numer-
9-tetrahydrocannabinol (9-THC), has received ous single compounds extracted from plants.
the most attention. However, numerous phyto- Each one of these compounds is usually not a
cannabinoids with very weak or no psychotropic magic bullet, as desired for pharmaceutical
effects can be extracted from the cannabis plant. drugs, but rather a molecule with various sites of
These include, among others, cannabidiol (CBD), action that mildly modulates physiological pro-
cannabigerol (CBG), cannabichromene (CBC), cesses. For example, CBD, the major nonpsycho-
9-tetrahydrocannabivarin (9-THCV), canna- tropic compound of cannabis, has been reported
bidivarin (CBDV), 9-tetrahydrocannabinolic to exert its effects on signaling pathways by bind-
acid (9-THCA), and cannabidiolic acid (CBDA) ing to at least 22 different sites (Izzo etal., 2009).
(Izzo et al., 2009). There is evidence of ancient Its influence on intracellular calcium concen-
medical use of cannabis in China (named trations (a major modulator of physiologic and
ma-fen), Assyria (azallu or gan-zi-gun-nu), pathologic processes in the CNS) is of particu-
and India (Indian hemp). Cannabis was pre- lar interest, since CBD was shown to increase or
scribed for the treatment of a variety of systemic decrease those concentrations, depending on the
and neurological conditions, including fever, excitatory status of the cell, therefore exerting
arthritis, weight loss, injuries, depression, impo- its action by promoting intracellular homeosta-
tence, pain, inflammation, epilepsy, migraines, sis of this important element (Ryan etal., 2009).
neuralgia, cramps, and tetanus (Mechoulam and Though many of the clinical consequences of
Hanu, 2001). The purported ability to ameliorate CBD have been associated with its mechanisms of
a large diversity of pathologic conditions suggests action, other associations remain to be revealed.
the possibility of a general, homeostatic effect of Another example of a homeostatic herbal
this plants constituent compounds. However, compound is the alkaloid Huperzine A, extracted
this effect ceases to exist beyond a certain dose from the plant Huperzia serrata. This plant has
of the remedy, when adverse effects are fre- been traditionally used in China for the treat-
quently encountered. This limitation of cannabis ment of many conditions, including contusions,
has been well known, and its psychotropic and strains, swelling, and schizophrenia (Ma et al.,
gait-disturbing properties when taken in excess 2007). Huperzine A is approved in China for
were mentioned even in ancient Chinese records treatment of dementia. Arandomized controlled
(Mechoulam and Hanu,2001). trial performed in the United States showed only
Modern science has managed to decipher classIII evidence of its efficacy in mild to mod-
many of the balancing effects of cannabis by erate Alzheimers disease, demonstrating cogni-
identifying different mechanisms of action of tive benefit after 16 weeks of treatment with 0.8
many of its compounds. For example, 9-THC mg Huperzine Aa day compared to placebo, and
was found to increase appetite and stabilize failed to show efficacy at the primary end point of
weight in patients with AIDS (Beal etal., 1995), improvement in scores on the cognitive subscale
and CBD showed anxiolytic activity and increase of the Alzheimer's Disease Assessment Scale with
in serotonergic activity in animal models (Izzo a daily dose of 0.4 mg (Rafii etal., 2011). However,
etal., 2009), properties that may explain its use many preclinical studies reported cognitive
for the treatment of depression. The potential improvement after treatment with Huperzine A,
of cannabis to help in the treatment of epilepsy and numerous mechanisms of action support the
336 Part III:Homeostatic Manipulators

same line of evidence (Zhang et al., 2008). The can be used in combination, based on their
main mechanisms of action of Huperzine Aare mechanisms of action, to achieve best therapeu-
inhibition of acetylcholinesterase (Tang et al., tic results; and botanicals may lead to discov-
1989) and antagonism at the NMDA receptor ery of new target sites for an enhanced rational
(Zhang and Hu, 2001). Additionally, it was found approach to treatment combining the best of
to exert other influences in laboratory studies rel- traditional and modern approaches. However,
evant to cognitive dysfunction, as well as other these great opportunities are limited by numer-
neurological disorders, such as multiple sclero- ous challenges encountered when performing
sis, amyotrophic lateral sclerosis, pain, epilepsy, clinical studies and by the incomplete informa-
and stroke, by protecting against oxidative stress, tion available to date on the biological actions of
inhibiting apoptosis, promoting neurogenesis, botanicals in various CNS disorders (Head and
decreasing inflammation, and modulating ion Kelly, 2009; Izzo et al., 2009; Namjooyan et al.,
currents (Bialer etal., 2009; Hemendinger etal., 2014; Sucher,2006).
2008; Li and Hu, 2002a, 2002b; Ma et al., 2013;
Pohanka etal., 2011; Tian etal., 2013; Wang etal., I N T E G R AT I N G H E R B S A N D
2008; Yu etal., 2013). The homeostatic properties HERBAL COMPOUNDS IN
of Huperzine A, similarly to those of other herbal WESTERN MEDICINE AFTER
products, are dose-dependent, implying the need THEIR RIGOROUS TESTING
for physician-guided administration, despite BY THE PRINCIPLES OF
the common notion that natural means safe EVIDENCE-BASED MEDICINE
and thus that the consumption of herbal reme- Despite their thousands of years of use, most
dies does not have to be as rigorously monitored herbal remedies have never been rigorously
as do pharmaceuticals. This dose-dependent tested using modern preclinical animal mod-
homeostasis is reflected by dual opposite actions els or in human randomized controlled trials
of the compound: it supports the proliferation for safety, efficacy, and tolerability, as required
of cultured neuronal stem cells when used in by regulatory agencies such as the US Food and
low concentrations as opposed to inhibition of Drug Administration, before they can be pre-
their proliferation in high concentrations (Ma scribed for specific diseases. The most promising
etal.,2013). botanicals should be studied using a bench-to-
bedside paradigm with full incorporation of
I M P R O V I N G T R E AT M E N T regulatory oversight. Such an approach for the
OF CNS DISORDERS BASED treatment of epilepsy is the Harvard Epilepsy
ON LESSONS FROM USE OF Botanical Program (Schachter, 2009) launched
B O TA N I C A L S by one of the authors (SS). This program was
The main protocol for drug development in established to support preclinical evaluation of
the modern pharmaceutical industry has been herbal therapies for epilepsy. Its goals are (a) to
target-based. This approach implies in-depth identify herbal therapies and compounds iso-
acquaintance with disease processes and with the lated from them that have promising activity
mechanisms of action of candidate compounds. in animal epilepsy models and relevant in vitro
However, since the productivity of this approach assays, (b)to conduct the preclinical studies nec-
has waned, for example in epilepsy, the need for essary to proceed with early-stage clinical stud-
a paradigm shift emphasizing the disease to be ies, (c)to plan and initiate these clinical studies.
treated and its manifestations has been stressed As part of the program, numerous mixtures and
(Schmidt and Schachter, 2014). Our knowledge individual herbal extracts and isolated com-
of mechanisms involved in pathophysiological pounds have been tested through the National
processes of diseases and exerted by drugs is at Institute of Neurological Disorders and Strokes
best only partial (Sams-Dodd, 2013), and tradi- Anticonvulsant Screening Project and in in vitro
tional herbal remedies represent a rich potential assays of neuronal receptor or ion channel func-
source for a pipeline of novel, systems-based, tion. More than half of these products (70%) have
disease-centered medications (Sucher, 2013). shown promise in preclinical testing. The lead
In this way, botanicals can be used according product of this pipeline is Huperzine A, men-
to their traditional indications after acquiring tioned earlier. This compound was found to be
stringent evidence for their safety and efficacy, extremely potent in suppressing seizures in ani-
using modern testing protocols and manufac- mal models of seizures (Bialer etal., 2009)and is
turing methods. Constituent herbal compounds now advancing to clinical trials.
Botanicals 337

The main challenges limiting the design of the highest research standards when studying
clinical studies on the use of botanicals and the botanicals.
interpretation of their results are the consequence
of the large variability in plants contents. The pre- M A N U FA C T U R I N G P R O D U C T S
cise chemical composition of plants extracts are T O A F F E C T N E W LY
not only influenced by the exact strain, place of D I S C O V E R E D TA R G E T S
growth, and weather but also by even minor dif- Botanicals may lead to discovery of new physi-
ferences in the extraction method. Additionally, ologic pathways that will help us better under-
the interpretation of results of studies performed stand disease processes and develop therapies.
in regions of the world where herbal medicine For example, research on the mechanisms of
has been traditionally implemented is problem- action of cannabis compounds revealed their
atic, since the practice of diagnostics is differ- receptors (Matsuda et al., 1990; Munro et al.,
ent in these regions than in Western medicine, 1993), which had been previously unknown, and
using a more holistic approach. Furthermore, consequently the endocannabinoid (Mechoulam
the prescription of natural remedies usually and Ben-Shabat, 1999) system was revealed.
includes individualized mixtures of botani- The elements of this complex system were then
cals, aimed not only to be directly ingested but promptly proposed as targets for the pharmaceu-
sometimes prepared as infusions or inhaled. The tical industry (Piomelli etal.,2000).
relative ease of purchasing herbal remedies as Traditionally used herbal medications may
compared to prescription medications and the also lead to discovery of new pathways and
perception of botanicals as safe lead in many possible pharmaceutical targets involved in
cases to their worldwide empirical use in practice the pathophysiology of various medical condi-
well before they have been stringently studied. tions. For example, as mentioned earlier, cur-
However, these obstacles should not prevent the cumin has been shown to ameliorate various
medical community from aiming to implement brain disorders, including epilepsy, depression,

FIGURE19.1: Traditional medical systems make use of mixtures of botanicals containing combinations of herbs,
and, in turn, numerous single chemical compounds.
338 Part III:Homeostatic Manipulators

and Alzheimers disease (Kulkarni and Dhir, Blume, W.T., Luders, H.O., Mizrahi, E., Tassinari,
2010). Treatment with curcumin in an animal C., van Emde Boas, W., and Engel, J. Jr. (2001).
model of epilepsy was superior to phenytoin Glossary of descriptive terminology for ictal
in alleviating memory deficits in addition to semiology: Report of the ILAE task force on
seizures and depressive symptoms. Since, in classification and terminology. Epilepsia 42,
contrast to phenytoin, curcumin also normal- 12121218.
ized brain levels of neuroepinephrine and ace- Bodalia, P.N., Grosso, A.M., Sofat, R., Macallister, R.J.,
tylcholinesterase activity (Choudhary et al., Smeeth, L., Dhillon, S., Casas, J.P., Wonderling, D.,
2013), these mechanisms could be targeted and Hingorani, A.D. (2013). Comparative efficacy
by the pharmaceutical industry in the devel- and tolerability of anti-epileptic drugs for refrac-
opment of novel antiepileptic drugs. Other tory focal epilepsy:Systematic review and network
possible targets revealed by the bioactivity meta-analysis reveals the need for long term com-
of curcumol, a volatile essential oil extracted parator trials. Br J Clin Pharmacol 76, 649667.
from the curcumin plant, are the 2 and sub- Choudhary, K.M., Mishra, A., Poroikov, V.V.,
and Goel, R.K. (2013). Ameliorative effect of
units of the glycine receptor, expressed mainly
Curcumin on seizure severity, depression like
in the hippocampus (Wang et al., 2012). The
behavior, learning and memory deficit in post-
hippocampal glycine receptors, which induce
pentylenetetrazole-kindled mice. Eur J
tonic inhibition, have been implied in the
Pharmacol 704,3340.
pathophysiology of learning and memory defi-
Eichler, S.A., Kirischuk, S., Juttner, R., Schaefermeier,
cits (Wang etal., 2012)and of epilepsy (Eichler
P.K., Legendre, P., Lehmann, T.N., Gloveli, T.,
etal., 2008). Current modern methods for stud- Grantyn, R., and Meier, J.C. (2008). Glycinergic
ying the biological actions of small molecules tonic inhibition of hippocampal neurons with
should be adapted to facilitate the study of the depolarizing GABAergic transmission elicits
mechanisms of action of herbal medicines and histopathological signs of temporal lobe epilepsy.
to find new therapeutic targets. J Cell Mol Med 12, 28482866.
Fisher, R.S., van Emde Boas, W., Blume, W., Elger, C.,
SUMMARY Genton, P., Lee, P., and Engel, J., Jr. (2005).
Treatment of CNS disorders with botanicals may Epileptic seizures and epilepsy: Definitions
re-establish homeostasis by their complementary proposed by the International League Against
multitargeted effects at the levels of mixtures, Epilepsy (ILAE) and the International Bureau for
extracts, and compounds (Figure 19.1). These Epilepsy (IBE). Epilepsia 46, 470472.
effects may be exploited for therapeutic benefit, Head, K.A., and Kelly, G.S. (2009). Nutrients and
either by use of the herbal remedies themselves botanicals for treatment of stress: Adrenal
after modern testing or by targeting new disease fatigue, neurotransmitter imbalance, anxiety,
pathways revealed by the botanicals with indi- and restless sleep. Altern Med Rev 14, 114140.
vidual compounds. Hemendinger, R.A., Armstrong, E.J., 3rd, Persinski, R.,
Todd, J., Mougeot, J.L., Volvovitz, F., and
References Rosenfeld, J. (2008). Huperzine Aprovides neu-
Barnes, P.M., Bloom, B., and Nahin, R.L. (2008). roprotection against several cell death inducers
Complementary and alternative medicine use using in vitro model systems of motor neuron
among adults and children: United States, cell death. Neurotox Res 13,4961.
2007. National Healthy Statistics Report 12 Hopkins, A.L. (2007). Network pharmacology. Nat
(Atlanta: Centers for Diease Control and Biotechnol 25, 11101111.
Prevention). National Center for Complementary and Integrative
Beal, J.E., Olson, R., Laubenstein, L., Morales, J.O., Health. (2015). https://nccih.nih.gov/
Bellman, P., Yangco, B., Lefkowitz, L., Plasse, T.F., National Institute of Neurological Disorders and
and Shepard, K.V. (1995). Dronabinol as a treat- Stroke. (2014). 2014 NINDS benchmarks for
ment for anorexia associated with weight loss in epilepsy research. http://www.ninds.nih.gov/
patients with AIDS. J Pain Symptom Manage re sea rch /epi leps y web/2 014 B ech ma rk s-Fi
10,8997. nal-PDF.pdf
Bialer, M., Johannessen, S.I., Levy, R.H., Perucca, E., Ideker, T., Galitski, T., and Hood, L. (2001). A new
Tomson, T., and White, H.S. (2009). Progress approach to decoding life: Systems biology.
report on new antiepileptic drugs:Asummary of Annu Rev Genomics Hum Genet 2, 343372.
the Ninth Eilat Conference (EILAT IX). Epilepsy Izzo, A.A., Borrelli, F., Capasso, R., Di Marzo, V.,
Res 83,143. and Mechoulam, R. (2009). Non-psychotropic
Botanicals 339

plant cannabinoids:New therapeutic opportuni- Munro, S., Thomas, K.L., and Abu-Shaar, M. (1993).
ties from an ancient herb. Trends Pharmacol Sci Molecular characterization of a peripheral recep-
30, 515527. tor for cannabinoids. Nature 365,6165.
Keith, C.T., Borisy, A.A., and Stockwell, B.R. (2005). Namjooyan,F.,Ghanavati,R.,Majdinasab,N.,Jokari,S.,
Multicomponent therapeutics for networked sys- and Janbozorgi, M. (2014). Uses of complemen-
tems. Nat Rev Drug Discov 4,7178. tary and alternative medicine in multiple sclero-
Kitano, H. (2002). Systems biology:Abrief overview. sis. J Tradit Complement Med 4, 145152.
Science 295, 16621664. Piomelli, D., Giuffrida, A., Calignano, A., and
Kulkarni, S.K., and Dhir, A. (2010). An overview of Rodriguez de Fonseca, F. (2000). The endocan-
curcumin in neurological disorders. Indian J nabinoid system as a target for therapeutic drugs.
Pharm Sci 72, 149154. Trends Pharmacol Sci 21, 218224.
Li, Q., Chen, X., He, L., and Zhou, D. (2009). Pohanka, M., Hrabinova, M., Zemek, F., Drtinova,
Traditional Chinese medicine for epilepsy. L., Bandouchova, H., and Pikula, J. (2011).
Cochrane Database Syst Rev, CD006454. Huperzine induces alteration in oxidative bal-
Li, S., and Zhang, B. (2013). Traditional Chinese ance and antioxidants in a guinea pig model.
medicine network pharmacology:Theory, meth- Neuro Endocrinol Lett 32 Suppl 1, 95100.
odology and application. Chin J Nat Med 11, Rafii, M.S., Walsh, S., Little, J.T., Behan, K., Reynolds,
110120. B., Ward, C., Jin, S., Thomas, R., and Aisen, P.S.
Li, Y., and Hu, G.Y. (2002a). Huperzine A, a noot- (2011). A phase II trial of huperzine A in mild
ropic agent, inhibits fast transient potassium to moderate Alzheimer disease. Neurology 76,
current in rat dissociated hippocampal neurons. 13891394.
Neurosci Lett 324,2528. Ross, H.R., Napier, I., and Connor, M. (2008).
Li, Y., and Hu, G.Y. (2002b). Huperzine A inhibits Inhibition of recombinant human T-type cal-
the sustained potassium current in rat dissoci- cium channels by Delta9-tetrahydrocannabinol
ated hippocampal neurons. Neurosci Lett 329, and cannabidiol. J Biol Chem 283, 1612416134.
153156. Ryan, D., Drysdale, A.J., Lafourcade, C., Pertwee,
Ma, T., Gong, K., Yan, Y., Zhang, L., Tang, P., Zhang, R.G., and Platt, B. (2009). Cannabidiol targets
X., and Gong, Y. (2013). Huperzine Apromotes mitochondria to regulate intracellular Ca2+ lev-
hippocampal neurogenesis in vitro and in vivo. els. J Neurosci 29, 20532063.
Brain Res 1506,3543. Sams-Dodd, F. (2013). Is poor research the cause of
Ma, X., Tan, C., Zhu, D., Gang, D.R., and Xiao, P. the declining productivity of the pharmaceuti-
(2007). Huperzine Afrom Huperzia speciesan cal industry? An industry in need of a paradigm
ethnopharmacolgical review. J Ethnopharmacol shift. Drug Discov Today 18, 211217.
113,1534. Schachter, S.C. (2009). Botanicals and herbs:Atra-
Ma, Y.L., Weston, S.E., Whalley, B.J., and Stephens, ditional approach to treating epilepsy.
G.J. (2008). The phytocannabinoid Delta(9)- Neurotherapeutics 6, 415420.
tetrahydrocannabivarin modulates inhibi- Schmidt, D., and Schachter, S.C. (2014). Drug treat-
tory neurotransmission in the cerebellum. Br J ment of epilepsy in adults. BMJ 348,g254.
Pharmacol 154, 204215. Sucher, N.J. (2006). Insights from molecular inves-
Margineanu, D.G. (2013). Systems biology, complex- tigations of traditional Chinese herbal stroke
ity, and the impact on antiepileptic drug discov- medicines:Implications for neuroprotective epi-
ery. Epilepsy Behav. 38, 131142. lepsy therapy. Epilepsy Behav 8, 350362.
Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, Sucher, N.J. (2013). The application of Chinese medi-
A.C., and Bonner, T.I. (1990). Structure of a can- cine to novel drug discovery. Expert Opin Drug
nabinoid receptor and functional expression of Discov 8,2134.
the cloned cDNA. Nature 346, 561564. Tang, X.C., De Sarno, P., Sugaya, K., and Giacobini,
Mechoulam, R., and Ben-Shabat, S. (1999). E. (1989). Effect of huperzine A, a new cholines-
From gan-zi-gun-nu to anandamide and terase inhibitor, on the central cholinergic sys-
2-arachidonoylglycerol: The ongoing story of tem of the rat. J Neurosci Res 24, 276285.
cannabis. Nat Prod Rep 16, 131143. Tian, G.X., Zhu, X.Q., Chen, Y., Wu, G.C., and
Mechoulam, R., and Hanu, L. (2001). The cannabi- Wang, J. (2013). Huperzine Ainhibits CCL2 pro-
noids: An overview. Therapeutic implications duction in experimental autoimmune encepha-
in vomiting and nausea after cancer chemo- lomyelitis mice and in cultured astrocyte. Int J
therapy, in appetite promotion, in multiple scle- Immunopathol Pharmacol 26, 757764.
rosis and in neuroprotection. Pain Res Manag Wang, L., Li, W.G., Huang, C., Zhu, M.X., Xu, T.L.,
6,6773. Wu, D.Z., and Li, Y. (2012). Subunit-specific
340 Part III:Homeostatic Manipulators

inhibition of glycine receptors by curcumol. J Zhang, G.B., Li, Q.Y., Chen, Q.L., and Su, S.B.
Pharmacol Exp Ther 343, 371379. (2013). Network pharmacology: A new
Wang, Z.F., Wang, J., Zhang, H.Y., and Tang, X.C. approach for chinese herbal medicine research.
(2008). Huperzine Aexhibits anti-inflammatory Evid Based Complement Alternat Med 2013,
and neuroprotective effects in a rat model of 621423.
transient focal cerebral ischemia. J Neurochem Zhang, H.Y., Yan, H., and Tang, X.C. (2008).
106, 15941603. Non-cholinergic effects of huperzine A:Beyond
Yu, D., Thakor, D.K., Han, I., Ropper, A.E., Haragopal, inhibition of acetylcholinesterase. Cell Mol
H., Sidman, R.L., Zafonte, R., Schachter, S.C., and Neurobiol 28, 173183.
Teng, Y.D. (2013). Alleviation of chronic pain fol- Zhang, J.M., and Hu, G.Y. (2001). Huperzine A,
lowing rat spinal cord compression injury with a nootropic alkaloid, inhibits N-methyl-D-
multimodal actions of huperzine A. Proc Natl aspartate-induced current in rat dissociated hip-
Acad Sci USA 110, E746E755. pocampal neurons. Neuroscience 105, 663669.
20
The Role ofAcupuncture
inNociception Homeostasis
N A N N A G O L D M A N , TA K A H I R O TA K A N O , B E N J A M I N T. K R E S S ,
AND MAIKEN NEDERGA ARD

CHALLENGES AND PROGRESS to result in diseases (Yang etal., 2011), however


I N U N D E R S TA N D I N G the metaphysical nature of this hypothesis has
ACUPUNCTURE hindered acupuncture from being the subject of
Acupuncture is a form of traditional Chinese empiricalstudy.
medicine that has been practiced for more than The evolving political climate in China also
4,000 years. Despite widespread use, however, contributed to the lack of information about
there is a lack of understanding of how acu- the biological mechanisms of acupuncture.
puncture derives its therapeutic efficacy. This Due to widespread epidemics of infectious dis-
is true in part because the indications for acu- eases and the comparative success of Western
puncture treatment are exceedingly diverse medicine, particularly at promoting notions of
and may include wide-ranging applications for public hygiene, the Kuomintang government
gastrointestinal (e.g., heartburn) dermatologic in China outlawed many traditional Chinese
(e.g., acne), or psychiatric disorders (e.g., anxi- medical practices in the early 20th century and,
ety, insomnia, or depression), among many oth- in 1929, closed schools devoted to the teaching
ers. The most common use of acupuncture is for of traditional Chinese medicine (Veith, 1973;
the relief from pain. Although the exact protocol Yang et al., 2011). This decision resulted in at
often varies depending on the practitioner, the least two decades of official abandoning of acu-
treatment for pain typically utilizes a fine needle puncture. It was not until Mao Tse-Tung and the
that is inserted a depth of ~1 to 2inches into spe- Communist Party rose to power in 1949 that the
cific points (acupoints) on the patients body. The use of traditional Chinese medicine regained
needle is then intermittently rotated to initiate widespread popularity. However, in order to
acupunctures analgesic properties, or, in certain limit the growing influence of Western powers
variations, heat or a low-grade electric current over Chinese culture, the Communist Party did
are applied to the needles in order to amplify or not encourage objective studies of acupuncture,
prolong the treatments therapeutic efficacy. which in turn created a distrust in the Western
Many historical events contribute to why so world toward acupunctures therapeutic effi-
little is known about the pathways involved in cacy. Several clinical studies have addition-
acupuncture-mediated analgesia. Acupuncture is ally reported a lack of benefit of acupuncture
based on the philosophical principles of Eastern over placebo (Moore and Berk, 1976;Petrie and
medicine that traditionally do not search for bio- Hazleman, 1986), which has further hindered
logical mechanisms as underlying causes of dis- the integration of acupuncture into Western
ease. For example, the anatomical and functional medical practices.
distinctions of acupoints are based on the ancient Other studies, however, support the clini-
theory of communication channels, called merid- cal benefits of acupuncture, including those
ians, that are said to facilitate transmission of the conducted in experimental chronic pain
bodys energy source, or Qi. According to this animal models, which have the advantage
theory, it is critical to have a balance of energy of lacking potentially confounding placebo
streaming through the major 12 meridians in the effects (Cantwell, 2010; Koski, 2011). A recent
body. Blockade of such energy flow is thought meta-analysis of 35 eligible randomized
342 Part III:Homeostatic Manipulators

controlled trials indicates that acupuncture has received the greatest attention in the field
provides a significant benefit over placebo when of pain research due to its widely distributed
used for treatment of chronic neck, back, or expression pattern in the dorsal spinal cord,
osteoarthritis pain (Vickers et al., 2012). Such especially in lamina II (substantia gelatinosa;
data have contributed to the growing popular- Choca et al., 1988; Horiuchi et al., 2010). In
ity of acupuncture and the growing recognition this region, most afferent sensory nerves
of its therapeutic value by Western agencies make connections with postsynaptic neurons.
such as the World Health Organization and Similarly important for antinociception is the
the US National Institutes of Health (NIH) localized expression pattern of A1 receptors in
(World Health Organization, 2003). For the descending projection of the dorsal horn
example, acupuncture is currently practiced (Choca etal.,1988).
in more than 160 countries around the world In both the peripheral and central nervous
(Zhao, 2008), and in a consensus statement in system, the endogenous release of adenosine
1997 the NIH recognized that acupuncture inhibits pain transmission by inhibiting the
may serve as an adjunct treatment or accept- activity of nociceptive pathways (Guo et al.,
able alternative therapy for postoperative den- 2013; Taylor, 2009), and many groups have
tal pain, headache, menstrual cramps, tennis identified A1 receptors as the main adenosine
elbow, fibromyalgia, myofascial pain, osteoar- receptor involved in the analgesic actions of
thritis, carpal tunnel syndrome, and low back adenosine (Burnstock, 2007; Sawynok, 1998;
pain (Acupuncture, 1997). According to the Sawynok and Liu, 2003). Interestingly, manipu-
NIH National Center for Complementary and lations such as depolarization (high K+) or the
Alternative Medicine, the 2007 National Health application of agonists, including capsaicin,
Interview Survey revealed that an estimated substance P, N-methyl-D-aspartate (NMDA),
3.1million US adults and 150,000 children used morphine, or serotonin (5-HT), have all been
acupuncture in 2006. Nevertheless, the lack of shown to trigger elevations in extracellular
a defined mechanistic basis of acupuncture adenosine concentrations (Latini and Pedata,
has led to skepticism by physicians who do not 2001), suggesting that adenosine release may
subscribe to its underlying philosophical prin- constitute a physiological regulator of pain
ciples. In particular, the absence of verifiable, signaling. In support of this concept, acti-
anatomical acupoints or meridians represents vation of peripheral and central vanillinoid
a major conceptual barrier to Western medi- receptors (TRPV1) among primary afferent
cine (Ahn et al., 2008). As a result, until very C-fibers has been shown to trigger adenosine
recently, acupuncture has not been the focus of release that attenuated inflammatory and ther-
rigorous empirical study, and thus is not com- mal pain transmission directly by inhibiting
monly recommended as a first-line treatment. TRPV1-mediated Ca 2+ entry into dorsal root
Consequently, the general public is forced to ganglion neurons (Puntambekar et al., 2004).
seek treatment from potentially underquali- The results of that study also reported that
fied practitioners, in many cases under uncon- enhanced capsaicin concentrations overcame
trolled conditions. Defining the mechanistic the adenosine-induced inhibition of nocicep-
basis for acupuncture-mediated analgesia is tive signaling, suggesting that adenosine and
thus a critical step for its acceptance to the capsaicin exert competitive inhibition of the
health-care system and also an essential step TRPV1 receptor. The response profile to cap-
for improving its safety and clinical benefits. saicin is particularly interesting because it sug-
Although skepticism remains, California and gests that agonists of TRPV1 and adenosine
Maryland recently mandated acupuncture cov- exert homeostatic control over nociceptive
erage in all health insurance plans for individu- signaling, presumably to facilitate rapid acti-
als and small groups. vation (detection and propagation of a noxious
thermal or inflammatory stimulus) followed
ANALGESIC ACTIONS by rapid reset, respectively, of the nociceptive
OFADENOSINE circuit (to maintain the functional sensory unit
Adenosine, a purine nucleoside, has potent and prepare for detection of future threats).
antinociceptive properties in both animals In further support of this concept, systemic
and humans. Adenosine signals through administration of various A1 receptor agonists
four distinct receptors: A1, A 2A, A 2B, and A 3. has been shown to produce analgesic effects
Of the adenosine receptors, the A1 receptor in a variety of acute pain models in animals
The Role of Acupuncture in Nociception Homeostasis 343

(Gong etal., 2010). For example, several studies ADENOSINE SIGNALING


show that intrathecal injection of A1 receptor I S A K E Y M E D I AT O R
agonists induced analgesia in various animal OFTHE ANALGESIC ACTIONS
models of acute pain. These tests include tail OFACUPUNCTURE
flick, tail immersion, hot-plate, formalin, acetic While most investigations of the biological mech-
acid, capsaicin models, and others (Nascimento anisms of acupuncture have focused on central
et al., 2010; Song, 2011; Zahn et al., 2007). endorphin signaling (Han, 2004;Neumann etal.,
Similarly, mice lacking the A1 receptor (knock- 1996; Zhao, 2008), other lines of work indicate
out animals) exhibit a lower pain threshold in that acupuncture produces analgesia by act-
hyperalgesia tests (Wu et al., 2005). Further, ing locally (Melzack et al., 1977). For example,
the analgesic effect of intrathecal adenosine acupuncture-induced analgesia is most potent
administration was abolished in A1 receptor when applied close to rather than distant to
knockout mice (Johansson etal.,2001). the locus of pain and normally restricted to the
In addition to suppressing acute pain trans- ipsilateral side (Lao et al., 2004; WM Li, 2005),
mission, agonists of A1 receptors have also been pointing to local mechanisms. In addition, many
shown to reduce chronic pain (Figure 20.1). acupoints reside along the major nerves (Gunn
The agonist R-PIA inhibits mechanical allo- et al., 1976). In fact, a systematic analysis con-
dynia induced by spinal nerve ligation in cluded that 70% of acupoints are found either
rats (Hwang et al., 2005; Song, 2011). R-PIA along the pathways of major peripheral nerves,
also increases the heat pain threshold in rats superficial branches of peripheral nerves, or at
that underwent an injury in the spinal cord sites where cutaneous nerves emerge from the
(Horiuchi et al., 2010). Another selective A1 deep fascia (Bossy, 1979). For example, Zusanli
receptor agonist, CPA, inhibits pain induced by (ST36), a widely used and studied acupoint, is
arthritis or neuropathy in rats (Curros-Criado located above the deep peroneal nerve, and, upon
and Herrero,2005). stimulation, the needle tip lands in close proxim-
ity to the nerve fiber (Yang etal.,2007).
Multiple lines of work have documented
that adenosine triphosphate (ATP) is released
in response to either mechanical and electrical
stimulation or heat (Sauer et al., 2000; White
Spinal cord etal., 1982). Once released, ATP acts as a trans-
mitter that binds to purinergic receptors, includ-
Systemic
ing P2X and P2Y receptors (Abbracchio et al.,
Acupoint 2009; Burnstock, 2007). ATP is not transported
back into the cell but rather is rapidly degraded to
Site of pain adenosine, inosine, or IMP by ectonucleotidases
or deaminases before reuptake (Burnstock, 2007).
Based the literature discussed previously, it was
hypothesized that the mechanical stimulation
FIGURE 20.1: Site in which injection of A1 receptor over the unique anatomical sites in which acu-
agonists have been documented to have an analgesic puncture is commonly applied leads to increased
effect in chronicpain. levels of extracellular adenosine, which in turn
Intratecal administration (Curros-Criado and Herrero, mediates the analgesic actions of acupuncture.
2005; Horiuchi etal., 2010; Hwang etal, 2005; Johansson The first evidence in support of this hypothesis
etal., 2001; Song etal., 2011; Zahn etal.,2007)
was documented in a series of experiments con-
Systemic administration (Gong etal., 2010; Nascimento
ducted by the Nedergaard lab group (Goldman
etal.,2010)
et al., 2010). To determine whether adenosine
Acupoints (Goldman etal., 2010; Hurt and Zylka,2010)
was involved in the antinociceptive effects of acu-
Local administration of adenosine receptor agonist at the
puncture, Goldman and colleagues first asked
locus of pain. A1 receptor agonists have analgesic effects
whether the extracellular concentration of aden-
while A2 receptors mediate proalgesic effects (Karlsten
osine increases during the needle insertion and
etal., 1992; Taiwo and Levine,1990).
gentle manipulation associated with a typical
The agonists in all studies were injected locally, except acupuncture treatment. The results showed that,
for the systemic administration in which the agents in mice, acupuncture triggered a localized release
were delivered intravascular. of tri-, di-, and mono-phosphorylated adenosine
344 Part III:Homeostatic Manipulators

nucleotides (ATP, ADP, and AMP) in addition to knockout mice and wild-type mice. As a result
adenosine. This was consistent with the obser- of the CFA injection, both groups of mice devel-
vation that tissue damage is associated with an oped mechanical allydonia (Von Frey filament
increase in extracellular nucleotides and adeno- touch test) and thermal hyperalgesia (thermal
sine (Fredholm, 2007). The authors acquired test). When the adenosine A1 receptor ago-
direct confirmation of their hypothesis by collect- nist CCPA was locally injected into the Zusanli
ing samples of interstitial fluid by a microdialysis acupoint, it produced transient antinociceptive
probe implanted in the tibialis anterior muscle/ effects reflected by a reduction of both mechani-
subcutis of adult mice at a distance of ~0.5mm cal and thermal hypersensitivity. However, in A1
from the Zusanli point, which is located below receptor knockout mice, CCPA failed to reduce
and slightly lateral for the midline of the knee pain. These observations indicated that A1 recep-
(Zhao, 2008). Adenine nucleotides and adeno- tor expression was necessary for CCPA-mediated
sine were quantified using high-performance pain suppression in inflammatory pain.
liquid chromatography before, during, and after Neuropathic pain was modeled using the ligation
acupuncture (Cui etal., 2010;Volonte etal., 2004). injury of the sciatic nerve (Vadakkan etal., 2005),
At baseline, the concentrations of ATP, ADP, in which pain peaked 5 to 7days after nerve liga-
AMP, and adenosine were in the low nanomolar tion. CCPA injected in the Zusanli point of the
range, as previously reported (J Li etal., 2003; J ipsilateral leg reduced neuropathic pain with an
Li etal., 2005). Acupuncture applied by manual efficacy that was comparable to its suppression
rotation of the acupuncture needle every 5 min of inflammatory pain (Figure 20.2c, d). In each
for a total of 30 min sharply increased the extra- model, the antinociceptive effect of CCPA was
cellular concentrations of all purines detected. transient and consistent with a local mechanism
Adenosine concentration increased 24-fold dur- of pain suppression, as it did not alter sensitivity
ing the 30-min acupuncture session. to painful stimulation in the contralateral leg. In
The critical role of adenosine in mediating addition, injection of CCPA into the contralat-
the analgesic effects of acupuncture was further eral leg did not alter the pain threshold of the
tested by locally injecting the selective A1 recep- ipsilateral leg. These experiments revealed that
tor agonist, 2-chloro-N(6)-cyclopentyladenosine localized stimulation of A1, whether through an
(CCPA; Lohse etal., 1998)in two murine mod- agonist or via acupuncture, alleviated symptoms
els of either inflammatory pain or neuropathic associated with inflammatory pain and neuro-
pain (Goldman etal., 2010). Inflammatory pain pathic pain (Figure20.2).
was assessed by quantification of the withdrawal Goldman and colleagues observations indi-
threshold from a thermal or tactile stimulus cated that the antinociceptive action of acupunc-
following the injection of complete Freunds ture is mediated by activation of A1 receptors
adjuvant (CFA) into the right paw of both A1 located on ascending nerves. Thus medications

Acupuncture

Autocrine action to fibroblasts


ATP

ATP AMP Adenosine A1R


ADP
IMP Inosine
Fibroblasts Nociceptive
Skeletal muscle nerve

FIGURE 20.2: Proposed mechanism of acupuncture-induced analgesia. Insertion and rotation of acupuncture
needle triggers release of adenosine triphosphate (ATP) from tissue fibroblasts and skeletal muscle induces release
of ATP. Extracellular ATP is in part converted to adenosine and activates A1 receptors. ADP=adenosine diphph-
sphate, AMP=adenosine monophosphate, IMP=inosine monophosphate.
The Role of Acupuncture in Nociception Homeostasis 345

that interfere with A1 receptors or adenosine to the small body weight of mice. Takano and
metabolism may improve the clinical benefit of colleagues, also from the Nedergaard lab group,
acupuncture. For example, it has been previously addressed this important question directly
shown that inhibition of adenosine metabo- by collecting microdialysis samples of inter-
lism prolongs the biological effects of adenosine stitial fluid before, during, and after deliver-
(Sawynok, 2007). In accordance with this find- ing 30 min of conventional acupuncture in the
ing, the Nedergaard group hypothesized that Zusanli point in human volunteers (Takano
the inhibition of adenosine deaminase would et al., 2012). Similar to the findings from mice,
prolong the beneficial effects of acupuncture. the analysis showed that the interstitial adeno-
The concept was tested using an Food and Drug sine concentration increased significantly dur-
Administrationapproved adenosine deaminase ing acupuncture and remained elevated for
inhibitor, deoxycoformycin. Combined injections 30 min after the acupuncture (Figure 20.3;
of deoxycoformycin with acupuncture resulted Takano et al., 2012). Interestingly, the analy-
in antinociceptive effects that lasted 2.0 to 2.5 hrs sis showed that acupuncture not only had to be
longer than acupuncture alone (Goldman etal., delivered at the Zusanli point in order to achieve
2010). In addition to further proving that acu- analgesia but that the needle had to be rotated to
puncture acts via adenosine receptors, the results evoke significant adensoine release (Figure 20.3).
of these studies imply that it is possible to extend Acupuncture-mediated adenosine release was
the clinical utility of acupuncture as a safe and not observed if acupuncture was delivered in tis-
effective treatment for pain. The observations sue outside the Zusanli point (Figure20.3).
may be particularly important for the manage- Other studies of acupuncture have sug-
ment of patients with chronic pain who require gested that the analgesic properties may depend
long-term treatment, as expanding the duration on the proximity not only to peripheral nerves
of acupunctures analgesic effects could require but also to regions of enhanced vascularization.
fewer sessions to achieve continued therapeutic Consistent with this concept, a recent study of
benefits. acupoints suggested that the ST36 (Zusanli) and
ST37 (Shangjuxu) acupuncture points exhibit
ACUPUNCTURE TRIGGERS distinct structural characteristics with a higher
RELEASE OFADENOSINE complexity of the local vasculature (C Liu etal.,
INHUMAN SUBJECTS 2014). X-ray phase contrast imaging in this study
An open question is whether acupuncture per- revealed that at the acupuncture points, the
formed by an experienced practitioner, and microvascular bed included multiple larger size
according to standard technique, similarly trig- vessels (~50100 m in diameter) with bifurca-
gers the release of adenosine in human subjects. tions and smaller vessels (1550m in diameter),
The possibility existed that adenosine release whereas nonacupuncture point areas exhib-
in mice exposed to acupuncture was an experi- ited few larger vessels and essentially none of
mental artifact, since the acupuncture needles the smaller vessels. Combined with our finding
utilized were disproportionally large compared that robust adenosine release and subsequent

Adenosine
Percent change from baseline

A B AMP
C # Adenosine
250 # AMP
I. Acupoint with/ * ADP 250
* ATP ADP
without rotation 200 **
Percent change

* 200 ATP
II. Non-acupoint *
150 150
with rotation
100 100
Microdialysis
probe 50 50
0 0
Baseline Recovery 1 Acupuncture Without Non-acupoint
Acupuncture Recovery 2 rotation

FIGURE20.3: Acupuncture induces release of adenosine in human subjects. (A)Extracellular purines were col-
lected by a microdialysis probe while acupuncture session was given. (B)Transient increase of tissue adenosine by
acupuncture in human. (C)Adenosine increase was observed only when a needle was inserted in an acupoint and
rotated (Takano etal., 2012). AMP=adenosine monophosphate, ADP=adenosine diphphsphate, ATP=adeno-
sine triphosphate.
346 Part III:Homeostatic Manipulators

analgesia were detected if the acupuncture nee- connexin 43, which mediates purine release in
dle was inserted and rotated in the Zusanli multiple cell types (Kang etal., 2008;Wilgenbus
point, whereas no analgesia was detected when et al., 1992). Subsequent studies showed that
the needle was simply inserted without rota- the acupuncture-induced shape change is also
tion, these observations suggest that the proxim- associated with a sustained release of ATP from
ity of acupuncture placement to the peripheral fibroblasts and is dependent of P2Y purinergic
nerves and to regions of dense vascularization receptors (Goldman etal., 2013; Langevin etal.,
are important factors that influence the analgesic 2013). However, it appears that the changes in
properties of acupuncture. They further suggest fibroblast shape are not mediated by adenosine,
that an important determinant of acupunctures since A1 receptor antagonists failed to inhibit
analgesic benefits may be the mode of the applied cytoskeletal reorganization induced by ATP
stimulus, such as the tissue stretching that results (Goldman etal.,2013).
from needle rotation, which may offer further The same cellular cascade can be observed
insights into the analgesic properties of other by simply stretching the tissue for approximately
alternative therapies. These potentially include 30 min, about the same time duration of needle
certain forms of therapeutic massage, such as the retention during a typical acupuncture treat-
practice known as acupressure, which involves ment. Interestingly, and perhaps holding impor-
mechanical manipulation of the acupoints with- tant implications for enhancing the therapeutic
out the need for needle insertion. benefit of acupuncture, the work by Langavin
etal. also found that letting go of the needle after
O T H E R P U R I N E R G I C - M E D I AT E D needle rotation does not cause the tissue to imme-
SIGNALING MECHANISMS diately unwind from the needle. The whorl of
A C T I VAT E D B Y A C U P U N C T U R E connective tissue remains intact as long as the
Goldman and Takanos studies of experimental needle remains under the skin, causing the tissue
acupuncture in mice and traditional acupunc- to be stretched for a prolonged period. Langevins
ture in humans strengthened the role of adeno- team found that the tissue changes with acu-
sine in acupuncture-mediated antinociception. puncture are associated with a large-scale relaxa-
However, neither study established the source or tion of the connective tissue wherein fibroblasts
mechanism of adenosine release. Work done by initiated a specific Rho-dependent cytoskeletal
Helene Langevin has made major strides in this reorganization that was required for full tissue
area and has greatly expanded what we know relaxation. Rho is an intracellular signaling mol-
today about the cellular response to acupuncture ecule known to play a role in cell motility and the
needle insertion. Langevin et al. first observed remodeling of cell-surface proteins that connect
that acupuncture points seem to be mainly located the fibroblast to its surrounding matrix.
along connective-tissue planes between muscles, Altogether, multiple lines of work have doc-
or between muscle and bone. Her analysis showed umented that the extracellular concentration
that more than 80% of acupuncture points in the of ATP, ADP, AMP, and adenosine increases
arm are located along connective-tissue planes locally in tissue exposed to traditional mechan-
(Langevin and Yandow, 2002). Langevins team ical acupuncture. While adenosine and A1
next observed that the fibroblasts residing in the receptor activation play a key role in the anal-
connective tissue, as far as several centimeters gesic action of acupuncture (Goldman et al.,
away from the needle, reorganized their internal 2010), indirect evidence points to the impor-
cytoskeleton and changed shape upon insertion tance of P2Y receptor mediated cytoskeletal
of the acupuncture needle, becoming large and remodeling of fibroblasts (Goldman etal., 2013;
flat (Langevin etal., 2007). The analysis showed Langevin etal., 2013). Similarly, P2X receptors
that fibroblasts in connective tissue respond to are regarded as pain receptors, and both P2X4
static stretching of the tissue by expanding and and P2X3 receptor antagonists have shown
remodeling their cytoskeleton within minutes promising effects in preclinical and clinical
both ex vivo and in vivo. In search of a mecha- studies (Burnstock, 2013; Tsuda et al., 2013).
notransduction mechanism, Langevin et al. Acupuncture is usually not linked to pain sen-
tested the hypothesis that the mechanism of sation, likely because the manipulation of the
fibroblast expansion in response to tissue stretch needles is slow and gentle, such that ATP con-
involves extracellular ATP signaling. In support centration does not rise fast enough to over-
of this concept, fibroblasts express high levels of come desensitization and reach the threshold
The Role of Acupuncture in Nociception Homeostasis 347

A B 100 Control
Deoxycoformycin
ATP Adenosine 80

Touch test (%)


release release
60 ** ** **
** ** ** **
PAP **
A1R 40
CD39 CD73 cAMP
ATP AMP Adenosine PKA 20
Deaminases
ADP 0
IMP
T 0.5h 1h 1.5h 2h 2.5h 3h 3.5h 4h
Inosine
DCF Before acupuncture
Before CFA

FIGURE 20.4: Ectonucleotidase activity. (A) Extracellular adenosine can be produced from the breakdown
of adenosine triphosphate (ATP) by CD39 and subsequently by prostatic acid phosphatase (PAP) or CD73.
Deoxycoformycin (DCF) blocks AMP/adenosine deaminases. (B)Administration of DCF prolonged the analgesic
effect of acupuncture (Goldman etal., 2010). ADP=adenosine diphosphate, AMP=adenosine monophosphate,
IMP=inosine monophaosphate, PKA=protein kinase, ACFA=complete Freunds adjuvant.

for activation of P2X receptors. Excessive nee- analgesic effect of acupuncture was prolonged
dle handling, however, will trigger pain, which (Figure 20.4B; Goldman et al., 2010). In an ele-
may reflect a consequence of higher amplitude gant series of experiments, Mark Zylkas group
(suprathreshold) increases in interstitial ATP recently showed that local injection of prostatic
that then exceed the capacity for degradation to acid phosphatase, which hydrolyses a phosphate
ATPs antinociceptive purine derivatives. Thus group from a wide variety of proteins and other
acupuncture appears to exert therapeutic prop- biomolecules including AMP, attenuated chronic
erties by capitalizing on innate homeostatic pain for 3 days without acupuncture treatment,
mechanisms of purine-mediated nociceptive suggesting that endogenous extracellular AMP
activation and inhibition, such as evoking the is a sufficient source for production of analgesic
controlled release of ATP and its inhibitory levels of adenosine (Hurt and Zylka,2012).
degradation products. Acupuncture treatment for chronic pain
is typically given at multiple sessions to pro-
LOCAL INJECTION OF duce longer lasting reductions in pain (Guerra
ECTONUCLEOTIDASES: de Hoyos et al., 2004;Thomas et al., 2005).
A POTENTIAL NOVEL Acupuncture used in this manner is interest-
A N T I N O C I C E P T I V E S T R AT E G Y ing because long-lasting activation of A1 recep-
Activation of A1 receptors by endogenous adeno- tors may activate downstream processes that
sine has been implicated as key step in mediating counteract inflammation and neural plasticity
actupunctures analgesic properties. In light of that is associated with chronic pain. Adenosine
this, molecular targets important for purinergic has in multiple lines of work been shown to
metabolism may be highly relevant for the treat- have potent anti-inflammatory roles (Flogel
ment of pain. Adenosine is primarily metabo- etal., 2012; Hasko and Pacher, 2008; Ohta and
lized by adenosine deaminase, which removes an Sitkovsky, 2001; Sawynok, 1998). Adenosine
amine group from adenosine to form inosine in interacts with specific G protein-coupled recep-
the subcutis and muscles, (Figure 20.4a; Cunha tors on inflammatory and immune cells to
and Sebastiao, 1991). AMP is readily converted regulate their function, and injurious stimuli,
to IMP by AMP deaminase, while the kinetics ischemia, and inflammation all result in release
of the dephosphorylation of AMP to adenosine of adenosine in tissue (Fredholm, 2007). The
are slow (Goldman et al., 2010). IMP, inosine, released adenosine then acts as an immunosup-
and AMP are not ligands for purine receptors, pressant. For example, mice that were deficient
as these degradation pathways quickly eliminate in A2 receptors showed increased inflamma-
adenosine. Of the purine ligands, only adenosine tion tissue damage (Ohta and Sitkovsky, 2001).
can activate the A1 receptor, albeit a recent report Novel insights into how acupuncture achieves
suggested that AMP also binds to A1 receptors therapeutic benefits open up new strategies for
(Rittiner et al., 2012). When deoxycoformy- treating painsuch as manipulation of local
cin was locally administered to inhibit deami- adenosine signalingthat do not necessarily
nase activity and adenosine metabolism, the involve acupuncture.
348 Part III:Homeostatic Manipulators

POTENTIAL CONFOUNDING countries. Mice received plantar incision surgery


EFFECT OFCAFFEINE and were then treated with acupuncture nee-
Caffeine is a nonspecific antagonist of adenosine dling after administration of acute and chronic
receptors that is present in a wide variety of food caffeine. For the chronic preadministration of
and beverages (Marchand etal., 1995). It is esti- caffeine, two different doses of caffeine were
mated that 90% of the population worldwide con- administered, one that mimicked the average
sumes caffeine daily, making it the most widely daily consumption in Western countries and one
consumed psychoactive drug (Frary etal., 2005). that mimicked the average daily consumption in
The popularity of caffeine can be attributed to its China. The analysis showed that acute preadmin-
stimulatory effects, including wakefulness and istration of caffeine (10mg/kg, i.p.) completely
enhanced mood (Haskell etal., 2005; Lieberman reversed acupuncture analgesia in both mechan-
et al., 2002). Caffeines high blood-brain bar- ical and electro-acupuncture. Interestingly, the
rier permeability allows it to readily enter the Western dose of caffeine (70mg/kg/day) admin-
central nervous system, where it then produces istered during 8days in the drinking water also
psychoactive effects largely through downstream reversed acupuncture analgesia. However, the
consequences of interfering with noradre- Chinese dose (4mg/kg/day) administered during
negic, dopaminergic, or serotonergic signaling the same period did not reverse the effects of acu-
via A1 receptor activation (Nehlig et al., 1992). puncture (Mor et al., 2013). It is interesting to
Neurons in these nuclei are highly sensitive to note that the number of studies in China favoring
caffeine and their metabolic activity activated acupuncture treatment is much higher than in
after intake of just 1 mg/kg caffeine (Nehlig etal., Western countries. Daily caffeine consumption
1992). Recently, positron emission tomography is a plausible explanation for this discrepancy.
was used to visualize binding sites of caffeine in
the human brain, and the analysis established CONCLUSION
that there was 50% occupancy of the brains Chronic pain, often defined as pain persist-
A1 adenosine receptors when individuals con- ing longer than six months, is a heterogeneous
sumed caffeine throughout the day (Elmenhorst group of medical conditions that have significant
etal.,2012). impact on an individuals physical, emotional,
The relationship between caffeine and acu- psychological, and financial distress. According
puncture is intriguing because it could explain to an NIH estimate in 2001, more than 50million
why a number of clinical trials have found that Americans suffer from chronic pain, and approx-
acupuncture is not always effective. Caffeines imately 45% of the population visit clinical offices
competition with adenosine binding at the A1 for pain at some point in their lives (National
receptor would interfere with the antinociceptive Institutes of Health, 2001). Although the anal-
mechanism of acupuncture. However, the bio- gesic effect of acupuncture for the treatment of
logical effects of caffeine are complex, making it chronic pain is well documented (Kelly 2009;
difficult to predict its effect on pain. For example, Weidenhammer etal., 2007; Zhao, 2008), its bio-
caffeine has analgesic effects at nonphysiologi- logical basis is not well understood, and it has
cal high concentrations and may, at lower doses, only recently been the subject of extensive study.
enhance efficacy of other analgesics by actions Clear benefits of acupuncture set the procedure
not related to the A1 receptors. For example, caf- apart from traditional analgesic treatments,
feine can improve drug absorption, reduce drug which include nonsteroidal anti-inflammatory
metabolic clearance, activate norepinephrine agents (such as ibuprofen and naproxen), and
signaling, and alter mood and alertness (Derry opioids (such as morphine and codeine). Aspirin
et al., 2012;Sawynok and Yaksh, 1993). Caffeine and other nonsteroidal anti-inflammatory agents
may also indirectly interfere with the antinoci- are generally ineffective in treating neuropathic
ceptive action of several agents such as amitrip- pain (Portenoy, 2000). While they are effective
tyline and oxcarbazepine by blocking the A1 in inflammatory pain states, there is a ceiling
receptor (Sawynok,2011). effect to the pain relief they provide, and adverse
A recent study revealed very interesting effects to the gastric system may become a serious
dose-dependent interactions when caffeine health concern with high doses or with long-term
is combined with both manual and electro- use (Ashburn and Staats, 1999; Mukherjee etal.,
acupuncture. Mor and colleges took into 2001; Ong etal., 2007; Portenoy, 2007). Opioids
account that the average daily caffeine consump- are frequently used for cancer pain manage-
tion in China is 20 times lower than Western ment, but due to the risks of increased tolerance,
The Role of Acupuncture in Nociception Homeostasis 349

dependency, and psychological and hormonal Burnstock G. Purinergic mechanisms and painan
adverse effects, many providers are now ques- update. Eur J Pharmacol 2013;716(13):2440.
tioning the expanded role of long-term opioid Cantwell SL. Traditional Chinese veterinary medi-
therapy (Ballantyne and Mao, 2003;Noble etal., cine: The mechanism and management of acu-
2010; Rowbotham etal., 2003). Thus the lack of puncture for chronic pain. Top Companion
alternative approaches to treat chronic pain is a Anim Med 2010;25(1):5358.
large societal problem. Choca JI, Green RD, Proudfit HK. Adenosine A1
Acupuncture is an effective and relatively and A2 receptors of the substantia gelatinosa are
cheap procedure that in controlled clinical stud- located predominantly on intrinsic neurons:An
ies has been shown to effectively lessen the burden autoradiography study. J Pharmacol Exp Ther
of chronic pain. Acupuncture is also a procedure 1988;247(2):757764.
that is rarely associated with side effects when ster- Cui M, Tang X, Christian WV, Yoon Y, Tieu K.
ile needles are utilized (Lao, 1996). Another key Perturbations in mitochondrial dynamics
advantage is that acupuncture does not adversely induced by human mutant PINK1 can be rescued
by the mitochondrial division inhibitor mdivi-1. J
affect cognitive functions and presents no risk for
Biol Chem 2010;285(15):1174011752.
dependence (Yamashita etal., 2000). Unraveling
Cunha RA, Sebastiao AM. Extracellular metabo-
the mechanistic foundation of acupuncture has
lism of adenine nucleotides and adenosine in
identified adenosine and A1 receptors as key
the innervated skeletal muscle of the frog. Eur J
effectors of acupunctures analgesic properties.
Pharmacol 1991;197(1):8392.
Manipulations of adenosine metabolism which
Curros-Criado MM, Herrero JF. The antinocicep-
prolong A1 receptors activation (CFA and pros- tive effects of the systemic adenosine A1 receptor
tatic acid phosphatase) have already been shown agonist CPA in the absence and in the presence
to enhance the beneficial effects of acupunc- of spinal cord sensitization. Pharmacol Biochem
ture, whereas A1 receptor antagonists potently Behav 2005;82(4):721726.
inhibit acupunctures analgesic actions. These Derry CJ, Derry S, Moore RA. Caffeine as an anal-
observations indicate that acupunctures analge- gesic adjuvant for acute pain in adults. Cochrane
sic properties have a clear molecular basisone Database Syst Rev 2012;3:CD009281.
that is susceptible to other forms of manipula- Elmenhorst D, Meyer PT, Matusch A, Winz OH,
tion and may form the basis of novel treatments Bauer A. Caffeine occupancy of human cere-
for chronic pain. Future work may even design bral A1 adenosine receptors: In vivo quantifi-
local approaches to reduce the burden of chronic cation with 18F-CPFPX and PET. J Nucl Med
pain by manipulation of purine signaling in the 2012;53(11):17231729.
absence of acupuncture itself. Flogel U, Burghoff S, van Lent PL, Temme S, Galbarz L,
Ding Z, El-Tayeb A, Huels S, Bonner F, Borg N,
References Jacoby C, Muller CE, van den Berg WB, Schrader
Acupuncture. NIH Consensus Statement 1997 J. Selective activation of adenosine A2A recep-
Nov 35. 1997;15(5):134. tors on immune cells by a CD73-dependent
Abbracchio MP, Burnstock G, Verkhratsky A, prodrug suppresses joint inflammation in exper-
Zimmermann H. Purinergic signalling in the imental rheumatoid arthritis. Sci Transl Med
nervous system: An overview. Trends Neurosci 2012;4(146):146ra108.
2009;32(1):1929. Frary CD, Johnson RK, Wang MQ. Food sources
Ahn AC, Colbert AP, Anderson BJ, Martinsen OG, and intakes of caffeine in the diets of per-
Hammerschlag R, Cina S, Wayne PM, Langevin sons in the United States. J Am Diet Assoc
HM. Electrical properties of acupuncture 2005;105(1):110113.
points and meridians: A systematic review. Fredholm BB. Adenosine, an endogenous distress
Bioelectromagnetics 2008;29(4):245256. signal, modulates tissue damage and repair. Cell
Ashburn MA, Staats PS. Management of chronic Death Differ 2007;14(7):13151323.
pain. Lancet 1999;353(9167):18651869. Goldman N, Chandler-Militello D, Langevin HM,
Ballantyne JC, Mao J. Opioid therapy for chronic Nedergaard M, Takano T. Purine receptor medi-
pain. N Engl J Med 2003;349(20):19431953. ated actin cytoskeleton remodeling of human
Bossy J. Neural mechanisms in acupuncture analge- fibroblasts. Cell Calcium 2013;53(4):297301.
sia. Minerva Med 1979;70(24):17051715. Goldman N, Chen M, Fujita T, Xu Q, Peng W, Liu
Burnstock G. Physiology and pathophysiology of W, Jensen TK, Pei Y, Wang F, Han X, Chen JF,
purinergic neurotransmission. Physiol Rev Schnermann J, Takano T, Bekar L, Tieu K,
2007;87(2):659797. Nedergaard M. Adenosine A1 receptors mediate
350 Part III:Homeostatic Manipulators

local anti-nociceptive effects of acupuncture. Karlsten R, Gordh T, Post C. Local antinociceptive


Nature Neurosci 2010;13(7):883888. and hyperalgesic effects in the formalin test after
Gong QJ, Li YY, Xin WJ, Wei XH, Cui Y, Wang J, peripheral administration of adenosine ana-
Liu Y, Liu CC, Li YY, Liu XG. Differential effects logues in mice. Pharmacol Toxicol 1992;70(6 Pt 1):
of adenosine A1 receptor on pain-related behav- 434438.
ior in normal and nerve-injured rats. Brain Res Kelly RB. Acupuncture for pain. Am Fam Physician
2010;1361:2330. 2009;80(5):481484.
Guerra de Hoyos JA, Andres Martin Mdel C, Bassas y Koski MA. Acupuncture for zoological companion
Baena de Leon E, Vigara Lopez M, Molina Lopez T, animals. Vet Clin North Am Exot Anim Pract
Verdugo Morilla FA, Gonzalez Moreno MJ. 2011;14(1):141154.
Randomised trial of long term effect of acupunc- Langevin HM, Bouffard NA, Churchill DL, Badger GJ.
ture for shoulder pain. Pain 2004;112(3):289298. Connective tissue fibroblast response to acu-
Gunn CC, Ditchburn FG, King MH, Renwick GJ. puncture:Dose-dependent effect of bidirectional
Acupuncture loci: A proposal for their clas- needle rotation. J Altern Complement Med
sification according to their relationship to 2007;13(3):355360.
known neural structures. Am J Chin Med Langevin HM, Fujita T, Bouffard NA, Takano
1976;4(2):183195. T, Koptiuch C, Badger GJ, Nedergaard M.
Guo W, Zhang Z, Liu X, Burnstock G, Xiang Z, Fibroblast cytoskeletal remodeling induced
He C. Developmental expression of P2X5 by tissue stretch involves ATP signaling. J Cell
receptors in the mouse prenatal central and Physiol 2013;228(9):19221926.
peripheral nervous systems. Purinergic Signal Langevin HM, Yandow JA. Relationship of acupunc-
2013;9(2):239248. ture points and meridians to connective tissue
Han JS. Acupuncture and endorphins. Neurosci Lett planes. Anat Rec 2002;269(6):257265.
2004;361(13):258261. Lao L. Safety issues in acupuncture. J Altern
Haskell CF, Kennedy DO, Wesnes KA, Scholey AB. Complement Med 1996;2(1):2731.
Cognitive and mood improvements of caf- Lao L, Zhang RX, Zhang G, Wang X, Berman BM, Ren
feine in habitual consumers and habitual K. A parametric study of electroacupuncture on
non-consumers of caffeine. Psychopharmacology persistent hyperalgesia and Fos protein expres-
2005;179(4):813825. sion in rats. Brain Res 2004;1020(12):1829.
Hasko G, Pacher P. A2A receptors in inflammation Latini S, Pedata F. Adenosine in the central
and injury:Lessons learned from transgenic ani- nervous system: Release mechanisms and
mals. J Leukoc Biol 2008;83(3):447455. extracellular concentrations. J Neurochem
Horiuchi H, Ogata T, Morino T, Yamamoto H. 2001;79(3):463484.
Adenosine A1 receptor agonists reduce hyperal- Li J, King NC, Sinoway LI. ATP concentrations and
gesia after spinal cord injury in rats. Spinal Cord muscle tension increase linearly with muscle
2010;48(9):685690. contraction. J Appl Physiol 2003;95(2):577583.
Hurt JK, Zylka MJ. PAPupuncture has localized and Li J, King NC, Sinoway LI. Interstitial ATP and nor-
long-lasting antinociceptive effects in mouse epinephrine concentrations in active muscle.
models of acute and chronic pain. Mol Pain Circulation 2005;111(21):27482751.
2012;8:28. Li WM, Cui KM, Li N, Gu QB, Schwarz W, Ding
Hwang JH, Hwang GS, Cho SK, Han SM. Morphine GH, Wu GC. Analgesic effect of electroacupunc-
can enhance the antiallodynic effect of intra- ture on complete Freunds adjuvant-induced
thecal R-PIA in rats with nerve ligation injury. inflammatory pain in mice:Amodel of antipain
Anesth Analg 2005;100(2):461468. treatment by acupuncture in mice. Jpn J Physiol
Johansson B, Halldner L, Dunwiddie TV, Masino SA, 2005;55(6):339344.
Poelchen W, Gimenez-Llort L, Escorihuela RM, Lieberman HR, Tharion WJ, Shukitt-Hale B,
Fernandez-Teruel A, Wiesenfeld-Hallin Z, Speckman KL, Tulley R. Effects of caffeine,
Xu XJ, Hardemark A, Betsholtz C, Herlenius sleep loss, and stress on cognitive perfor-
E, Fredholm BB. Hyperalgesia, anxiety, and mance and mood during U.S. Navy SEAL
decreased hypoxic neuroprotection in mice lack- training. Sea-air-land. Psychopharmacology
ing the adenosine A1 receptor. Proc Natl Acad 2002;164(3):250261.
Sci USA 2001;98(16):94079412. Liu C, Xiaohu W, Hua X, Fang L, Dang R, Zhang D,
Kang J, Kang N, Lovatt D, Torres A, Zhao Z, Zhang X, Xie H, Xiao T. X-ray phase-contrast CT
Lin J, Nedergaard M. Connexin 43 hemi- imaging of the acupoints based on synchrotron
channels are permeable to ATP. J Neurosci radiation. J Electron Spectros Relat Phenomena
2008;28(18):47024711. 2014;196;8084.
The Role of Acupuncture in Nociception Homeostasis 351

Lohse MJ, Klotz KN, Schwabe U, Cristalli G, Vittori S, Puntambekar P, Van Buren J, Raisinghani M,
Grifantini M. 2-Chloro-N6-cyclopentyladenosine: Premkumar LS, Ramkumar V. Direct interac-
Ahighly selective agonist at A1 adenosine recep- tion of adenosine with the TRPV1 channel pro-
tors. Naunyn-Schmiedebergs Arch Pharmacol tein. J Neurosci 2004;24(14):36633671.
1988;337(6):687689. Rittiner JE, Korboukh I, Hull-Ryde EA, Jin J,
Marchand S, Li J, Charest J. Effects of caffeine on Janzen WP, Frye SV, Zylka MJ. AMP is an
analgesia from transcutaneous electrical nerve adenosine A1 receptor agonist. J Biol Chem
stimulation. N Engl J Med 1995;333(5):325326. 2012;287(8):53015309.
Melzack R, Stillwell DM, Fox EJ. Trigger points and Rowbotham MC, Twilling L, Davies PS, Reisner L,
acupuncture points for pain: Correlations and Taylor K, Mohr D. Oral opioid therapy for
implications. Pain 1977;3(1):323. chronic peripheral and central neuropathic pain.
Moore ME, Berk SN. Acupuncture for chronic shoul- N Engl J Med 2003;348(13):12231232.
der pain:An experimental study with attention Sauer H, Hescheler J, Wartenberg M. Mechanical
to the role of placebo and hypnotic susceptibility. strain-induced Ca(2+) waves are propa-
Ann Intern Med 1976;84(4):381384. gated via ATP release and purinergic recep-
Mor AO, Cidral-Filho FJ, Mazzardo-Martins L, tor activation. Am J Physiol Cell Physiol
Martins DF, Nascimento FP, Li SM, Santos 2000;279(2):C295C307.
ARS. Caffeine at moderate doses can inhibit Sawynok J. Adenosine receptor activation and noci-
acupuncture-induced analgesia in a mouse ception. Eur J Pharmacol 1998;347(1):111.
model of postoperative pain. J Caffeine Res Sawynok J. Adenosine and ATP receptors. Handb
2013;3(3):143148. Exp Pharmacol 2007(177):309328.
Mukherjee D, Nissen SE, Topol EJ. Risk of cardio- Sawynok J. Caffeine and pain. Pain 2011;
vascular events associated with selective COX-2 152(4):726729.
inhibitors. JAMA 2001;286(8):954959. Sawynok J, Liu XJ. Adenosine in the spinal cord and
Nascimento FP, Figueredo SM, Marcon R, Martins DF, periphery:Release and regulation of pain. Prog
Macedo SJ, Jr., Lima DA, Almeida RC, Ostroski Neurobiol 2003;69(5):313340.
RM, Rodrigues AL, Santos AR. Inosine reduces Sawynok J, Yaksh TL. Caffeine as an analgesic adju-
pain-related behavior in mice: Involvement of vant:Areview of pharmacology and mechanisms
adenosine A1 and A2A receptor subtypes and of action. Pharmacol Rev 1993;45(1):4385.
protein kinase C pathways. J Pharmacol Exp Song JG, Hahm KD, Kim YK, Leem JG, Lee C,
Ther 2010;334(2):590598. Jeong SM, Park PH, Shin JW. Adenosine
Nehlig A, Daval JL, Debry G. Caffeine and the cen- triphosphate-sensitive potassium channel
tral nervous system:Mechanisms of action, bio- blockers attenuate the antiallodynic effect
chemical, metabolic and psychostimulant effects. of R-PIA in neuropathic rats. Anesth Analg
Brain Res Brain Res Rev 1992;17(2):139170. 2011;112(6):14941499.
Neumann S, Doubell TP, Leslie T, Woolf CJ. Taiwo YO, Levine JD. Direct cutaneous hyper-
Inflammatory pain hypersensitivity mediated by algesia induced by adenosine. Neuroscience
phenotypic switch in myelinated primary sen- 1990;38(3):757762.
sory neurons. Nature 1996;384(6607):360364. Takano T, Chen X, Luo F, Fujita T, Ren Z, Goldman
Noble M, Treadwell JR, Tregear SJ, Coates VH, N, Zhao Y, Markman JD, Nedergaard M.
Wiffen PJ, Akafomo C, Schoelles KM. Long-term Traditional acupuncture triggers a local
opioid management for chronic noncancer pain. increase in adenosine in human subjects. J Pain
Cochrane Database Syst Rev 2010(1):CD006605. 2012;13(12):12151223.
Ohta A, Sitkovsky M. Role of G-protein-coupled ade- Taylor BK. Spinal inhibitory neurotransmission
nosine receptors in downregulation of inflam- in neuropathic pain. Curr Pain Headache Rep
mation and protection from tissue damage. 2009;13(3):208214.
Nature 2001;414(6866):916920. National Institutes of Health. Program announce-
Ong CK, Lirk P, Tan CH, Seymour RA. An evidence- ment: The management of chronic pain.
based update on nonsteroidal anti-inflammatory Washington, DC: National Institutes of
drugs. Clin Med Res 2007;5(1):1934. Health;2001.
Petrie JP, Hazleman BL. A controlled study of Thomas KJ, MacPherson H, Ratcliffe J, Thorpe L,
acupuncture in neck pain. Br J Rheumatol Brazier J, Campbell M, Fitter M, Roman M,
1986;25(3):271275. Walters S, Nicholl JP. Longer term clinical and
Portenoy RK. Current pharmacotherapy of chronic economic benefits of offering acupuncture care
pain. J Pain Symptom Manage 2000;19(1 to patients with chronic low back pain. Health
Suppl):S16S20. Technol Assess 2005;9(32):iiiiv, ixx,1109.
352 Part III:Homeostatic Manipulators

Tsuda M, Masuda T, Tozaki-Saitoh H, Inoue K. Cx32 and Cx43 gap junction proteins in nor-
P2X4 receptors and neuropathic pain. Front Cell mal and neoplastic human tissues. Int J Cancer
Neurosci 2013;7:191. 1992;51(4):522529.
Vadakkan KI, Jia YH, Zhuo M. A behavioral model World Health Organization. Acupuncture: Review
of neuropathic pain induced by ligation of and analysis of reports on controlled clinical tri-
the common peroneal nerve in mice. J Pain als. Geneva:World Health Organization;2003.
2005;6(11):747756. Wu WP, Hao JX, Halldner L, Lovdahl C, DeLander
Veith I. Acupuncture in traditional Chinese GE, Wiesenfeld-Hallin Z, Fredholm BB, Xu
medicine: An historical review. Calif Med XJ. Increased nociceptive response in mice
1973;118(2):7079. lacking the adenosine A1 receptor. Pain
Vickers AJ, Cronin AM, Maschino AC, Lewith G, 2005;113(3):395404.
Macpherson H, Foster NE, Sherman KJ, Yamashita H, Tsukayama H, Hori N, Kimura T,
Witt CM, Linde K. Acupuncture for chronic Tanno Y. Incidence of adverse reactions associ-
pain: Individual patient data meta-analysis. ated with acupuncture. J Altern Complement
Arch Intern Med 2012;172:14441453. Med 2000;6(4):345350.
Volonte MG, Yuln G, Quiroga P, Consolini Yang ES, Li PW, Nilius B, Li G. Ancient Chinese
AE. Development of an HPLC method for medicine and mechanistic evidence of acu-
determination of metabolic compounds in puncture physiology. Pflugers Arch 2011;
myocardial tissue. J Pharm Biomed Anal 462(5):645653.
2004;35(3):647653. Yang J, Yang Y, Chen JM, Liu WY, Wang CH,
Weidenhammer W, Linde K, Streng A, Hoppe A, Lin BC. Effect of oxytocin on acupuncture
Melchart D. Acupuncture for chronic low back analgesia in the rat. Neuropeptides 2007;
pain in routine care:Amulticenter observational 41(5):285292.
study. Clin J Pain 2007;23(2):128135. Zahn PK, Straub H, Wenk M, Pogatzki-Zahn
White T, Potter P, Moody C, Burnstock G. EM. Adenosine A1 but not A2a receptor ago-
Tetrodotoxin-resistant release of ATP from nist reduces hyperalgesia caused by a surgi-
guinea-pig taenia coli and vas deferens dur- cal incision in rats: a pertussis toxin-sensitive
ing electrical field stimulation in the presence G protein-dependent process. Anesthesiology
of luciferin-luciferase. Can J Physiol Pharmacol 2007;107(5):797806.
1981;59(10):10941100. Zhao ZQ. Neural mechanism underlying acu-
Wilgenbus KK, Kirkpatrick CJ, Knuechel R, puncture analgesia. Prog Neurobiol 2008;
Willecke K, Traub O. Expression of Cx26, 85(4):355375.
21
Meditation
GALLE DESBORDES

INTRODUCTION 2004). Allostatic load has been proposed as


Meditation has been practiced for centuries by a marker of cumulative biological risk and a
people across the world to improve well-being predictor of mortality and decline in physi-
and reduce suffering. Scientific research from cal functioning status (Beckie, 2012; McEwen
the past several decades suggests that medita- and Seeman, 1999; Seeman et al., 2001, 2010).
tion practice can yield objective, measurable Allostatic load adversely affects multiple physi-
health benefits. In particular, evidence is grow- ological systems in the organism (including the
ing that meditation practice may offer protection nervous, endocrine, and immune systems) and
against the deleterious effects of chronic stress, can manifest in the form of chronic inflamma-
which engenders a dysregulation of homeostatic tion, overactivation of the sympathetic nervous
and allostatic processes and can lead to seri- system, underactivation of the parasympathetic
ous health disorders. The brain is both affected nervous system, and premature cellular aging
by and centrally involved in the regulation of (e.g., shorter telomeres). Allostatic load has been
chronic stress (Maier, 2003). It has been pro- proposed as a major contributor to the increased
posed that meditation practice is a form of mind occurrence of a broad range of modern-day ill-
training that can enhance health by improving nesses, including brain disorders such as major
emotional regulation and increasing resiliency depression, posttraumatic stress disorder, and
to stress (Benson, 1975; Kabat-Zinn, 1990), pos- other chronic anxiety disorders (McEwen,
sibly via the generation of enduring, beneficial 2003). Recent studies support the intriguing
changes in the brain through better regulation possibility that these deleterious effects may
of inflammation and neuroplasticity mecha- be counteracted by specific brain training
nisms (Davidson and McEwen, 2012; Lutz programs (Bryck and Fisher, 2012), includ-
et al., 2007; Ornish et al., 2008, 2013; Slagter ing meditation-based interventions designed
etal.,2011). to promote well-being and prosocial behavior
Allostasis (stability through change) is a (Davidson and McEwen,2012).
model of physiological regulation that is a gener- While the field of meditation science has
alization of the well-known concept of homeosta- not yet directly addressed how meditation may
sis and accounts for cases in which physiological affect homeostatic control of the brain, a grow-
set points are not constant but vary as a func- ing number of studies suggest that meditation
tion of bodily needs and competing motivation. affects physiological mechanisms that can dis-
Allostatic regulation necessitates the extensive rupt or restore homeostasis in the brain. In this
involvement of the central nervous system as the chapter we review the current body of literature
main coordinator of regulatory responsesboth relating to the short- and long-term effects of
behavioral and physiological (Schulkin, 2004; meditation practice on the central nervous sys-
Sterling and Eyer,1988). tem and on peripheral systems that are directly
While the stress response is adaptive in the or indirectly involved in homeostaticor
short term, chronic stress exacts a cost on the allostaticregulation of the brain. Specifically,
organism known as allostatic load, the cumu- the topics covered include the effects of medita-
lative physiological burden enacted on the body tion training on (i)blood pressure, (ii) immune
through attempts to adapt to lifes demands function, (iii) telomeres, (iv) autonomic regula-
that can accelerate disease processes (McEwen, tion of the heart, (v)HPA axis regulation, and (vi)
1998, 2004; McEwen and Stellar, 1993; Schulkin, neuroplasticity.
354 Part III:Homeostatic Manipulators

W H AT A R E M E D I TAT I O N mindfulness-based stress reduction (MBSR),


PR ACTICES? which is the original format (Kabat-Zinn, 1990,
Meditation practices have existed since prehis- 2013); mindfulness-based cognitive therapy
toric times (Johnson, 1982) and are currently (MBCT), which is a hybrid between MBSR and
practiced globally by many people, both in reli- cognitive behavioral therapy originally designed
gious and secular (including clinical) contexts. to prevent depression relapse in patients with a
Any attempt to define meditation is fraught history of major depression but that has been
with debate, and this review is not the place to increasingly applied to other clinical and non-
enter into these academic considerations. As a clinical populations (Segal et al., 2013; Teasdale
starting point, most meditative practices dis- etal., 1995); and other mindfulness-based inter-
cussed in this chapter are more or less adequately ventions with varying duration and components
described by the following working definition, (Kabat-Zinn, 2003). Other practices derived
offered by Shapiro (1982): Meditation refers to from mindfulness include third-wave forms
a family of techniques which have in common a of psychotherapy such as acceptance and com-
conscious attempt to focus attention in a nonana- mitment therapy (Hayes, 2004; Hayes et al.,
lytical way and an attempt not to dwell on discur- 1999) and dialectical behavioral therapy (Lau
sive, ruminating thought. However, other types and McMain, 2005; Linehan, 1993; Robins etal.,
of meditation not covered by this definition exist, 2004). A growing scientific literature points to
such as the loving-kindness and compassion wide-ranging benefits from mindfulness-based
meditation practices described later. Different programs, from decreases in anxiety, depression,
traditions and schools of thought offer a wide and chronic pain to improvements in immune
variety of meditation practices, and interested function and more (Kabat-Zinn, 2013). However,
readers are referred to the vast literature dedi- the methodological quality of the scientific litera-
cated to these topics. Overall, the full spectrum ture on meditation for health has been criticized
of meditation practiceswhich vary in terms of (Ospina etal., 2007). Arecent review concludes
their purpose, specific instructions, and expected that mindfulness-based programs show moder-
resultsis therefore extremely broad. The generic ate evidence of improved anxiety, depression,
term meditation (often used without further and pain but low evidence of improved stress/
elaboration in the scientific literature) can refer distress, mental health related quality of life, and
to widely diverse practices, likely with varying positive mood (Goyal et al., 2014), even though
effects on physiology. Recent attempts to organ- participants in mindfulness-based interventions
ize these practices into a few categories for the often report an increase in well-being (reviewed
purpose of scientific research resulted in active in Chambers etal., 2009; Grossman, 2004; Rubia,
debate (Awasthi, 2012; Josipovic, 2010; Lutz etal., 2009). Clearly, more high-quality research is
2008; Travis and Shear, 2010a, 2010b). In sum- needed, but the preliminary evidence to date is
mary, experts agree that there is no such thing very encouraging, as we reviewlater.
as meditation as a single, monolithic practice. Other types of meditative or contempla-
This important distinction can help explain the tive practices are receiving increasing interest
variety of physiological effects reported in the from the scientific community. Practices col-
scientific literature todate. lectively referred to as loving-kindness or
Some of the first meditation practices investi- compassion meditation (which encompass
gated scientifically, in nonexpert as well as expert several distinct forms of practices) are aimed at
practitioners, were Transcendental Meditation, cultivating loving-kindnessa form of uncondi-
also known as TM (Rosenthal, 2011; Wallace tional love toward all beings (Salzberg, 1995)or
et al., 1971), and the Relaxation Response compassionthe feeling that arises in witnessing
(Benson, 1975; Benson et al., 1974b). Since the anothers suffering and that motivates a subse-
1980s, another increasingly popular type of med- quent desire to help (Goetz et al., 2010). Unlike
itation practice offered in the clinical context is mere empathic resonance, which can lead to
mindfulness, first introduced by Kabat-Zinn burnout, secondary traumatic stress, or compas-
(1982). Nowadays, numerous mindfulness-based sion fatigue (a deterioration of ones resiliency,
interventions exist that are centered on mind- coping, and empathic abilities), true compas-
fulness meditation instruction and practice, sion is considered beneficial to the individual
typically in the form of a six- to eight-week who experiences it (Klimecki etal., 2014; Singer
group intervention. These programs include and Bolz, 2013). In recent years, a number of
Meditation 355

programs have been proposed to train indi- several intercorrelated physiological systems such
viduals in cultivating loving-kindness and com- as the sympathetic and parasympathetic nervous
passion via meditative practices presented in systems and the hypothalamicpituitaryadrenal
a secular format adapted to a modern lifestyle (HPA) axis (McEwen and Seeman, 1999; Seeman
(Germer, 2009; Gilbert, 2005; Hofmann et al., etal., 2001,2010).
2011; Jazaieri et al., 2013; Makransky, 2007;
Ozawa-de Silva and Negi, 2013; Salzberg, 1995; Blood Pressure
Wallmark et al., 2012). Recent scientific studies Early studies suggested that Transcendental
indicate that loving-kindness and compassion Meditation and the related Relaxation Response
practices may have multiple beneficial effects, practice could reduce blood pressure and nor-
such as improvements in chronic pain and psy- malize hypertension (Benson et al., 1974b;
chological distress (Carson et al., 2005); reduc- Wallace etal., 1971). Later studies seem to con-
tions in depression, anxiety, rumination, and firm this finding, albeit amid some controversy
self-criticism (Gilbert and Procter, 2006; Kemeny (Canter and Ernst, 2004; Orme-Johnson et al.,
et al., 2012); reduced inflammation response 2005; Parati and Steptoe, 2004). A more recent
(Pace etal., 2009, 2010, 2013); and increased vagal meta-analysis of nine randomized controlled
tone as assessed by heart rate variability (HRV; trials concluded that TM could decrease blood
Kok etal., 2013). Afew studies have also begun pressure, with an effect size of 4.7mm Hg (95%
to investigate the neural correlates of these prac- confidence interval:7.4 to 1.9mm Hg) for sys-
tices, although much more work is needed in tolic blood pressure and 3.2mm Hg (95% confi-
this area (Desbordes etal., 2012b; Garrison etal., dence interval:5.4 to 1.3mm Hg) for diastolic
2014; Klimecki etal., 2013, 2014; Lee etal., 2012; blood pressure, respectivelyeffect sizes that
Leung et al., 2013; Mascaro et al., 2013; Weng are considered clinically meaningful (Anderson
etal.,2013). et al., 2008). There is also preliminary evidence
Other types of meditative practices have suggesting that the MBSR intervention can
been scientifically studied, for instance visu- reduce blood pressure as well. Asingle-arm study
alization practices (Kozhevnikov et al., 2009); of breast and prostate cancer patients undergo-
gTummo or inner heat yoga (Benson et al., ing MBSR found a significant pre- to postinter-
1982; Kozhevnikov etal., 2013); and body aware- vention 2.1 mm Hg decrease in systolic blood
ness practices such as yoga, qigong, tai-chi, the pressure but no effect on diastolic blood pressure
Alexander Technique, the Feldenkrais Method, (Carlson et al., 2007). A randomized controlled
and so on (Mehling etal., 2011; Schmalzl etal., trial of MBSR in prehypertensive patients, with
2014), which are beyond the scope of this review. an active control condition consisting of progres-
sive muscle relaxation training, found that MBSR
E F F E C T S O F M E D I TAT I O N could significantly reduce blood pressure in this
O N A L L O S TAT I C M A R K E R S population compared to the control intervention,
Early scientific experiments on meditators with a 4.8-mm Hg reduction in systolic blood
seemed to suggest that meditative states were pressure and a 1.9-mm Hg reduction in diastolic
associated with a hypometabolic state and blood pressure (Hughes etal.,2013).
had calming, relaxing, stress-reducing effects
(Benson etal., 1974a; Wallace etal., 1971; Young Immune Function
and Taylor, 1998), even though it was recognized Some evidence suggests that meditation training
early on that some types of meditation could may improve immune function in healthy indi-
also increase arousal (Amihai and Kozhevnikov, viduals. Based on previous studies indicating that
2014; Corby et al., 1978; Shapiro, 1982; Shapiro chronic stress can diminish antibody response to
and Walsh,1984). vaccines (Glaser etal., 1998; Kiecolt-Glaser etal.,
The benefits of meditation practice for stress 1996; Yang and Glaser, 2002), Davidson et al.
reduction purposes have been widely reported. (2003) conducted a randomized controlled trial
In this section we review some of the most nota- of MBSR in a workforce population in which
ble studies to date showing the effects of medi- subjects in both the intervention group and the
tation practices on some allostatic biomarkers. waitlist control group received the influenza vac-
Allostatic load can be estimated from markers of cine at the end of the eight-week period. Subjects
inflammation, metabolism, blood pressure, and who had received MBSR (N=25) showed small
telomeres, as well as by measuring the integrity of but statistically significant increases in antibody
356 Part III:Homeostatic Manipulators

titers to influenza vaccine compared with control psychological stress in the laboratory. The TSST
subjects (N=16). consists of a public speaking and mental arith-
On a broader level, it has been proposed that metic task performed in front of a panel of emo-
meditation practice might help regulate chronic tionally nonresponsive confederates presented as
inflammation (e.g., Oke and Tracey, 2009). behavioral experts. Even though the stressor
Chronic inflammation is especially relevant itself lasts only a few minutes, the TSST reliably
to homeostatic regulation of the brain, as the activates the autonomic nervous system, HPA
immune system and the central nervous system axis, and innate immune inflammatory response
form a bidirectional communication network over a time course of several hours (Dickerson
(Maier, 2003; McEwen, 2006). Chronic inflam- and Kemeny, 2004; Kirschbaum etal.,1993).
mation has been associated with a number of Several studies of meditation have used the
diseases that can affect the brain (Anthony and TSST to assess changes in inflammatory response
Pitossi, 2013), including inflammatory diseases to stress after several weeks of meditation train-
of the blood vessel wall (with increased risk of ing. In a randomized controlled trial of compas-
thrombotic stroke) and other cardiovascular sion meditation compared to an active control
diseases, Alzheimers disease (Ferreira et al., intervention (based on health discussion groups)
2014; Maccioni et al., 2009; Rubio-Perez and conducted in healthy college students, Pace etal.
Morillas-Ruiz, 2012), Parkinsons disease (Hirsch (2009) found a post hoc association between com-
et al., 2012; More et al., 2013), brain metastasis passion meditation practice time and decreased
(Hamilton and Sibson, 2013), and even type II plasma levels of interleukin-6 (a proinflammatory
diabetes (Donath and Shoelson, 2011)which cytokine), although there was no group differ-
can be associated with brain dysfunction (Biessels ence between the two interventions. In a rand-
etal., 2014). Chronic inflammation may also be omized controlled trial of MBSR compared to the
involved in the process of age-related cognitive Health Enhancement Program (an ad hoc active
decline (Simen et al., 2011) and normal aging control intervention), Rosenkranz et al. (2013)
(Pizza etal., 2011; Sparkman and Johnson, 2008). also found comparable cortisol responses to the
Chronic inflammation is believed to be caused by TSST after both interventions but significantly
a combination of genetic predisposition, lifestyle, smaller flare in the MBSR group in response to
and psychosocial factors; it is, therefore, suscep- topical application of capsaicin cream to forearm
tible to sociobehavioral changes such as those skin, indicating reduced inflammation response.
promoted by meditation-based interventions and In another randomized controlled trial of MBSR
perhaps by other meditation-specific neuroplas- with an active control group, levels of adrenocor-
ticity mechanisms (Coe and Laudenslager, 2007; ticotrophin hormone (the hormone that stimu-
Davidson and McEwen,2012). lates cortisol release) during the TSST were lower
Some recent studies indicate that meditation in the MBSR group than in the control group
interventions may indeed reduce inflammation. (Hoge etal.,2012).
In a randomized controlled trial in a popula- Several recent studies also suggest that medi-
tion of foster-care adolescents (N=71) exposed tative states may have immediate effects on
to early life adversity (a known risk factor for inflammatory gene expression. In a recent study
increased inflammation), a six-week compassion conducted on experienced meditators taking
meditation intervention had some effects on sali- part in a meditation retreat, Kaliman etal. (2014)
vary concentration in C-reactive protein (CRP), found a downregulation of the proinflammatory
an inflammatory marker. While there was no genes RIPK2 and COX2 and of several histone
significant difference between the intervention deacetylase (HDAC) genes after engaging in
group and the control group in CRP levels over- mindfulness meditation for eight hours. In addi-
all, the number of practice sessions attended by tion, the extent to which some of those genes were
the subjects in the meditation group was corre- downregulated was associated with faster corti-
lated with a pre- to postintervention reduction sol recovery to a TSST. In a study of both experi-
in CRP, suggesting a possible dose-response rela- enced and novice practitioners of the Relaxation
tionship (Pace etal.,2013). Response, Bhasin etal. (2013) found that imme-
Inflammation can also be assessed in diately after engaging in the Relaxation Response
response to an acute stressor, such as the Trier practice, the expression of genes associated with
Social Stress Test (TSST), a commonly used energy metabolism, mitochondrial function,
standardized protocol for reproducibly inducing insulin secretion, and telomere maintenance was
Meditation 357

enhanced, whereas the expression of genes linked women, both the mindfulness group and the
to inflammatory response and stress-related waitlist control group showed an increase in tel-
pathways was reduced, with stronger effects in omerase activity over four months, with nega-
experienced practitioners than in novices. While tive correlations between changes in telomerase
these recent findings are very promising, more activity and changes in chronic stress, anxiety,
research is needed to determine whether medi- dietary restraint, dietary fat intake, cortisol,
tation affects inflammatory gene expression over and glucose (Daubenmier etal., 2012). This pre-
the longterm. liminary finding is in line with a previous study
of intensive lifestyle changes (including daily
Telomeres meditation practice) in prostate cancer patients
Telomeres are protective caps at the ends of who showed significantly increased telomerase
chromosomes. Their maintenance requires the activity after three months, where the increases
action of the ribonucleoprotein enzyme known in telomerase activity were significantly associ-
as telomerase. The degradation, or shortening, of ated with decreases in low-density lipoprotein
telomeres threatens chromosome integrity and is (LDL) cholesterol and decreases in psychological
implicated in the process of aging as well as cancer distress (Ornish etal., 2008)and with increased
(Aubert and Lansdorp, 2008; Corey, 2009; Eitan relative telomere length after five years of follow
etal., 2014; Falandry etal., 2014; Kong etal., 2013; up (Ornish etal.,2013).
Oeseburg etal., 2010; Zhu etal., 2011). Measures Finally, a recent pilot study on a small num-
of telomere length and telomerase activity have ber (N = 15) of experienced practitioners of
been used to assess cellular aging (Aubert and loving-kindness meditation and matched control
Lansdorp, 2008; Mather et al., 2011; Njajou subjects suggested that, in women, the amount
et al., 2009). Importantly, stress can accelerate of loving-kindness meditation practice over the
telomere shortening (reviewed in Starkweather lifetime was associated with longer telomeres
etal., 2014). Epel etal. (2004) found that women (Hoge etal.,2013).
with the highest levels of perceived stress had Overall, these preliminary findings support
telomeres shorter on average by the equivalent the possibility that meditation training may have
of at least one decade of additional aging com- positive impacts on telomere length and telomer-
pared to low-stress women. In addition, telom- ase activity.
erase activity in peripheral-blood mononuclear
cells, although not telomere length, was inversely Autonomic Regulation oftheHeart
associated with six major cardiovascular disease One of the main markers of allostatic load is HRV,
risk factors in healthy women, suggesting that the healthy beat-to-beat fluctuations in heart
telomerase activity may be a more direct and rate that reflect autonomic (sympathetic and
potentially earlier predictor than telomere length parasympathetic) influences on cardiac activity
of long-term cellular viability, genomic stability, (Malik etal., 1996). Diminished HRV is an inde-
and disease processes (Epel etal.,2006). pendent risk factor for mortality in patients with
In the field of meditation science, telomeres heart disease (Berntson et al., 1997; Freeman,
and telomerase activity are gaining interest as 2006; Friedman and Thayer, 1998; Malik et al.,
possible objective markers of health improve- 1996; Malliani et al., 1991; Mujica-Parodi et al.,
ments associated with meditation practice. For 2009; Stein etal., 1994)and has been associated
example, in a study of long-term meditation prac- with a variety of pathological states and disposi-
titioners engaged in a three-month-long, intense tions, including anxiety and depression (Gorman
meditation retreat (known as the Shamatha pro- and Sloan, 2000). For instance, low HRV is asso-
ject), telomerase activity was significantly greater ciated with both acute (state) anxiety (Fuller,
in retreat participants at the end of the retreat 1992; Jnsson, 2007; Watkins et al., 1998) and
than in matched control subjects, and mediation chronic (trait) anxiety and clinical anxiety dis-
analyses based on longitudinal psychological orders (Cohen and Benjamin, 2006; Cohen etal.,
assessments suggested that increases in perceived 2000; Friedman, 2007; Klein et al., 1995; Licht
control and decreases in negative affectivity con- etal., 2009; McCraty etal., 2001). HRV decreases
tributed to increased telomerase activity (Jacobs in relation to increased depression severity, even
etal.,2011). in the absence of cardiovascular disease (Brown
In a recent pilot study of mindfulness train- et al., 2009; Kemp et al., 2010). Consequently,
ing for healthy eating in overweight and obese high HRV has been proposed as an indicator of
358 Part III:Homeostatic Manipulators

good physical and psychological health (Porges, participants who underwent either type of medi-
2011; Thayer etal., 2009,2012). tation training (mindful-attention or compas-
Meditative states are usually associated with sion) showed an increase in resting HRV pre- to
higher vagal parasympathetic activity, as indi- postintervention, with a significant correlation
cated by higher HRV either during or imme- between increased HRV and decreased depres-
diately after engaging in a meditation practice sion score (Desbordes etal., 2012a).
session (Cysarz and Bssing, 2005; Ditto et al., In summary, while specific meditative states
2006; Kubota et al., 2001; Nesvold et al., 2012; are associated with a variety of changes in auto-
Peng etal., 1999, 2004; Phongsuphap etal., 2008; nomic activation, recent evidence suggests that
Takahashi etal., 2005; Tang etal., 2009; Wolever the regular practice of meditation may have
etal., 2012; Wu and Lo, 2008). However, it should lasting, beneficial influences on autonomic
be noted that certain advanced forms of medi- functioning.
tation practices seem to increase sympathetic It should be noted that the autonomic nerv-
activation in expert practitioners (Amihai and ous system and the HPA axis can be indepen-
Kozhevnikov, 2014; Corby etal., 1978; Kox etal., dently affected by a psychosocial stressor such
2012). Areview of the literature on the complex as the TSST (Schommer etal., 2003). Therefore,
autonomic changes associated with meditative it does not necessarily follow from the effects of
states is beyond the scope of this chapter. The meditation on autonomic function that medita-
question of interest here is whether the prac- tion will have comparable effects on the HPA
tice of meditation has lasting, beneficial influ- axis (e.g., as assessed by cortisol levels). Next we
ences on autonomic functioning that may affect review how meditation may affect regulation of
homeostatic regulation of the brain. In other the HPAaxis.
words, is autonomic function altered in medita-
tion practitioners, even outside periods of for- HPA Axis Regulation
mal meditation practice, such as during normal One of the main systems involved in allostatic
resting conditions or during tasks that challenge regulation is the HPA axis, a major neuroen-
the autonomic nervous system? Recent studies docrine subsystem that controls physiological
have linked even brief training in meditation responses to stress, such as increased levels of cor-
(from five days to several weeks) to a longitu- tisol (Dickerson and Kemeny, 2004; Hellhammer
dinal increase in resting HRV. In a single-arm et al., 2009; Kirschbaum et al., 1993; Marques
pilot study of patients presenting with depres- et al., 2010; Steptoe et al., 2007). Cortisol levels
sion and anxiety six months after surgery for have been proposed as an objective measure of
spontaneous subarachnoid hemorrhage, rest- HPA axis activation in chronic stress (Herbert
ing HRV was found to significantly increase and Cohen, 1993) and as an outcome measure
pre- to post-MBSR training, while depression for meditation interventions (Matousek et al.,
scores significantly decreased (Joo et al., 2010). 2010). Salivary cortisol is considered the method
Longitudinal increases in resting HRV have also of choice for measuring free cortisol levels in the
been found with meditation-based interventions context of stress research (Hellhammer et al.,
other than MBSR. In a waitlist-controlled field 2009). However, obtaining meaningful measures
experiment of university workers taking part in of cortisol can be challenging. Cortisol levels
a six-week program for learning loving-kindness exhibit wide variations throughout a 24-hour
meditation (with a one-hour class per week that cycle, following a circadian rhythm controlled
included guided meditation practice and group by the hypothalamic suprachiasmatic nucleus
discussion), the experimental condition (medi- (part of the HPA axis). While the daily cortisol
tation versus waitlist control) significantly pre- curve normally follows a stereotypical profile,
dicted an increase in resting HRV (Kok et al., with a sharp morning rise and a slow descending
2013). In our own recent study, healthy adults slope in the afternoon and evening, reaching its
were randomized to either mindful-attention minimum level during the night (Krieger etal.,
meditation training, compassion meditation 1971), many factors can perturb this rhythm,
training, or an active control intervention based and erratic cortisol patterns have been associated
on health education classes (see Desbordes etal., with metabolic abnormalities, fatigue, depres-
2012b). One of our assessments measured resting sion, psychosocial factors, and poor quality of
HRV before and after each of the three eight-week life in general (Chida and Steptoe, 2009; Debono
interventions. Preliminary analyses indicate that et al., 2009). In addition, flattened or abnormal
Meditation 359

diurnal cortisol rhythms are a predictor of early training (Rosenkranz etal., 2013). More longitu-
mortality in some types of cancer (Sephton etal., dinal studies with careful methodology for corti-
2000,2013). sol assessments are needed to elucidate the effects
To obtain meaningful cortisol measurements of meditation training on HPA axis activation in
as part of a research study, it is therefore neces- various populations.
sary to measure cortisol at very specific times The hypothesis that meditation practice may
relative to the subjects waking time, if possible contribute to lower abnormally elevated corti-
at multiple time points during a 24-hour cycle, sol levels, at least in some clinical populations,
or at short intervals immediately after waking should be of interest in the context of homeo-
to assess the cortisol awakening response (Clow static control of the brain since high concentra-
etal., 2004; Fries etal., 2009; Hellhammer etal., tions of cortisol may have deleterious effects on
2009; Pruessner et al., 1997). Alternatively, one the brain, especially in the hippocampus. It is
can measure cortisol response to acute stress- well known that the hippocampus becomes atro-
ors such as the TSST (Kirschbaum et al., 1993). phied and presents functional abnormalities in
Finally, it should be noted that cortisol levels vary humans and animals exposed to severe stress or
with age, gender, and certain pharmacological depression (Bora etal., 2012; Bremner etal., 2000;
treatments, including oral contraceptives. Colla etal., 2007; Davidson etal., 2002; Malykhin
Previous reports of the effects of medita- etal., 2010; Shah etal., 1998; Sheline etal., 1996,
tion training on cortisol levels showed mixed 1999). It has been suggested that this hippocam-
results (for a review see Matousek et al., 2010). pal atrophy results from prolonged exposure to
Some studies in cancer patients found that high concentrations of cortisol (Lupien et al.,
participation in MBSR was associated with a 1998). While normal levels of cortisol activate
global decrease in cortisol levels (Carlson etal., mineralocorticoid receptors and facilitate hip-
2007; Witek-Janusek etal., 2008), while another pocampal long-term potentiation (and mem-
study reported an increase in cortisol awaken- ory functions), high concentrations of cortisol
ing response (Matousek et al., 2011)arguably additionally activate glucocorticoid receptors
an improvement in this patient population since that have debilitating effects on hippocampal
stressed individuals tend to exhibit a blunted function, including a dampening of long-term
(i.e., abnormally flat) cortisol daily profile (Chida potentiation (Kerr et al., 1989). Therefore, cor-
and Steptoe, 2009). Other studies found no sig- tisol exerts a concentration-dependent biphasic
nificant changes in cortisol after MBSR, although (inverted U-shape) influence on the expression
their methodological design has been criticized of hippocampal plasticity (Diamond etal., 1992).
on the basis of inadequate cortisol sampling (e.g., While these mechanisms have been investigated
insufficient number of time points), small sample mostly in animal models, studies in humans
sizes, or lack of control over confounding varia- also indicate that elevated basal cortisol levels
bles such as diet, physical exercise, or sleepwake predict reduced hippocampal volume and defi-
cycle (Matousek et al., 2010). However, even cits in hippocampus-dependent memory tasks
carefully designed randomized controlled trials in the aging population (Lupien et al., 1998;
have also yielded unclear cortisol outcomes. For Sapolsky, 1992). These complex interactions
example, a longitudinal study of MBCT in remit- between cortisol levels and hippocampal struc-
ted depression patients with six-month follow-up ture and function may underlie the connection
showed no significant changes in any cortisol between stressful experiences and dampening of
measure (cortisol awakening response, diurnal hippocampal neurogenesis (Gould and Tanapat,
slope, or area under the curve; Gex-Fabry etal., 1999; Gould etal., 1998; Rao etal., 2010). The link
2012), although it may be due to the fact that cor- between cortisol levels and hippocampus size in
tisol patterns are erratic and difficult to detect chronic stress may take the form of a positive
in depressed patients (Peeters et al., 2004). In a feedback loop between pathologically high lev-
nonclinical community sample, a randomized els of cortisol and neuronal damage in the hip-
controlled trial of MBSR compared to an active pocampus, which may in turn further reduce
control intervention found no difference between HPA downregulation and promote hypercorti-
the groups in posttraining cortisol response to solemia (reviewed in Davidson etal.,2002).
a TSST, although the amount of mindfulness Most interestingly, hippocampal atrophy
practice (i.e., dose) predicted a steeper (arguably seems to be reversible to some extent (Gould etal.,
more salubrious) diurnal cortisol slope following 2000; Jacobs etal., 2000; Malykhin etal., 2010).
360 Part III:Homeostatic Manipulators

For example, chronic treatment with antidepres- grey matter volume in relation to attentional
sants increases neurogenesis in the hippocampus performance in Zen meditators compared with
(Santarelli et al., 2003; Vermetten et al., 2003). matched controls. They found that, contrary to
Since hippocampal atrophy can be reverted when the control subjects, meditators did not display
stress is reduced, the question naturally arises the expected effects of age on grey matter volume
whether similar effects can be achieved by reduc- and on performance in a computerized sustained
ing perceived stress, via the practice of medita- attention task. Vestergaard-Poulsen etal. (2009)
tion for example. Recent studies suggest that found higher grey matter density in the brain
meditation practice may indeed promote growth stem of experienced practitioners of Tibetan
(or prevent shrinkage) in the hippocampus. Dzogchen meditation compared to matched
Long-term meditation practitioners have larger controls, particularly in the medulla oblongata
hippocampi and greater grey matter density in region of the dorsal brain stem, which contains
the hippocampal complex than matched control autonomic nuclei involved in cardiac and res-
subjects (Hlzel etal., 2008; Luders etal., 2009, piratory functions. Grant etal. (2010) found that
2013a, 2013b). Remarkably, hippocampus growth compared to matched controls, Zen meditators
was also observed longitudinally after only eight had both lower pain sensitivity and greater cor-
weeks of meditation practice in meditation-nave tical thickness in pain-related areas (the dorsal
participants, in healthy subjects undergoing anterior cingulate and secondary somatosensory
MBSR (Hlzel et al., 2011) and in healthy sub- cortex), with a positive correlation between corti-
jects undergoing mindful-attention or compas- cal thickness in the anterior cingulate and years
sion meditation training (Desbordes etal., 2014). of meditation practice. A series of studies by
In addition, neuroplastic effects of meditation Luders and colleagues conducted on long-term
have been reported not only in the hippocam- meditators from various traditions demonstrated
pal complex but also in other brain regions, as widespread differences between meditators and
reviewednext. matched controls in several brain structural
measures. These measures included larger grey
Neuroplasticity matter volume in the right orbito-frontal cortex
In a seminal study of the effects on brain (Luders et al., 2009), larger hippocampus and
structure of lifetime meditation experience, greater grey matter in the hippocampal complex
Lazar etal. (2005) found greater cortical thick- (Luders et al., 2013a, 2013b); greater structural
ness in the right anterior insula and dorsolat- connectivity within major white matter path-
eral prefrontal cortex regions in Vipassana ways (Luders etal., 2011); local (voxel-wise) dif-
(or Insight) meditation practitioners than in ferences in grey matter asymmetry between left
matched control subjects, with a significant cor- and right hemispheres (Kurth et al., 2015); and
relation between cortical thickness and years greater cortical gyrification (a measure of corti-
of meditation experience. This study was the cal folding curvature) in the fusiform gyrus, pre-
first to suggest that meditation practices might central gyrus, cuneus, and especially the right
offset age-related cortical thinning and grey anterior dorsal insula (where gyrification was
matterloss. positively correlated with years of meditation
Other studies (reviewed in Fox et al., 2014; practice; Luders et al., 2012), again pointing to
Tang et al., 2015) have since extended these an important role of the right anterior insula. In
findings. In another cross-sectional study of a study of Theravadan Buddhist practitioners of
Vipassana meditators and matched controls, loving-kindness meditation, Leung et al. (2013)
Hlzel et al. (2008) used voxel-based morpho- found significantly greater grey matter volume
metry (Ashburner and Friston, 2000) to assess in meditators than in matched controls in the
grey matter concentration. This study confirmed right angular gyrus and right posterior parahip-
the earlier finding in Vipassana meditators of pocampal gyrus. While neuroplastic effects of
increased grey matter in the right anterior insula, meditation in the hippocampus had been found
which is involved in interoceptive awareness, in several other studies (reviewed previously),
presumably reflecting the cultivation of bodily this study was the first to highlight differences
awareness during this type of training. This study in the right angular gyrus, a region known to be
also reported greater grey matter concentration activated by cognitive empathy and perspective
in the left inferior temporal gyrus and right hip- taking and thereby potentially involved in com-
pocampus in meditators compared to matched passion and care for others (Adolphs, 2009; Van
controls. Pagnoni and Cekic (2007) investigated Overwalle, 2009; Saxe and Kanwisher,2003).
Meditation 361

While these studies suggest that the brains controlled trials with active control interven-
of meditators show some structural differ- tions, are warranted to rigorously test the effects
ences from the brains of nonmeditators, the of meditation-based interventions on different
cross-sectional nature of these studies does not physiological (and psychological) mechanisms
allow testing of whether meditation practice that may improve healthin particular regarding
caused these differences or whether individuals homeostatic and allostatic regulation processes
with particular brain features were more drawn in the brain and other physiological systems. The
to becoming long-term meditation practitioners. field is rapidly progressing and maturing into a
However, recent longitudinal studies of subjects full-fledged multidisciplinary research domain,
without prior meditation experience indicate with a growing number of investigators world-
that changes in brain structure can be detected wide collectively building a rigorous body of
after only a few weeks of meditation practice. evidence supporting the view that meditation
After participating in the eight-week MBSR practices may offer wide-ranging health benefits.
program, healthy subjects showed significantly The effects of meditation are especially remark-
higher longitudinal increases in grey matter able in the context of allostatic loadthe cumu-
concentration in the amygdala, hippocampus, lative, deleterious effects of chronic stress on our
posterior cingulate cortex, temporo-parietal bodies that not only diminish our quality of life
junction, and cerebellum compared with waitlist but can also promote chronic inflammation and
control subjects (Hlzel etal., 2010, 2011). After accelerate aging and disease processes. We expect
one month of integrative body-mind training that future research will further establish the
(a meditation training program based on tra- legitimacy of meditation practices as low-cost,
ditional Chinese medicine in the form of daily accessible therapeutic interventions for a variety
30-minute sessions totaling only 11 hours of of illnesses and preclinical conditions.
practice), a college-student population exhibited
increases in fractional anisotropy (a measure of References
white matter integrity) in the corona radiata, a Adolphs, R. (2009). The social brain: Neural basis
major white-matter tract connecting multiple of social knowledge. Annu. Rev. Psychol. 60,
brain structures to the anterior cingulate cortex, 693716.
a region previously implicated in self-regulation Amihai, I., and Kozhevnikov, M. (2014). Arousal
(Tang etal., 2010, 2012). In our longitudinal med- vs. relaxation:Acomparison of the neurophysi-
itation study mentioned previously (Desbordes ological and cognitive correlates of Vajrayana
etal., 2012b), preliminary analyses of structural and Theravada meditative practices. PLoS One
brain data suggest changes in several subcorti- 9, e102990.
cal regions (including increased hippocampal Anderson, J.W., Liu, C., and Kryscio, R.J. (2008).
volume) after eight weeks of training in either Blood pressure response to transcendental medi-
mindful-attention meditation or compassion tation: A meta-analysis. Am. J. Hypertens. 21,
meditation (Desbordes etal., 2014). In the com- 310316.
Anthony, D.C., and Pitossi, F.J. (2013). Special issue
passion meditation participants, we also found
commentary:The changing face of inflammation
a significant increase in cortical thickness in the
in the brain. Mol. Cell. Neurosci. 53,15.
dorsomedial prefrontal cortex (Singleton et al.,
Ashburner, J., and Friston, K.J. (2000). Voxel-based
2014), a region activated during empathy tasks
morphometrythe methods. Neuroimage 11,
that, in another study, showed increased activa-
805821.
tion correlated with greater empathic accuracy
Aubert, G., and Lansdorp, P. (2008). Telomeres and
after eight weeks of training in compassion medi- aging. Physiol. Rev. 88, 557579.
tation (Mascaro etal.,2013). Awasthi, B. (2012). Issues and perspectives in medi-
Taken together, these findings support the tation research:In search for a definition. Front.
view that meditation practices, as a form of Psychol. 3,613.
brain training, may promote neuroplasticity Beckie, T.M. (2012). A systematic review of allostatic
and impact multiple brain functions, including load, health, and health disparities. Biol. Res.
homeostatic and allostatic processes. Nurs. 14(4), 31146.
Benson, H. (1975). The Relaxation Response
CONCLUSION (NewYork:William Morrow).
The scientific investigation of meditative prac- Benson, H., Beary, J.F., and Carol, M.P. (1974a).
tices is still at an early stage. Larger longitudi- The relaxation response. Psychiatry J. Study
nal studies, ideally in the form of randomized Interpers. Process. 37,3746.
362 Part III:Homeostatic Manipulators

Benson, H., Lehmann, J.W., Malhotra, M.S., reduction (MBSR) in breast and prostate cancer
Goldman, R.F., Hopkins, J., and Epstein, M.D. outpatients. Brain. Behav. Immun. 21, 10381049.
(1982). Body temperature changes during the Carson, J.W., Keefe, F.J., Lynch, T.R., Carson, K.M.,
practice of g Tum-mo yoga. Nature 295, 234236. Goli, V., Fras, A.M., and Thorp, S.R. (2005).
Benson, H., Rosner, B.A., Marzetta, B.R., and Loving-kindness meditation for chronic low
Klemchuk, H.M. (1974b). Decreased blood- back pain: Results from a pilot trial. J. Holist.
pressure in pharmacologically treated hyperten- Nurs. 23, 287304.
sive patients who regularly elicited the relaxation Chambers, R., Gullone, E., and Allen, N.B. (2009).
response. Lancet 1, 289291. Mindful emotion regulation: An integrative
Berntson, G.G., Bigger, J.T., Eckberg, D.L., Grossman, review. Clin. Psychol. Rev. 29, 560572.
P., Kaufmann, P.G., Malik, M., Nagaraja, H.N., Chida, Y., and Steptoe, A. (2009). Cortisol awaken-
Porges, S.W., Saul, J.P., Stone, P.H., etal. (1997). ing response and psychosocial factors:Asystem-
Heart rate variability: Origins, methods, and atic review and meta-analysis. Biol. Psychol. 80,
interpretive caveats. Psychophysiology 34, 265278.
623648. Clow, A., Thorn, L., Evans, P., and Hucklebridge,
Bhasin, M.K., Dusek, J.A., Chang, B.-H., Joseph, F. (2004). The awakening cortisol response:
M.G., Denninger, J.W., Fricchione, G.L., Benson, Methodological issues and significance. Stress
H., and Libermann, T.A. (2013). Relaxation 7,2937.
response induces temporal transcriptome Coe, C.L., and Laudenslager, M.L. (2007).
changes in energy metabolism, insulin secre- Psychosocial influences on immunity, including
tion and inflammatory pathways. PLoS One 8, effects on immune maturation and senescence.
e62817. Brain. Behav. Immun. 21, 10001008.
Biessels, G.J., Strachan, M.W.J., Visseren, F.L.J., Cohen, H., and Benjamin, J. (2006). Power spectrum
Kappelle, L.J., and Whitmer, R.A. (2014). analysis and cardiovascular morbidity in anxiety
Dementia and cognitive decline in type 2 dia- disorders. Auton. Neurosci. Basic Clin. 128,18.
betes and prediabetic stages: Towards targeted Cohen, H., Benjamin, J., Geva, A.B., Matar, M.A.,
interventions. Lancet. Diabetes Endocrinol. 2, Kaplan, Z., and Kotler, M. (2000). Autonomic
246255. dysregulation in panic disorder and in
Bora, E., Fornito, A., Pantelis, C., and Ycel, M. post-traumatic stress disorder: Application of
(2012). Gray matter abnormalities in major power spectrum analysis of heart rate variability
depressive disorder: A meta-analysis of voxel at rest and in response to recollection of trauma
based morphometry studies. J. Affect. Disord. or panic attacks. Psychiatry Res. 96,113.
138,918. Colla, M., Kronenberg, G., Deuschle, M., Meichel, K.,
Bremner, J.D., Narayan, M., Anderson, E.R., Staib, Hagen, T., Bohrer, M., and Heuser, I. (2007).
L.H., Miller, H.L., and Charney, D.S. (2000). Hippocampal volume reduction and HPA-system
Hippocampal volume reduction in major depres- activity in major depression. J. Psychiatr. Res. 41,
sion. Am. J.Psychiatry 157, 115118. 553560.
Brown, A.D.H., Barton, D.A., and Lambert, G.W. Corby, J.C., Roth, W.T., Zarcone, V.P., and Kopell,
(2009). Cardiovascular abnormalities in patients B.S. (1978). Psychophysiological correlates of the
with major depressive disorder: Autonomic practice of Tantric Yoga meditation. Arch. Gen.
mechanisms and implications for treatment. Psychiatry 35, 571577.
CNS Drugs 23, 583602. Corey, D.R. (2009). Telomeres and telomerase:From
Bryck, R.L., and Fisher, P.A. (2012). Training the discovery to clinical trials. Chem. Biol. 16,
brain:Practical applications of neural plasticity 12191223.
from the intersection of cognitive neuroscience, Cysarz, D., and Bssing, A. (2005). Cardiorespiratory
developmental psychology, and prevention sci- synchronization during Zen meditation. Eur.
ence. Am. Psychol. 67, 87100. J.Appl. Physiol. 95,8895.
Canter, P.H., and Ernst, E. (2004). Insufficient evi- Daubenmier, J., Lin, J., Blackburn, E., Hecht,
dence to conclude whether or not transcendental F.M., Kristeller, J., Maninger, N., Kuwata, M.,
meditation decreases blood pressure:Results of a Bacchetti, P., Havel, P.J., and Epel, E. (2012).
systematic review of randomized clinical trials. J. Changes in stress, eating, and metabolic factors
Hypertens. 22, 20492054. are related to changes in telomerase activity in
Carlson, L.E., Speca, M., Faris, P., and Patel, K.D. a randomized mindfulness intervention pilot
(2007). One year pre-post intervention follow-up study. Psychoneuroendocrinology 37, 917928.
of psychological, immune, endocrine and blood Davidson, R.J., and McEwen, B.S. (2012). Social
pressure outcomes of mindfulness-based stress influences on neuroplasticity: Stress and
Meditation 363

interventions to promote well-being. Nat. Epel, E.S., Blackburn, E.H., Lin, J., Dhabhar, F.S.,
Neurosci. 15, 689695. Adler, N.E., Morrow, J.D., and Cawthon, R.M.
Davidson, R.J., Kabat-Zinn, J., Schumacher, J., (2004). Accelerated telomere shortening in
Rosenkranz, M., Muller, D., Santorelli, S.F., response to life stress. Proc. Natl. Acad. Sci.
Urbanowski, F., Harrington, A., Bonus, K., and U.S.A. 101, 1731217315.
Sheridan, J.F. (2003). Alterations in brain and Epel, E.S., Lin, J., Wilhelm, F.H., Wolkowitz, O.M.,
immune function produced by mindfulness Cawthon, R., Adler, N.E., Dolbier, C., Mendes,
meditation. Psychosom. Med. 65, 564570. W.B., and Blackburn, E.H. (2006). Cell aging in
Davidson, R.J., Pizzagalli, D.A., Nitschke, J.B., and relation to stress arousal and cardiovascular dis-
Putnam, K. (2002). Depression: Perspectives ease risk factors. Psychoneuroendocrinology 31,
from affective neuroscience. Annu. Rev. Psychol. 277287.
53, 545574. Falandry, C., Bonnefoy, M., Freyer, G., and Gilson,
Debono, M., Ghobadi, C., Rostami-Hodjegan, A., E. (2014). Biology of cancer and aging: A com-
Huatan, H., Campbell, M.J., Newell-Price, J., plex association with cellular senescence. J. Clin.
Darzy, K., Merke, D.P., Arlt, W., and Ross, R.J. Oncol. 32, 26042610.
(2009). Modified-release hydrocortisone to Ferreira, S.T., Clarke, J.R., Bomfim, T.R., and De
provide circadian cortisol profiles. J. Clin. Felice, F.G. (2014). Inflammation, defective
Endocrinol. Metab. 94, 15481554. insulin signaling, and neuronal dysfunction in
Desbordes, G., Barbieri, R., Citi, L., Lazar, S., Negi, L., Alzheimers disease. Alzheimers. Dement. 10,
Raison, C., and Schwartz, E. (2012a). Assessment S76S83.
of autonomic tone at rest and during meditation Fox, K. C. R., Nijeboer, S., Dixon, M. L., Floman,
in a longitudinal study of an eight-week medita- J. L., Ellamil, M., Rumak, S. P., Christoff,
tion intervention. BMC Complement. Altern. K. (2014). Is meditation associated with altered
Med. 12,P46. brain structure? Asystematic review and meta-
Desbordes, G., Negi, L.T., Pace, T.W.W., Wallace, B.A., analysis of morphometric neuroimaging in med-
Raison, C.L., and Schwartz, E.L. (2012b). Effects itation practitioners. Neurosci. Behav. Physiol.
of mindful-attention and compassion medita- 43C, 4873.
tion training on amygdala response to emotional Freeman, R. (2006). Assessment of cardiovascular
stimuli in an ordinary, non-meditative state. autonomic function. Clin. Neurophysiol. 117,
Front. Hum. Neurosci. 6,292. 716730.
Desbordes, G., Negi, L.T., Pace, T.W.W., Wallace, B.A., Friedman, B.H. (2007). An autonomic f lexibility-
Raison, C.L., and Schwartz, E.L. (2014). Effects neurovisceral integration model of anxi-
of eight-week meditation training on hippocam- ety and cardiac vagal tone. Biol. Psychol. 74,
pal volume: A comparison of mindful atten- 185199.
tion training and cognitively-based compassion Friedman, B.H., and Thayer, J.F. (1998). Autonomic
training. J. Altern. Complement. Med. 20,A24. balance revisited: Panic anxiety and heart rate
Diamond, D.M., Bennett, M.C., Fleshner, M., and variability. J. Psychosom. Res. 44, 133151.
Rose, G.M. (1992). Inverted-U relationship Fries, E., Dettenborn, L., and Kirschbaum, C. (2009).
between the level of peripheral corticosterone The cortisol awakening response (CAR): Facts
and the magnitude of hippocampal primed burst and future directions. Int. J. Psychophysiol.
potentiation. Hippocampus 2, 421430. 72,6773.
Dickerson, S.S., and Kemeny, M.E. (2004). Acute Fuller, B.F. (1992). The effects of stress-anxiety
stressors and cortisol responses: A theoretical and coping styles on heart rate variability. Int.
integration and synthesis of laboratory research. J.Psychophysiol. 12,8186.
Psychol. Bull. 130, 355391. Garrison, K.A., Scheinost, D., Constable, R.T., and
Ditto, B., Eclache, M., and Goldman, N. (2006). Brewer, J.A. (2014). BOLD signal and functional
Short-term autonomic and cardiovascular effects connectivity associated with loving kindness
of mindfulness body scan meditation. Ann. meditation. Brain Behav. 4, 337347.
Behav. Med. 32, 227234. Germer, C.K. (2009). The Mindful Path to Self-
Donath, M.Y., and Shoelson, S.E. (2011). Type 2 Compassion:Freeing Yourself from Destructive
diabetes as an inflammatory disease. Nat. Rev. Thoughts and Emotions (New York: Guilford
Immunol. 11, 98107. Press).
Eitan, E., Hutchison, E.R., and Mattson, M.P. (2014). Gex-Fabry, M., Jermann, F., Kosel, M., Rossier, M.F.,
Telomere shortening in neurological disor- Van der Linden, M., Bertschy, G., Bondolfi, G.,
ders: An abundance of unanswered questions. and Aubry, J.-M. (2012). Salivary cortisol
Trends Neurosci. 37, 256263. profiles in patients remitted from
364 Part III:Homeostatic Manipulators

recurrent depression: One-year follow-up of a Hayes, S.C., Strosahl, K.D., and Wilson, K.G. (1999).
mindfulness-based cognitive therapy trial. J. Acceptance and Commitment Therapy: An
Psychiatr. Res. 46,8086. Experiential Approach to Behavior Change
Gilbert, P. (2005). Compassion:Conceptualisations, (NewYork:Guilford Press).
Research and Use in Psychotherapy (London: Hellhammer, D.H., Wst, S., and Kudielka, B.M.
Routledge). (2009). Salivary cortisol as a biomarker in
Gilbert, P., and Procter, S. (2006). Compassionate stress research. Psychoneuroendocrinology 34,
mind training for people with high shame 163171.
and self-criticism: Overview and pilot study Herbert, T.B., and Cohen, S. (1993). Stress and
of a group therapy approach. Clin. Psychol. immunity in humans: A meta-analytic review.
Psychother. 13, 353379. Psychosom. Med. 55, 364379.
Glaser, R., Kiecolt-Glaser, J.K., Malarkey, W.B., and Hirsch, E.C., Vyas, S., and Hunot, S. (2012).
Sheridan, J.F. (1998). The influence of psycholog- Neuroinflammation in Parkinsons dis-
ical stress on the immune response to vaccines. ease. Parkinsonism Relat. Disord. 18 Suppl 1,
Ann. N.Y. Acad. Sci. 840, 649655. S210S212.
Goetz, J.L., Keltner, D., and Simon-Thomas, E. Hofmann, S.G., Grossman, P., and Hinton, D.E.
(2010). Compassion: An evolutionary analysis (2011). Loving-kindness and compassion medi-
and empirical review. Psychol. Bull. 136, 351374. tation:Potential for psychological interventions.
Gorman, J.M., and Sloan, R.P. (2000). Heart rate Clin. Psychol. Rev. 31, 11261132.
variability in depressive and anxiety disorders. Hoge, E.A., Bui, T.H.E., Metcalf, C., Pollack, M.H.,
Am. Heart J. 140, S77S83. and Simon, N.M. (2012). Mindfulness training
Gould, E., and Tanapat, P. (1999). Stress and hip- improves resilience: Reductions in adrenocorti-
pocampal neurogenesis. Biol. Psychiatry 46, cotropic hormone (ACTH) response to laboratory
14721479. stress. Neuropsychopharmacology 38, S321S322.
Gould, E., Tanapat, P., McEwen, B.S., Flgge, G., and Hoge, E.A., Chen, M.M., Orr, E., Metcalf, C.A.,
Fuchs, E. (1998). Proliferation of granule cell Fischer, L.E., Pollack, M.H., De Vivo, I., and
precursors in the dentate gyrus of adult monkeys Simon, N.M. (2013). Loving-kindness medita-
is diminished by stress. Proc. Natl. Acad. Sci. tion practice associated with longer telomeres in
U.S.A. 95, 31683171. women. Brain. Behav. Immun. 32, 159163.
Gould, E., Tanapat, P., Rydel, T., and Hastings, N. Hlzel, B.K., Carmody, J., Evans, K.C., Hoge, E.A.,
(2000). Regulation of hippocampal neurogenesis Dusek, J.A., Morgan, L., Pitman, R.K., and Lazar,
in adulthood. Biol. Psychiatry 48, 715720. S.W. (2010). Stress reduction correlates with
Goyal, M., Singh, S., Sibinga, E.M.S., Gould, N.F., structural changes in the amygdala. Soc. Cogn.
Rowland-Seymour, A., Sharma, R., Berger, Z., Affect. Neurosci. 5,1117.
Sleicher, D., Maron, D.D., Shihab, H.M., et al. Hlzel, B.K., Carmody, J., Vangel, M.G., Congleton, C.,
(2014). Meditation programs for psychologi- Yerramsetti, S.M., Gard, T., and Lazar, S.W.
cal stress and well-being: A systematic review (2011). Mindfulness practice leads to increases
and meta-analysis. JAMA Intern. Med. 174, in regional brain gray matter density. Psychiatry
357368. Res. Neuroimaging 191,3643.
Grant, J.A., Courtemanche, J., Duerden, E.G., Hlzel, B.K., Ott, U., Gard, T., Hempel, H.,
Duncan, G.H., and Rainville, P. (2010). Cortical Weygandt, M., Morgen, K., and Vaitl, D. (2008).
thickness and pain sensitivity in Zen meditators. Investigation of mindfulness meditation prac-
Emotion 10,4353. titioners with voxel-based morphometry. Soc.
Grossman, P., Niemann, L., Schmidt, S., and Walach, Cogn. Affect. Neurosci. 3,5561.
H. (2004). Mindfulness-based stress reduc- Hughes, J.W., Fresco, D.M., Myerscough, R., van
tion and health benefits: A meta-analysis. J. Dulmen M.H., Carlson, L.E., and Josephson, R.
Psychosom. Res. 57,3543. (2013). Randomized controlled trial of
Hamilton, A., and Sibson, N.R. (2013). Role of the mindfulness-based stress reduction for prehy-
systemic immune system in brain metastasis. pertension. Psychosom. Med. 75, 721728.
Mol. Cell. Neurosci. 53,4251. Jacobs, B.L., van Praag, H., and Gage, F.H.
Hayes, S.C. (2004). Acceptance and commit- (2000). Adult brain neurogenesis and psy-
ment therapy and the new behavior therapies. chiatry: A novel theory of depression. Mol.
In: Mindfulness and Acceptance: Expanding Psychiatry 5, 262269.
the Cognitive-Behavioral Tradition, S.C. Jacobs, T.L., Epel, E.S., Lin, J., Blackburn, E.H.,
Hayes, V.M. Follette, and M.M. Linehan, eds. Wolkowitz, O.M., Bridwell, D.A., Zanesco, A.P.,
(NewYork:Guilford Press), pp.129. Aichele, S.R., Sahdra, B.K., MacLean, K.A., etal.
Meditation 365

(2011). Intensive meditation training, immune hippocampal potentials: Increased effect with
cell telomerase activity, and psychological medi- aging. Science 245, 15051509.
ators. Psychoneuroendocrinology 36, 664681. Kiecolt-Glaser, J.K., Glaser, R., Gravenstein, S.,
Jazaieri, H., McGonigal, K., Jinpa, T., Doty, J.R., Malarkey, W.B., and Sheridan, J. (1996). Chronic
Gross, J.J., and Goldin, P.R. (2013). A random- stress alters the immune response to influenza
ized controlled trial of compassion cultivation virus vaccine in older adults. Proc. Natl. Acad.
training: Effects on mindfulness, affect, and Sci. U.S.A. 93, 30433047.
emotion regulation. Motiv. Emot. 38,2335. Kirschbaum, C., Pirke, K.M., and Hellhammer, D.H.
Johnson, W.L. (1982). Riding the Ox Home:AHistory (1993). The Trier Social Stress Testa tool for
of Meditation from Shamanism to Science investigating psychobiological stress responses
(London:Rider). in a laboratory setting. Neuropsychobiology
Jnsson, P. (2007). Respiratory sinus arrhythmia as a 28,7681.
function of state anxiety in healthy individuals. Klein, E., Cnaani, E., Harel, T., Braun, S., and
Int. J.Psychophysiol. 63,4854. Ben-Haim, S.A. (1995). Altered heart rate vari-
Joo, H.M., Lee, S.J., Chung, Y.G., and Shin, I.Y. ability in panic disorder patients. Biol. Psychiatry
(2010). Effects of mindfulness based stress 37,1824.
reduction program on depression, anxiety and Klimecki, O.M., Leiberg, S., Lamm, C., and Singer,
stress in patients with aneurysmal subarach- T. (2013). Functional neural plasticity and asso-
noid hemorrhage. J. Korean Neurosurg. Soc. 47, ciated changes in positive affect after compassion
345351. training. Cereb. Cortex 23, 15521561.
Josipovic, Z. (2010). Duality and nonduality in medi- Klimecki, O.M., Leiberg, S., Ricard, M., and Singer,
tation research. Conscious. Cogn. 19, 11191121. T. (2014). Differential pattern of functional brain
Kabat-Zinn, J. (1982). An outpatient program in plasticity after compassion and empathy train-
behavioral medicine for chronic pain patients ing. Soc. Cogn. Affect. Neurosci. 9, 873879.
based on the practice of mindfulness medita- Kok, B.E., Coffey, K.A, Cohn, M.A, Catalino, L.I.,
tion:Theoretical considerations and preliminary Vacharkulksemsuk, T., Algoe, S.B., Brantley,
results. Gen. Hosp. Psychiatry 4,3347. M., and Fredrickson, B.L. (2013). How positive
Kabat-Zinn, J. (1990). Full Catastrophe Living:Using emotions build physical health: Perceived posi-
the Wisdom of Your Body and Mind to Face tive social connections account for the upward
Stress, Pain, and Illness (New York: Delacorte spiral between positive emotions and vagal tone.
Press). Psychol. Sci. 24, 11231132.
Kabat-Zinn, J. (2003). Mindfulness-based interven- Kong, C.M., Lee, X.W., and Wang, X. (2013).
tions in context:Past, present, and future. Clin. Telomere shortening in human diseases. FEBS J.
Psychol. Sci. Pract. 144156. 280, 31803193.
Kabat-Zinn, J. (2013). Full Catastrophe Living: Kox, M., Stoffels, M., Smeekens, S.P., van Alfen, N.,
Using the Wisdom of Your Body and Mind to Gomes, M., Eijsvogels, T.M.H., Hopman, M.T.E.,
Face Stress, Pain, and Illness. Rev ed. (New van der Hoeven, J.G., Netea, M.G., and Pickkers,
York: Bantam). P. (2012). The influence of concentration/medi-
Kaliman, P., Alvarez-Lpez, M.J., Cosn-Toms, M., tation on autonomic nervous system activity
Rosenkranz, M.A., Lutz, A., and Davidson, R.J. and the innate immune response:Acase study.
(2014). Rapid changes in histone deacetylases and Psychosom. Med. 74, 489494.
inflammatory gene expression in expert medita- Kozhevnikov, M., Elliott, J., Shephard, J., and
tors. Psychoneuroendocrinology 40, 96107. Gramann, K. (2013). Neurocognitive and
Kemeny, M.E., Foltz, C., Cavanagh, J.F., Cullen, M., somatic components of temperature increases
Giese-Davis, J., Jennings, P., Rosenberg, E.L., during g-tummo meditation:Legend and reality.
Gillath, O., Shaver, P.R., Wallace, B.A., et al. PLoS One 8, e58244.
(2012). Contemplative/emotion training reduces Kozhevnikov, M., Louchakova, O., Josipovic, Z., and
negative emotional behavior and promotes pro- Motes, M.A. (2009). The enhancement of visuo-
social responses. Emotion 12, 338350. spatial processing efficiency through Buddhist
Kemp, A.H., Quintana, D.S., Gray, M.A., deity meditation. Psychol. Sci. 20, 645653.
Felmingham, K.L., Brown, K., and Gatt, J.M. Krieger, D.T., Allen, W., Rizzo, F., and Krieger, H.P.
(2010). Impact of depression and antidepressant (1971). Characterization of the normal temporal
treatment on heart rate variability:Areview and pattern of plasma corticosteroid levels. J. Clin.
meta-analysis. Biol. Psychiatry 67, 10671074. Endocrinol. Metab. 32, 266284.
Kerr, D.S., Campbell, L.W., Hao, S.Y., and Landfield, Kubota, Y., Sato, W., Toichi, M., Murai, T., Okada,
P.W. (1989). Corticosteroid modulation of T., Hayashi, A., and Sengoku, A. (2001). Frontal
366 Part III:Homeostatic Manipulators

midline theta rhythm is correlated with cardiac Luders, E., Thompson, P.M., Kurth, F., Hong, J.-Y.,
autonomic activities during the performance of Phillips, O.R., Wang, Y., Gutman, B.A., Chou,
an attention demanding meditation procedure. Y.-Y., Narr, K.L., and Toga, A.W. (2013b). Global
Cogn. Brain Res. 11, 281287. and regional alterations of hippocampal anat-
Kurth, F., Mackenzie-Graham, A., Toga, A.W., and omy in long-term meditation practitioners.
Luders, E. (2015). Shifting brain asymmetry:The Hum. Brain Mapp. 34, 33693375.
link between meditation and structural lateral- Lupien, S.J., de Leon, M., de Santi, S., Convit, A.,
ization. Soc. Cogn. Affect. Neurosci. 10,5561. Tarshish, C., Nair, N.P., Thakur, M., McEwen,
Lau, M.A., and McMain, S.F. (2005). Integrating B.S., Hauger, R.L., and Meaney, M.J. (1998).
mindfulness meditation with cognitive and Cortisol levels during human aging predict hip-
behavioural therapies:The challenge of combin- pocampal atrophy and memory deficits. Nat.
ing acceptance- and change-based strategies. Neurosci. 1,6973.
Can. J.Psychiatry 50, 863869. Lutz, A., Dunne, J.D., and Davidson, R.J. (2007).
Lazar, S.W., Kerr, C.E., Wasserman, R.H., Gray, Meditation and the neuroscience of conscious-
J.R., Greve, D.N., Treadway, M.T., McGarvey, ness: An introduction. In: The Cambridge
M., Quinn, B.T., Dusek, J.A., Benson, H., et al. Handbook of Consciousness, P.D. Zelazo, M.
(2005). Meditation experience is associated with Moscovitch, and E. Thompson, eds. (Cambridge,
increased cortical thickness. Neuroreport 16, UK:Cambridge Univ. Press), pp. 499551.
18931897. Lutz, A., Slagter, H.A., Dunne, J.D., and Davidson,
Lee, T.M.C., Leung, M.-K., Hou, W.-K., Tang, R.J. (2008). Attention regulation and monitoring
J.C.Y., Yin, J., So, K.-F., Lee, C.-F., and Chan, in meditation. Trends Cogn. Sci. 12, 163169.
C.C.H. (2012). Distinct neural activity associ- Maccioni, R.B., Rojo, L.E., Fernndez, J.A., and
ated with focused-attention meditation and Kuljis, R.O. (2009). The role of neuroimmuno-
loving-kindness meditation. PLoS One 7, e40054. modulation in Alzheimers disease. Ann. N.Y.
Leung, M.-K., Chan, C.C.H., Yin, J., Lee, C.-F., So, Acad. Sci. 1153, 240246.
K.-F., and Lee, T.M.C. (2013). Increased gray Maier, S.F. (2003). Bi-directional immune-brain
matter volume in the right angular and posterior communication: Implications for understand-
parahippocampal gyri in loving-kindness medi- ing stress, pain, and cognition. Brain. Behav.
tators. Soc. Cogn. Affect. Neurosci. 8,3439. Immun. 17,6985.
Licht, C.M.M., de Geus, E.J.C., van Dyck, R., and Makransky, J. (2007). Awakening Through
Penninx, B.W.J.H. (2009). Association between Love: Unveiling Your Deepest Goodness
anxiety disorders and heart rate variability in the (Somerville, MA:Wisdom Publications).
Netherlands Study of Depression and Anxiety Malik, M., Bigger, J.T., Camm, A.J., Kleiger, R.E.,
(NESDA). Psychosom. Med. 71, 508518. Malliani, A., Moss, A.J., and Schwartz, P.J.
Linehan, M.M. (1993). Cognitive-Behavioral (1996). Heart rate variability:Standards of mea-
Treatment of Borderline Personality Disorder surement, physiological interpretation, and clin-
(NewYork:Guilford Press). ical use. Eur. Heart J. 17,354.
Luders, E., Toga, A.W., Lepore, N., and Gaser, C. Malliani, A., Pagani, M., Lombardi, F., and Cerutti,
(2009). The underlying anatomical correlates of S. (1991). Cardiovascular neural regulation
long-term meditation: Larger hippocampal and explored in the frequency domain. Circulation
frontal volumes of gray matter. NeuroImage 45, 84, 482492.
672678. Malykhin, N.V, Carter, R., Seres, P., and Coupland,
Luders, E., Clark, K., Narr, K.L., and Toga, A.W. N.J. (2010). Structural changes in the hippocam-
(2011). Enhanced brain connectivity in pus in major depressive disorder:Contributions
long-term meditation practitioners. Neuroimage of disease and treatment. J. Psychiatry Neurosci.
57, 13081316. 35, 337343.
Luders, E., Kurth, F., Mayer, E.A., Toga, A.W., Marques, A.H., Silverman, M.N., and Sternberg, E.M.
Narr, K.L., and Gaser, C. (2012). The unique (2010). Evaluation of stress systems by apply-
brain anatomy of meditation practitio- ing noninvasive methodologies: Measurements
ners: Alterations in cortical gyrification. Front. of neuroimmune biomarkers in the sweat,
Hum. Neurosci.6,34. heart rate variability and salivary cortisol.
Luders, E., Kurth, F., Toga, A.W., Narr, K.L., and Neuroimmunomodulation 17, 205208.
Gaser, C. (2013a). Meditation effects within the Mascaro, J.S., Rilling, J.K., Negi, L.T., and Raison,
hippocampal complex revealed by voxel-based C.L. (2013). Compassion meditation enhances
morphometry and cytoarchitectonic probabilis- empathic accuracy and related neural activity.
tic mapping. Front. Psychol. 4,398. Soc. Cogn. Affect. Neurosci. 8,4855.
Meditation 367

Mather, K.A., Jorm, A.F., Parslow, R.A., and with lowered heart rate variability and increased
Christensen, H. (2011). Is telomere length a bio- trait anxiety in healthy adults. Hum. Brain
marker of aging? Areview. J. Gerontol. A.Biol. Mapp. 30,4758.
Sci. Med. Sci. 66, 202213. Nesvold, A., Fagerland, M.W., Davanger, S.,
Matousek, R.H., Dobkin, P.L., and Pruessner, J.C. Ellingsen, ., Solberg, E.E., Holen, A., Sevre, K.,
(2010). Cortisol as a marker for improve- and Atar, D. (2012). Increased heart rate variabil-
ment in mindfulness-based stress reduction. ity during nondirective meditation. Eur. J.Prev.
Complement. Ther. Clin. Pract. 16,1319. Cardiol. 19, 773780.
Matousek, R.H., Pruessner, J.C., and Dobkin, Njajou, O.T., Hsueh, W.-C., Blackburn, E.H.,
P.L. (2011). Changes in the cortisol awaken- Newman, A.B., Wu, S.-H., Li, R., Simonsick,
ing response (CAR) following participation in E.M., Harris, T.M., Cummings, S.R., and
mindfulness-based stress reduction in women Cawthon, R.M. (2009). Association between
who completed treatment for breast cancer. telomere length, specific causes of death, and
Complement. Ther. Clin. Pract. 17,6570. years of healthy life in health, aging, and body
McCraty, R., Atkinson, M., Tomasino, D., and composition, a population-based cohort study. J.
Stuppy, W.P. (2001). Analysis of twenty-four Gerontol. A.Biol. Sci. Med. Sci. 64, 860864.
hour heart rate variability in patients with panic Oeseburg, H., de Boer, R.A., van Gilst, W.H., and van
disorder. Biol. Psychol. 56, 131150. der Harst, P. (2010). Telomere biology in healthy
McEwen, B.S. (1998). Protective and damaging aging and disease. Pflugers Arch. 459, 259268.
effects of stress mediators. N. Engl. J.Med. 338, Oke, S.L., and Tracey, K.J. (2009). The inflammatory
171179. reflex and the role of complementary and alter-
McEwen, B.S. (2003). Mood disorders and allostatic native medical therapies. Ann. N.Y. Acad. Sci.
load. Biol. Psychiatry 54, 200207. 1172, 172180.
McEwen, B.S. (2004). Protective and damag- Orme-Johnson, D.W., Barnes, V.A., Hankey, A.M.,
ing effects of the mediators of stress and and Chalmers, R.A. (2005). Reply to crit-
adaptation: Allostasis and allostatic load. ics of research on transcendental meditation
In: Allostasis, Homeostasis, and the Costs in the prevention and control of hyperten-
of Physiological Adaptation, J. Schulkin, ed. sion. J. Hypertens. 23, 11071108; author reply
(Cambridge, UK: Cambridge Univ. Press), 11081109.
pp.6598. Ornish, D., Lin, J., Chan, J.M., Epel, E., Kemp, C.,
McEwen, B.S. (2006). Protective and damaging Weidner, G., Marlin, R., Frenda, S.J., Magbanua,
effects of stress mediators: Central role of the M.J.M., Daubenmier, J., et al. (2013). Effect of
brain. Dialogues Clin. Neurosci. 8, 367381. comprehensive lifestyle changes on telomer-
McEwen, B.S., and Seeman, T.E. (1999). Protective ase activity and telomere length in men with
and damaging effects of mediators of stress. biopsy-proven low-risk prostate cancer: 5-year
Elaborating and testing the concepts of allo- follow-up of a descriptive pilot study. Lancet
stasis and allostatic load. Ann. N.Y. Acad. Sci. Oncol. 14, 11121120.
896,3047. Ornish, D., Lin, J., Daubenmier, J., Weidner, G.,
McEwen, B.S., and Stellar, E. (1993). Stress and the Epel, E.S., Kemp, C., Magbanua, M., Marlin, R.,
individual: Mechanisms leading to disease. Yglecias, L., and Carroll, P. (2008). Increased
Arch. Intern. Med. 153, 20932101. telomerase activity and comprehensive life-
Mehling, W.E., Wrubel, J., Daubenmier, J.J., Price, style changes: A pilot study. Lancet Oncol. 9,
C.J., Kerr, C.E., Silow, T., Gopisetty, V., and 10481057.
Stewart, A.L. (2011). Body awareness: A phe- Ospina, M.B., Bond, K., Karkhaneh, M., Tjosvold, L.,
nomenological inquiry into the common ground Vandermeer, B., Liang, Y., Bialy, L., Hooton, N.,
of mind-body therapies. Philos. Ethics. Humanit. Buscemi, N., Dryden, D.M., et al. (2007).
Med.6,6. Meditation practices for health: State of the
More, S.V., Kumar, H., Kim, I.S., Song, S.-Y., and research. Evid. Rep. Technol. Assess.,1263.
Choi, D.-K. (2013). Cellular and molecular medi- Ozawa-de Silva, B.R., and Negi, L.T. (2013).
ators of neuroinflammation in the pathogenesis Cognitively-based compassion training (CBCT):
of Parkinsons disease. Mediators Inflamm. 2013, Protocol and key concepts. In: Compassion:
952375. Bridging Practice and Science, T. Singer, and
Mujica-Parodi, L.R., Korgaonkar, M., Ravindranath, M. Bolz, eds. (Munich: Max Planck Society),
B., Greenberg, T., Tomasi, D., Wagshul, M., pp. 416438.
Ardekani, B., Guilfoyle, D., Khan, S., Zhong, Y., Pace, T.W.W., Negi, L.T., Adame, D.D., Cole, S.P.,
et al. (2009). Limbic dysregulation is associated Sivilli, T.I., Brown, T.D., Issa, M.J., and Raison,
368 Part III:Homeostatic Manipulators

C.L. (2009). Effect of compassion medita- marker for the assessment of adrenocortical
tion on neuroendocrine, innate immune and activity. Life Sci. 61, 25392549.
behavioral responses to psychosocial stress. Rao, U., Chen, L.-A., Bidesi, A.S., Shad, M.U.,
Psychoneuroendocrinology 34,8798. Thomas, M.A., and Hammen, C.L. (2010).
Pace, T.W.W., Negi, L.T., Dodson-Lavelle, B., Hippocampal changes associated with early-life
Ozawa-de Silva, B.R., Reddy, S.D., Cole, S.P., adversity and vulnerability to depression. Biol.
Danese, A., Craighead, L.W., and Raison, C.L. Psychiatry 67, 357364.
(2013). Engagement with cognitively-based Robins, C.J., Schmidt, H.I., and Linehan,
compassion training is associated with reduced M.M. (2004). Dialectical behavior therapy:
salivary C-reactive protein from before to after Synthesizing radical acceptance with skill-
training in foster care program adolescents. ful means. In: Mindfulness and Acceptance:
Psychoneuroendocrinology 38, 294299. Expanding the Cognitive-Behavioral Tradition,
Pace, T.W.W., Negi, L.T., Sivilli, T.I., Issa, M.J., Cole, S.P., S.C. Hayes, V.M. Follette, and M.M. Linehan,
Adame, D.D., and Raison, C.L. (2010). Innate eds. (NewYork:Guilford Press), pp.3044.
immune, neuroendocrine and behavioral Rosenkranz, M.A., Davidson, R.J., MacCoon, D.G.,
responses to psychosocial stress do not predict Sheridan, J.F., Kalin, N.H., and Lutz, A. (2013). A
subsequent compassion meditation practice comparison of mindfulness-based stress reduc-
time. Psychoneuroendocrinology 35, 310315. tion and an active control in modulation of neu-
Pagnoni, G., and Cekic, M. (2007). Age effects on rogenic inflammation. Brain. Behav. Immun. 27,
gray matter volume and attentional perfor- 174184.
mance in Zen meditation. Neurobiol. Aging 28, Rosenthal, N.E. (2011). Transcendence: Healing
16231627. and Transformation Through Transcendental
Parati, G., and Steptoe, A. (2004). Stress reduc- Meditation (NewYork:Tarcher).
tion and blood pressure control in hyperten- Rubia, K. (2009). The neurobiology of meditation
sion: A role for transcendental meditation? J. and its clinical effectiveness in psychiatric disor-
Hypertens. 22, 20572060. ders. Biol. Psychol. 82,111.
Peeters, F., Nicolson, N.A, and Berkhof, J. (2004). Rubio-Perez, J.M., and Morillas-Ruiz, J.M. (2012).
Levels and variability of daily life cortisol A review: Inflammatory process in Alzheimers
secretion in major depression. Psychiatry Res. disease, role of cytokines. Sci. World J. 2012,
126,113. 756357.
Peng, C.-K., Henry, I.C., Mietus, J.E., Hausdorff, Salzberg, S. (1995). Loving-Kindness: The
J.M., Khalsa, G., Benson, H., and Goldberger, Revolutionary Art of Happiness
A.L. (2004). Heart rate dynamics during three (Boston:Shambhala Publications).
forms of meditation. Int. J.Cardiol. 95,1927. Santarelli, L., Saxe, M., Gross, C., Surget, A.,
Peng, C.-K., Mietus, J.E., Liu, Y., Khalsa, G., Battaglia, F., Dulawa, S., Weisstaub, N., Lee, J.,
Douglas, P.S., Benson, H., and Goldberger, A.L. Duman, R.S., Arancio, O., et al. (2003).
(1999). Exaggerated heart rate oscillations dur- Requirement of hippocampal neurogenesis for
ing two meditation techniques. Int. J. Cardiol. the behavioral effects of antidepressants. Science
70, 101107. 301, 805809.
Phongsuphap, S., Pongsupap, Y., Chandanamattha, Sapolsky, R.M. (1992). Stress, the Aging Brain, and
P., and Lursinsap, C. (2008). Changes in heart the Mechanisms of Neuron Death (Cambridge,
rate variability during concentration meditation. MA:MIT Press).
Int. J.Cardiol. 130, 481484. Saxe, R., and Kanwisher, N. (2003). People think-
Pizza, V., Agresta, A., DAcunto, C.W., Festa, M., ing about thinking people: The role of the
and Capasso, A. (2011). Neuroinflammation and temporo-parietal junction in theory of mind.
ageing:Current theories and an overview of the Neuroimage 19, 18351842.
data. Rev. Recent Clin. Trials 6, 189203. Schmalzl, L., Crane-Godreau, M.A., and Payne, P.
Porges, S.W. (2011). The Polyvagal Theory: (2014). Movement-based embodied contempla-
Neurophysiological Foundations of Emotions, tive practices:Definitions and paradigms. Front.
Attachment, Communication, and Self- Hum. Neurosci. 8,205.
Regulation (Norton Series on Interpersonal Schommer, N.C., Hellhammer, D.H., and
Neurobiology) (New York: W. W. Norton & Co.). Kirschbaum, C. (2003). Dissociation between
Pruessner, J.C., Wolf, O.T., Hellhammer, D.H., reactivity of the hypothalamus-pituitary-adrenal
Buske-Kirschbaum, A., von Auer, K., Jobst, S., axis and the sympathetic-adrenal-medullary sys-
Kaspers, F., and Kirschbaum, C. (1997). Free cor- tem to repeated psychosocial stress. Psychosom.
tisol levels after awakening:Areliable biological Med. 65, 450460.
Meditation 369

Schulkin, J. (2004). Allostasis, Homeostasis, (2014). Cognitively-based compassion training


and the Costs of Physiological Adaptation yields increase in cortical thickness after eight
(NewYork:Cambridge Univ. Press). weeks. Paper presented at: 20th annual meet-
Seeman, T.E., Gruenewald, T., Karlamangla, A., ing, Organization for Human Brain Mapping,
Sidney, S., Liu, K., McEwen, B.S., and Schwartz, Hamburg,June.
J.E. (2010). Modeling multisystem biologi- Slagter, H.A., Davidson, R.J., and Lutz, A. (2011).
cal risk in young adults: The Coronary Artery Mental training as a tool in the neuroscientific
Risk Development in Young Adults Study. Am. study of brain and cognitive plasticity. Front.
J.Hum. Biol. 22, 463472. Hum. Neurosci.5,17.
Seeman, T.E., McEwen, B.S., Rowe, J.W., and Singer, Sparkman, N.L., and Johnson, R.W. (2008).
B.H. (2001). Allostatic load as a marker of cumu- Neuroinflammation associated with aging sensi-
lative biological risk:MacArthur studies of suc- tizes the brain to the effects of infection or stress.
cessful aging. Proc. Natl. Acad. Sci. U.S.A. 98, Neuroimmunomodulation 15, 323330.
47704775. Starkweather, A.R., Alhaeeri, A.A., Montpetit, A.,
Segal, Z. V., Williams, J.M.G., and Teasdale, J.D. Brumelle, J., Filler, K., Montpetit, M., Mohanraj,
(2013). Mindfulness-Based Cognitive Therapy for L., Lyon, D.E., and Jackson-Cook, C.K. (2014).
Depression. 2nd ed. (NewYork:Guilford Press). An integrative review of factors associated with
Sephton, S.E., Lush, E., Dedert, E.A., Floyd, A.R., telomere length and implications for biobehav-
Rebholz, W.N., Dhabhar, F.S., Spiegel, D., and ioral research. Nurs. Res. 63,3650.
Salmon, P. (2013). Diurnal cortisol rhythm as a Stein, P.K., Bosner, M.S., Kleiger, R.E., and Conger,
predictor of lung cancer survival. Brain. Behav. B.M. (1994). Heart rate variability: A measure
Immun. 30 Suppl, S163S170. of cardiac autonomic tone. Am. Heart J. 127,
Sephton, S.E., Sapolsky, R.M., Kraemer, H.C., and 13761381.
Spiegel, D. (2000). Diurnal cortisol rhythm as Steptoe, A., Hamer, M., and Chida, Y. (2007). The
a predictor of breast cancer survival. J. Natl. effects of acute psychological stress on circulat-
Cancer Inst. 92, 9941000. ing inflammatory factors in humans: A review
Shah, P.J., Ebmeier, K.P., Glabus, M.F., and Goodwin, and meta-analysis. Brain. Behav. Immun. 21,
G.M. (1998). Cortical grey matter reductions 901912.
associated with treatment-resistant chronic uni- Sterling, P., and Eyer, J. (1988). Allostasis:Anew para-
polar depression. Controlled magnetic resonance digm to explain arousal pathology. In:Handbook
imaging study. Br. J.Psychiatry 172, 527532. of Life Stress, Cognition and Health, S. Fisher,
Shapiro, D.H. Jr. (1982). Overview: Clinical and and J. Reason, eds. (NewYork:John Wiley), pp.
physiological comparison of meditation with 629649.
other self-control strategies. Am. J. Psychiatry Takahashi, T., Murata, T., Hamada, T., Omori,
139, 267274. M., Kosaka, H., Kikuchi, M., Yoshida, H., and
Shapiro, D.H. Jr., and Walsh, R.N. (1984). Meditation: Wada, Y. (2005). Changes in EEG and auto-
Classic and Contemporary Perspectives (New nomic nervous activity during meditation and
York: Aldine). their association with personality traits. Int.
Sheline, Y.I., Sanghavi, M., Mintun, M.A., and Gado, J.Psychophysiol. 55, 199207.
M.H. (1999). Depression duration but not age Tang, Y.-Y., Hlzel, B.K., & Posner, M.I. (2015). The
predicts hippocampal volume loss in medically neuroscience of mindfulness meditation. Nat.
healthy women with recurrent major depression. Rev. Neurosci. 16, 213225.
J. Neurosci. 19, 50345043. Tang, Y.-Y., Lu, Q., Fan, M., Yang, Y., and Posner,
Sheline, Y.I., Wang, P.W., Gado, M.H., Csernansky, M.I. (2012). Mechanisms of white matter changes
J.G., and Vannier, M.W. (1996). Hippocampal induced by meditation. Proc. Natl. Acad. Sci.
atrophy in recurrent major depression. Proc. U.S.A. 109, 1057010574.
Natl. Acad. Sci. U.S.A. 93, 39083913. Tang, Y.-Y., Lu, Q., Geng, X., Stein, E.A., Yang, Y.,
Simen, A.A., Bordner, K.A., Martin, M.P., Moy, L.A., and Posner, M.I. (2010). Short-term medita-
and Barry, L.C. (2011). Cognitive dysfunction tion induces white matter changes in the ante-
with aging and the role of inflammation. Ther. rior cingulate. Proc. Natl. Acad. Sci. U.S.A. 107,
Adv. Chronic Dis. 2, 175195. 1564915652.
Singer, T., and Bolz, M. (2013). Compassion:Bridging Tang, Y.-Y., Ma, Y., Fan, Y., Feng, H., Wang, J., Feng,
Practice and Science (Munich: Max Planck S., Lu, Q., Hu, B., Lin, Y., Li, J., et al. (2009).
Society). Central and autonomic nervous system interac-
Singleton, O., Desbordes, G., Negi, L.T., Pace, T.W.W., tion is altered by short-term meditation. Proc.
Wallace, B.A., Raison, C.L., and Schwartz, E.L. Natl. Acad. Sci. U.S.A. 106, 88658870.
370 Part III:Homeostatic Manipulators

Teasdale, J.D., Segal, Z.V., and Williams, J.M. (1995). Wallace, R.K., Benson, H., and Wilson, A.F. (1971).
How does cognitive therapy prevent depres- A wakeful hypometabolic physiologic state. Am.
sive relapse and why should attentional control J.Physiol. 221, 795799.
(mindfulness) training help? Behav. Res. Ther. Wallmark, E., Safarzadeh, K., Daukantait, D.,
33,2539. and Maddux, R.E. (2012). Promoting altruism
Thayer, J.F., hs, F., Fredrikson, M., Sollers, J.J., through meditation: An 8-week randomized
and Wager, T.D. (2012). A meta-analysis of controlled pilot study. Mindfulness 4, 223234.
heart rate variability and neuroimaging stud- Watkins, L., Grossman, P., Krishnan, R., and
ies: Implications for heart rate variability as a Sherwood, A. (1998). Anxiety and vagal control
marker of stress and health. Neurosci. Biobehav. of heart rate. Psychosom. Med. 60, 498502.
Rev. 36, 747756. Weng, H.Y., Fox, A.S., Shackman, A.J., Stodola, D.E.,
Thayer, J.F., Hansen, A.L., Saus-Rose, E., and Caldwell, J.Z.K., Olson, M.C., Rogers, G.M.,
Johnsen, B.H. (2009). Heart rate variability, and Davidson, R.J. (2013). Compassion training
prefrontal neural function, and cognitive per- alters altruism and neural responses to suffering.
formance: The neurovisceral integration per- Psychol. Sci. 24, 11711180.
spective on self-regulation, adaptation, and Witek-Janusek, L., Albuquerque, K., Chroniak, K.R.,
health. Ann. Behav. Med. 37, 141153. Chroniak, C., Durazo-Arvizu, R., and Mathews,
Travis, F., and Shear, J. (2010a). Focused atten- H.L. (2008). Effect of mindfulness based stress
tion, open monitoring and automatic reduction on immune function, quality of life and
self-transcending: Categories to organize medi- coping in women newly diagnosed with early stage
tations from Vedic, Buddhist and Chinese tradi- breast cancer. Brain. Behav. Immun. 22, 969981.
tions. Conscious. Cogn. 19, 11101118. Wolever, R.Q., Bobinet, K.J., McCabe, K., Mackenzie,
Travis, F., and Shear, J. (2010b). Reply to Josipovic: E.R., Fekete, E., Kusnick, C.A., and Baime, M.
Duality and non-duality in meditation research. (2012). Effective and viable mind-body stress
Conscious. Cogn. 19, 11221123. reduction in the workplace: A randomized con-
Van Overwalle, F. (2009). Social cognition and the trolled trial. J. Occup. Health Psychol. 17, 246258.
brain: A meta-analysis. Hum. Brain Mapp. 30, Wu, S.-D., and Lo, P.-C. (2008). Inward-attention
829858. meditation increases parasympathetic activ-
Vermetten, E., Vythilingam, M., Southwick, ity: A study based on heart rate variability.
S.M., Charney, D.S., and Bremner, J.D. (2003). Biomed. Res. 29, 245250.
Long-term treatment with paroxetine increases Yang, E. V, and Glaser, R. (2002). Stress-associated
verbal declarative memory and hippocampal immunomodulation and its implications for
volume in posttraumatic stress disorder. Biol. responses to vaccination. Expert Rev. Vaccines
Psychiatry 54, 693702. 1, 453459.
Vestergaard-Poulsen, P., van Beek, M., Skewes, J., Young, J.D.-E., and Taylor, E. (1998). Meditation as a
Bjarkam, C.R., Stubberup, M., Bertelsen, J., voluntary hypometabolic state of biological esti-
and Roepstorff, A. (2009). Long-term medi- vation. News Physiol. Sci. 13, 149153.
tation is associated with increased gray mat- Zhu, H., Belcher, M., and van der Harst, P. (2011).
ter density in the brain stem. Neuroreport 20, Healthy aging and disease: Role for telomere
170174. biology? Clin. Sci. (Lond). 120, 427440.
22
Neurotrophic Regulation
inNeurorestoration oftheBrain
C A I X I N S U , M I C H E L R AT H B O N E , A N D S H U C U I J I A N G

HISTORY N E U R O R E S T O R AT I O N ,
O F N E U R O R E S T O R AT I O N TOBRING BACK
As far back as 1700 bc, ancient Egyptians docu- T H E H O M E O S TA S I S
mented their observations that injuries to the OFTHECNS
head and spinal cord resulted in permanent Equipped with the knowledge from scientific
paralysis, loss of speech, and other functions that studies of the CNS, we are now beginning to
were not recoverable (Cunha, 1949). Over time, understand how the CNS integrates the regula-
clinicians began to focus on neuroprotective tory and restorative forces of the body to foster
measures that limit further injury after central health and cope with environmental changes
nervous system (CNS) insults and surgeries to after CNS disorders. Under normal physiologi-
ablate, remove, or reduce the circumstances that cal conditions, the CNS manages a multitude of
impact the functionality. highly complex interactions to maintain its bal-
About 100years ago, with the development of ance and return systems to functioning within a
microscopy techniques, scientists began to study normal range. Homeostasis is achieved through
the CNS in more detail at the cellular level. The a delicate balance of electrical, chemical, molecu-
work from Santiago Ramon y Cajal, the father lar, cellular, hormonal, immunological, spatial,
of modern neuroscience, laid the foundation for and behavioral factors. To function properly,
future studies on CNS function restoration. He neurons require a fairly strict electrical potential
pointed out that the restoration of nerve paths gradient across the neural membranes, and this
in the CNS is frustrated by two negative condi- gradient is determined by the acid-base balance
tions:the lack of substances able to sustain and and the concentration of ions, both intra- and
invigorate the indolent growth of the sprouts and extracellularly, the availability of neurotrans-
the absence of catalytic agents capable of attract- mitters and receptor sites, and the blood levels
ing and directing the axonic current to its desti- of a variety of metals (Leisman, 1990). However,
nation (Lobato,2008). neurons do not function independently; they are
Neurorestoration, as a new concept, emerged affected by other cellular or even bodily activi-
in the 1990s. With the escalating development ties. For example, astrocytes, the guardians of
in the basic science field, in particular molecu- neurons, constantly modulate neuronal activity
lar biology, electrophysiology, and computa- and support neuron survival and synaptic trans-
tional neuroscience, scientists became able to mission by regulating neurotransmitters and
examine the CNS in more detail, from molecu- neurotrophic factors (Benarroch, 2005; Seifert
lar and cellular levels to circuitry and system and Steinhauser, 2013). Even respiration and
levels. Neuromodulation, gene therapy, and heart rate (which change blood pH, oxygen, and
cellular grafting techniques were fervently dis- glucose availability in the CNS) can contribute
cussed in scientific research. While there is no to maintaining the complex homeostasis of the
general agreement on the standard definition CNS. The interconnection of the homeostasis
of neurorestoration, Bednar defined it as the web is virtually endless.
structural and/or functional improvement in the Homeostatic regulatory mechanisms only
pathophysiology of the CNS following an insult work within certain limits to buffer the CNS
(Bednar and Perry,2012). from changes in the internal milieu. In 1939,
372 Part III:Homeostatic Manipulators

Walter Cannon postulated the theory that there adult humans. These findings not only confirmed
is a safety threshold in homeostatic regulation that CNS axons were able to regenerate but also
that ensures that variations from basal physi- reinforced a concept that has driven virtually all
ological levels (set points) do not reach danger- subsequent research in this field: that the CNS
ous extremes that might impair the functions of nonpermissive environment inhibits axonal
the cells or threaten the existence of the organ- regeneration following injury.
ism because adaptive reactions are automatically Neurorestoration is both a process and a goal
triggered to return the affected physiological and includes, but is not limited to, neuroregener-
system to a basal state (Cacioppo and Berntson, ation. The goal of neurorestoration is to improve
2011). Beyond that threshold, under a pathologi- the structure of the tissue or organ involved
cal condition such as a CNS disorder, the system (Bednar, 2008). Ideally, neuroprotection at the
will react by utilizing any or all compensatory early stage is mandatory to limit further injury.
bodily mechanisms to re-establish the dynamic However, effective neuroprotectants have largely
homeostasis necessary for proper function- failed over a range of CNS injury types and mod-
ing. Unfortunately, CNS disorders such as head els, and the search for an effective therapy is
trauma, spinal cord injury, stroke, epilepsy, and ongoing. Angiogenesis, as an antecedent require-
Alzheimers disease (AD) do not all affect the ment for neuroplasticity, stimulates endogenous
same area or portion of neural tissue. Rather, recovery mechanisms, including neurogen-
they result in a disturbance in the delicate and esis, synaptogenesis, and neuronal and synaptic
precisely tuned homeostatic environment. plasticity. These events are all involved in the
long-term repair and restoration process of the
N E U R O R E S T O R AT I O N , brain after an ischemic event (Ergul etal.,2012).
INCLUDING BUTNOT LIMITED
T O N E U R O R E G E N E R AT I O N AN APPROACH TOREDUCE
The adult CNS normally has a very limited capac- THENONPERMISSIVE AND
ity to recover after the loss of function due to the S T I M U L AT E T H E P E R M I S S I V E
difficulty of neuronal regeneration after axon ENVIRONMENTCUES
injury. For a very long time, the prevailing dogma CNS disorders are caused by neurodevelop-
held that there is no regeneration in the adult mental insults or occur when neurodegenera-
CNS. However, some optimistic scientists contin- tive processes disrupt the homeostatic balance.
ued conducting research on CNS repair. In a 1974 A detailed discussion of the pathological fea-
study, researchers observed widespread collateral tures of CNS disorders is beyond the scope of
sprouting of nociceptive dorsal root axons in the this chapter and has been discussed elsewhere
dorsal horn of the spinal cord following rhizot- (Dumont etal., 2001; Freire, 2012; Menon, 1999).
omy (Goldberger and Murray, 1974). Similarly, However, only when the pathophysiological
others demonstrated that lesioned noradren- changes and principles governing recovery of
ergic and cholinergic neurons were capable of function are more adequately understood will it
extending axons over long distances within the be possible to formulate strategies to restore the
brain and spinal cord (Bjorklund et al., 1971; function ofCNS.
1975; Katzman etal., 1971). Later, using the new As Cajal opined more than a century ago,
horseradish peroxidase tracing technique, David and as many researchers (Hou etal., 2008; Yang
and Aguayo (1981) examined the origin, termi- and Yang, 2012) have echoed more recently,
nation, and length of axonal growth after focal the fundamental inhibition to CNS functional
CNS injury in the adult rat. When peripheral recovery is the nonpermissive environment.
nerve segments were used as bridges between Under extreme conditions, such as spinal cord or
the medulla and spinal cord, they demonstrated brain trauma, the tissue goes through mechani-
that axons from neurons at both these levels cal injury:compression, impact, distraction, lac-
grew approximately 30 mm. The turning point eration/transection, and so on. Neurons that do
for CNS regeneration, however, was the publica- survive the initial injury are further bombarded
tion of the landmark paper from Eriksson etal. by secondary injuries such as neurogenic shock,
(1998). For the first time, they showed that new hemorrhage, ischemia, excitotoxicity, calcium
neurons, as defined by bromodeoxyuridine labe- and fluid-electrolyte disturbances; immunologic
ling and neuronal markers, were generated from injury; apoptosis; and disturbances in mito-
dividing progenitor cells in the dentate gyrus of chondrial function (Dumont etal., 2001). These
Neurotrophic Regulation in Neurorestoration of theBrain 373

secondary injury mechanisms can exacerbate neurotrophic factors, growth-promoting extra-


damage, limit restorative processes, and contrib- cellular matrix molecules, cell adhesion mol-
ute to overall morbidity and mortality. ecules, and other small molecules. While they
Regeneration follows degeneration. A bur- are present in a healthy and intact CNS, stimula-
geoning body of evidence has shown that the tion by the endogenous regulatory mechanisms
failure of axon regeneration after a CNS insult is after CNS injury has not yielded a positive result
due to an inadequate or inappropriate regenera- as the molecules are not sustained long enough
tive response from damaged CNS axons and to to promote regeneration. If their outgrowth and
a CNS environment that inhibits regeneration. regenerative abilities could be extended, the end
This inhibitory environment actually contains result might change.
many molecules that promote axon growth as As has been widely demonstrated, neuro-
well as molecules that inhibit it, but the balance trophic factors play a pivotal role in neurores-
of activities in the damaged CNS does not favor toration and neuroprotection in CNS disorders
the regeneration of adult CNS axons, with the such as Alzheimers disease (AD), Parkinsons
inhibitory factors usually upregulated after the disease (PD), amyotrophic lateral sclerosis (ALS),
disorder. Huntingtons disease, spinal cord and brain inju-
CNS neuronal inhibitors fall into three classes. ries, and other neurodegenerative and neuro-
The first class is secreted by glial scar, which is the traumatic pathologies (Aloe etal., 2012; Connor
main physical and biochemical barrier to axonal and Dragunow, 1998; Dreyfus, 1989). While
regeneration after CNS injury (Bovolenta et al., our understanding of their roles in CNS disor-
1991; Sandvig et al., 2004). Numerous studies ders is not complete, nerve growth factor (NGF),
have identified extracellular matrix component brain-derived neurotrophic factor (BDNF), and
chondroitin sulphate proteoglycan (Fitch and fibroblast growth factor 2 (FGF2) have been
Silver, 1997; Mironova and Giger, 2013)and kera- examined in numerous studies, which investi-
tan sulfate proteoglycan (Imagama etal., 2011)as gated both the preventive and therapeutic effects
key molecules contributing to glial scar inhibi- on neuronal survival as well as the negative
tion. An abundance of individual chondroitin impact of neurotrophin stimulation. Another
sulphate proteoglycans have been found, includ- emerging area of interest is the therapeutic
ing neurocan (Asher etal., 2000; McKeon etal., potential of purinergic signaling in the patho-
1999), neural/glial antigen (Levine, 1994), versi- physiology of CNS disorders. Geoffrey Burnstock
can (Asher et al., 2002), and brevican (Yamada (1972, 2013)introduced the concept of purinergic
etal.,1997). signaling when he presented the first evidence of
Compelling evidence has also revealed that the action of adenosine triphosphate (ATP) as a
CNS myelin formed by mature oligodendrocytes neurotransmitter. Broadly speaking, the puriner-
profoundly inhibits neurite outgrowth. A num- gic system includes purines in many different
ber of myelination-related inhibitory molecules formats: purine bases such as adenine and gua-
have been identified, including myelin-associated nine; their corresponding nucleosides (adenosine
glycoprotein (Chiquet, 1989; Domeniconi et al., and guanosine and their metabolic products), as
2002; Filbin, 1995), NogoA (Chen et al., 2000; well as purine nucleotides. With the cloning of
Schwab, 2004), and oligodendrocyte myelin gly- various purinergic receptor subtypes, such as P1
coprotein (Guo et al., 2007; Raiker et al., 2010). and P2, the roles of adenosine and ATP as neu-
In addition to glial scar and myelination-related rotransmitters and neuromodulators in the CNS
inhibitors, several axon guidance molecules have have become well accepted (Burnstock, 2013).
been found to cause growth cone collapse after Chapter 2 of this book discusses adenosine and
CNS injury, including semaphorins, ephrins, ATP in the homeostatic control of CNS disorders
netrins, and slits (Fan et al., 2013; Pasterkamp in detail.
and Verhaagen, 2006). This list of inhibitory mol- In contrast to the enthusiasm shown for
ecules is by no means exhaustive; many more adenine-based purines, little attention has
have yet to be identified. It is generally believed been given to the potential extracellular roles
that these inhibitors collectively contribute to the of nonadenosine-based purine nucleosides or
regenerative failure of injured CNS axons invivo. nucleotides. However, there is some evidence
At the other end of the spectrum are mecha- that these purines, particularly guanosine, may
nisms that promote neuronal outgrowth and also act as neurotransmitters or neuromodula-
regeneration after CNS injury. These include tors in the CNS. Our lab, together with others,
374 Part III:Homeostatic Manipulators

has shown evidence that guanosine may play an of neurotrophic factors as they can play dual roles
important role in CNS signaling and may exert and can elicit opposing signals in different dis-
neuroprotective and neurorestorative effects in eases (Hefti etal., 2006; Mantyh etal.,2011).
response to physiological and pathological con- One of the central themes for neurorestora-
ditions (Rathbone etal., 1999; 2008), discussed in tion after CNS disorder is to understand which
detaillater. factors are required to stimulate recovery in
In summary, after a CNS disorder, there are surviving nerves so that the nervous system
both inhibitory and stimulating environmental can rewire, leading to recovery of function.
factors at work; unfortunately, the stimulating In addition to disease-specific allopathic treat-
effects are overwhelmed by the inhibitory effects. ment, global holistic approaches that focus on
Reducing the nonpermissive environmental cues maintaining, regulating, and restoring CNS
while stimulating a more permissive environ- homeostasis will offer a long-lasting and system-
ment is the primary goal of neurorestoration. atic improvement. Many neurotrophic factors
share common signal transduction pathways
S T R AT E G I E S that influence numerous aspects of neuronal
Neurorestoration is interpreted differently by function and survival. The convergent factors in
various professions. For example, a surgeon may these signaling transduction cascades provide
employ a surgical intervention to effect neurores- new target sites for the development of novel
toration. Rehabilitation, physical and occupa- agents that could be used to treat a variety of
tional therapists employ exercise, acupuncture, or CNS disorders.
noninvasive brain stimulation in the treatment of In this chapter we focus on several neuro-
many CNS disorders (Demirtas-Tatlidede et al., trophic factors and small molecules including
2013; Schroer and Adamson, 2011). Exercise guanosine, especially on their ability to stimulate
and acupuncture are discussed in c hapter20 in endogenous neurogenesis. The common signal-
thisbook. ing pathways are also examined to explore the
Cell-based replacement therapy is one of mechanisms behind their therapeutic effects.
the newest and most discussed neurorestora- Our intention is not to focus in detail on any one
tive strategies both in the literature and in regulatory factor but to examine from a global
clinical studies for CNS disorder treatment. perspective how the CNS integrates the regu-
Stem cell related transplant is the prevailing latory and restorative processes of the body to
approach: embryonic stem cell studies have foster health and cope with environmental chal-
gradually been replaced by research employ- lenges, highlighting what our group has added to
ing adult stem cells such as neural stem cells, the current knowledge of neurorestoration.
induced pluripotent stem cells, and mesenchy-
mal stromal stem cells (Dutta etal., 2013; Farin NEUROTROPHIC EFFECT
etal., 2009). In addition to stem cell transplanta- T O S T I M U L AT E P E R M I S S I V E
tion, Schwann cells (Duncan etal., 1981; Li and ENVIRONMENT
Lepski, 2013; Wiliams and Bunge, 2012), olfac- For many years, NGF was the only known neu-
tory ensheathing cells (Bretzner etal., 2010; Xu rotrophic factor. Recently, however, the list of
etal., 1995), and enteric glia (Jiang etal., 2003b, the substances that may be classified as potential
2003c) have also been extensively studied to neurotrophic factors has expanded considerably.
determine their potential benefits. Purinergic molecules like adenine-based purines
In non-cell-based strategies, a large body of (adenosine and ATP) and nonadenine-based
molecules stimulate the permissive microenvi- purines (guanosine) have garnered wide interest
ronment through neurotrophic effects, including due to their neurotrophic effects (Burnstock, 2013;
neurotrophic factors (including but not limited Rathbone etal., 1999). Some hormones and neu-
to NGF, BDNF, and FGF2), purinergic molecules, rotransmitters such as insulin, thyroid hormones,
and so on; other strategies aim at reducing inhi- and sex steroids have been shown to affect neurite
bition, either by blocking the existing inhibitors extension and survival and can also be categorized
or modulating receptors, using antibodies and/ as neurotrophic factors (Arnold and Gorski, 1984;
or receptor antagonists, or reducing apopto- Recio-Pinto etal., 1986). In addition, there is evi-
sis, inflammation, or immunity effects. Among dence that interleukins and other lymphokines
these molecules, neurotrophic factors have been (IL-1beta, IL-2, IL-6) may also function as growth
shown to play fundamental roles in neuroresto- factors for certain neuronal and glia-like cells in
ration. Caution should be taken in the utilization the brain (Rothwell and Strijbos,1995).
Neurotrophic Regulation in Neurorestoration of theBrain 375

NGF on nigrostriatal dopaminergic neurons and may


In the neurotrophin family, NGF was the first lead to better functional restoration (Chaturvedi
identified and is the best characterized mem- etal., 2006; Li etal., 2000; Olson etal., 1991). In
ber. Studies have shown that NGF is distributed Huntingtons disease, mitochondrial dysfunc-
in high levels in regions of the CNS innervated tion plays an important role, and one study has
by the magnocellular cholinergic neurons of shown that NGF signaling can activate mito-
the basal forebrain (hippocampus, olfactory chondrial regulator PGC-1 expression, improve
bulb, and neocortex) and in regions containing behaviour, and restore sensorimotor ability and
cell bodies of these neurons (septum, nucleus reduce neuronal loss (Chen etal.,2012).
of the diagonal band of Broca, nucleus basalis Studies have shown that NGF-dependent
of Meynert). The highest level of NGF mRNA functional improvement is due in part to
expression occurs within the hippocampus and increased neurogenesis. In a dementia rat model,
cortex (Connor and Dragunow, 1998; Goedert NGF, which was at least partially mediated
etal.,1986). through increased cholinergic tone, promoted
There has been widespread interest in NGF neurogenesis in the adult hippocampus, which
as a neurotrophic factor in the CNS since its dis- may be due to the nootropic action of NGF
covery. In advanced AD, cholinergic basal fore- (Frielingsdorf etal., 2007). In cerebral ischemia,
brain neurons, which provide the major source NGF administration improved functional recov-
of cholinergic innervation to the cerebral cor- ery by stimulating neurogenesis in the ipsilat-
tex and hippocampus, undergo selective and eral striatum and subventricular zones (SVZ;
severe degeneration. With evidence from both Tonchev, 2011; Zhu et al., 2011). In an in vitro
tissue and animal studies, and given the lack of model, the extracellular signals of NGF and bFGF
an effective treatment for AD, the application of induced cholinergic neuron differentiation from
NGF in AD patients seems justified (Everall and endometrial stem cells (Noureddini etal.,2012).
Kerwin, 1990; Harbaugh, 1989; Mobley, 1989). In Unfortunately, since NGF cannot cross the
1993 the first case of intracranial infusion of NGF blood-brain barrier, it has to be administered
to an Alzheimers patient was reported. After one directly into the brain, either intraventricularly
month of NGF infusion, tests of verbal episodic by surgery or biologically through gene ther-
memory were improved, but other cognitive tests apy. Intraventricular delivery is usually asso-
were not. No adverse effects of the NGF infu- ciated with negative side effects such as pain.
sion were found. The results of this single case Gene therapy, however, is promising. Three
indicated that NGF may counteract cholinergic recent Phase I clinical trials for AD are explor-
deficits in AD and suggested that further clini- ing this prospect. In the first study, human NGF
cal trials of NGF infusion in AD were warranted genetically engineered into autologous grafted
(Olson etal., 1992; Seiger etal., 1993). However, in fibroblasts was delivered into eight individuals
a three-patient case study, researchers concluded with mild AD. The results suggested diminu-
that while long-term intracerebro-ventricular ation in the rate of cognitive decline, and serial
NGF administration may cause certain poten- PET scans showed significant increases in cor-
tially beneficial effects, the intraventricular route tical 18-fluorodeoxyglucose after treatment
of administration is also associated with nega- (Tuszynski et al., 2005). Another study used an
tive side effects, including constant back pain adeno-associated virusbased gene delivery tech-
and rapid weight loss, and appear to outweigh nique to administer NGF to the brain to treat AD
the positive effects of the protocol. Alternative symptoms and progression (Mandel, 2010). This
routes of administration, and/or lower doses of study has passed Phase I clinical safety testing,
NGF, perhaps combined with low doses of other and a multicenter Phase II clinical trial is cur-
neurotrophic factors, may shift the balance more rently ongoing. Another recent Phase I clinical
positively (Eriksdotter-Jonhagen etal.,1998). trial using encapsulated cell biodelivery of NGF
In addition to extensive study in AD, NGF to the basal forebrain of AD patients showed pos-
has also been examined in other CNS disorders. itive neurological outcomes in two of six patients,
After focal ischemia or stroke, numerous studies and no NGF-related adverse events were found
have demonstrated that NGF, alone or combined (Eriksdotter-Jonhagen etal.,2012).
with other treatments, can improve functional Caution should be taken in the utilization of
recovery (Cheng et al., 2009; Luk et al., 2004; NGF in different diseases. While NGF plays a sig-
Zhu etal., 2011). In a PD model, NGF exhibited a nificant role in the survival of sensory and sympa-
potential neuroprotective and neurorescue effect thetic neurons after injury and inflammation, it is
376 Part III:Homeostatic Manipulators

also a major factor in pain promotion and hyper- central pain sensitization (Croll etal., 1999; Pezet
algesia (Hefti et al., 2006; Mantyh et al., 2011). etal., 2002). In epilepsy, however, studies showed
By binding to its receptor TrkA on nociceptors, that BDNF mRNA and protein are markedly
several different pathways are activated, including upregulated in the hippocampus by seizure activ-
ion channels and the transient receptor potential ity in both animal models and a patient sample
vanilloid receptor, and secondary neurotransmit- (Kokaia etal., 1995; Takahashi etal., 1999). Also,
ters, including substance P and BDNF (Cattaneo, overexpression of BDNF in transgenic mice led to
2010; Hefti etal., 2006; Mantyh etal., 2011), which spontaneous seizures (Croll et al., 1999). BDNF
cause immediate and long-term excitability. circulates systemically outside the CNS via the
Based on this information, in an example such as cardiovascular system. Golden et al. (2010)
back pain, it might be more beneficial to employ showed that elevated levels of circulating BDNF
NGF neutralizing antibodies or antagonists that in middle-aged and elderly adults increased the
have been designed to fight different kinds of pain risk of high blood pressure and cardiometabolic
rather than NGF itself (Cattaneo, 2010; Hefti etal., dysfunction. BDNF preferentially binds to p75
2006; Kivitz etal., 2013; Mantyh etal.,2011). neurotrophin receptor (p75NTR) rather than
TrkB on sympathetic neurons. Recent studies
BDNF show that selective activation of the p75NTR
BDNF was the second neurotrophic factor to be in sympathetic neurons causes axon degenera-
discovered. It is a protein with a molecular mass tion. Therefore, elevated BDNF in the left ven-
of 27 kD that was originally purified from pig tricle after ischemia-reperfusion may stimulate
brain and was shown to support the survival of, p75NTR-dependent denervation of peri-infarct
and fiber outgrowth from, cultured embryonic myocardium (Lorentz,2013).
chick sensory neurons in 1982 (Barde etal.,1982). Since the purification of BDNF in 1982,
BDNF has been observed to be widely distrib- extensive literature has been published regard-
uted within the CNS, including the hippocam- ing its central role in brain development, physi-
pus, amygdala, thalamus, projection areas of ology, and pathology. BDNF was demonstrated
the olfactory system, inner and outer pyramidal to have survival and growth-promoting actions
layers of the neocortex, claustrum, septum, cer- on both central and peripheral neurons (Acheson
ebellum, and superior colliculus (Connor and etal., 1995). BDNF exerts its growth-promoting
Dragunow, 1998; Gall etal., 1992; Phillips etal., effect mostly by encouraging synapse growth
1990). In neurodegenerative diseases, many and facilitating synaptic plasticity, stimulat-
studies have demonstrated reduced levels of ing synaptogenesis. Studies have shown that, in
BDNF (Murer et al., 2001). For example, BDNF postnatal rat hippocampal slice cultures, BDNF
mRNA expression was found to be reduced in increased spine density in apical dendrites of
AD hippocampus specimens (Murray et al., CA1 pyramidal neurons (Tyler and Pozzo-Miller,
1994; Phillips et al., 1991). A significant reduc- 2003) and enhanced the expression of synaptic
tion in BDNF protein within the hippocampus proteins (Tartaglia etal., 2001). Transgenic mice
and temporal cortex of the postmortem human overexpressing BDNF had an increased number
AD brain was also found in a later study (Connor of synapses (63%) and increased synaptic vesi-
et al., 1997). Decreased BDNF protein has also cle docking in area CA1 (Aguado et al., 2003).
been demonstrated in the substantia nigra in A blockade of BDNFTrkB signaling prevented
PD (Howells et al., 2000). Furthermore, stud- spine head enlargement, whereas synaptic stim-
ies have shown that in Huntingtons disease, ulation plus the addition of exogenous BDNF
loss of huntingtin-mediated BDNF transcrip- induced spine enlargement in the absence of
tion leads to loss of trophic support to striatal postsynaptic spikes (Tanaka etal.,2008).
neurons, which subsequently degenerate in the The importance of BDNF in regulating synap-
hallmark pathology of the disorder (Binder and tic transmission, plasticity, and growth suggests
Scharfman, 2004; Zuccato etal., 2001). In anxi- that it has a crucial role in cognitive function.
ety disorders, some studies showed reduced Indeed, numerous studies have demonstrated
BDNF levels in individuals; however, this was not that a reduction in BDNF level not only impaired
consistent across the various anxiety disorders long-term potentiation (LTP) and reduced the
(Suliman etal., 2013). In CNS pain, electrophysi- number of synapses but was associated with a
ological and behavioral data demonstrate that deficit in learning and memory (Linnarsson
inhibition of BDNF signal transduction inhibits etal., 1997; Mu etal.,1999).
Neurotrophic Regulation in Neurorestoration of theBrain 377

We now understand that the trophic prop- drug, other strategies to manipulate BDNFTrkB
erties of BDNF may lead to novel therapeutic signaling have also shown potential benefits.
options in traumatic brain injury and stroke, in Such strategies include small-molecule TrkB
neurodegenerative diseases such as PD and AD, agonists or modulators and TrkB agonistic anti-
and perhaps even in neuropsychiatric disorders bodies (Lin etal., 2008; Massa etal., 2010; Qian
(e.g., depression). In contrast, pathological levels et al., 2006). An even more promising strategy
of BDNF-dependent synaptic plasticity may con- is to pharmacologically enhance the expression
tribute to conditions such as epilepsy and chronic of endogenous BDNF in the brain using a small
pain sensitization (Binder and Scharfman,2004). molecule approach.
Preclinical studies have shown promising
results for BDNF as a synaptic repair molecule in N E U R O T R O P H I N ( N G F/ B D N F )
neurodegenerative diseases. For example, intra- S I G N A L I N G PAT H WAY
ventricular infusion of BDNF or adeno-associated There are dozens of neurotrophic factors, and by
virusinduced BDNF activity increases the examining their nucleotide sequences and, there-
number of neurons in the adult olfactory bulb, fore, evolutionary relatedness, they can be sepa-
striatum, septum, and thalamus (Benraiss etal., rated into different families. The neurotrophin
2001; Pencea et al., 2001; Zigova et al., 1998). family, including NGF, BDNF, NT-3, and NT-4,
Furthermore, in a neurotoxin 6-OHDA (oxido- is perhaps the best understood and most widely
pamine) PD model, BDNF attenuated the loss of expressed in the brain. Their intracellular signal-
dopaminergic axons and prevented rotational ing pathways have also been extensively studied.
asymmetry (Shults etal., 1995; Yoshimoto etal., Neurotrophins regulate neuronal survival
1995). Moreover, BDNF has been shown to pro- and other aspects of cellular and biological
tect and/or repair hippocampal neurons and functions through the activation of correspond-
synapses despite beta amyloid (A) build-up ing receptors. There are two distinctive classes
and neuronal toxicity in a mouse model of AD of neurotrophin receptors: p75NTR and the
(Nagahara and Tuszynski,2011). tyrosine kinases (Trk) receptor. Trk-mediated
Even though the beneficial effects of BDNF on signaling promotes survival and growth, while
neuronal functions have been discussed exten- p75NTR-mediated signaling regulates celldeath.
sively in the literature, few clinical trials using
BDNF have been conducted (four in ALS and T R K S I G N A L I N G PAT H WAY
one in diabetic neuropathy; Lu etal., 2013), and The Trk receptors, a closely related family of
the results have been inconclusive. In a Phase I/II receptor tyrosine kinases, recognize neuro-
open-label trial for ALS, subcutaneously admin- trophins with a relatively high degree of bind-
istered BDNF showed a delay in the percentage ing specificity. Biological effects of each of the
of forced vital capacity decline and an improve- four mammalian neurotrophins are mediated
ment in walking time, but a Phase II/III trial did through activation of one or more of the three
not replicate these benefits (Liu et al., 2012). Lu members of the Trk receptor: TrkA, TrkB, and
etal. (2013) postulated that the clinical failure of TrkC. TrkA preferentially binds NGF, TrkB pre-
BDNF may be due to the fact that BDNF is cleared fers BDNF and NT-4/5, and TrkC interacts only
rapidly in vivo from where it was administered with NT-3. In addition, NT-3 can bind with the
and does not easily penetrate into the spinal cord other Trk receptors with less affinity (Chao, 2003;
parenchyma. Reichardt, 2006). Upon neurotrophin-binding
Currently, several approaches are actively activation, the Trk receptor becomes autophos-
being explored to deliver BDNF into the CNS. phorylated, and the autophosphorylation sites
Invasive procedures include using a catheter form docking sites for the interaction of down-
or implantable pumps (Dittrich et al., 1996) or stream signaling molecules. These molecules
implantation of a biodegradable polyethyl- trigger distinct downstream cascades of target
ene glycol-based hydrogel device containing enzymes and other biological effects, includ-
polylactic-co-glycolic acid microparticles encap- ing neuronal differentiation and survival
sulated with BDNF (Bertram etal., 2010; Lampe (Chao, 2003; Huang and Reichardt, 2001). Three
et al., 2011). Noninvasive approaches such as well-conserved downstream signaling pathways
nanoparticle molecular carriers of BDNF are also have been characterized, including Ras/extracel-
being explored (Gabathuler, 2010; Gral et al., lular signal-regulated kinase (ERK)which is
2013). In addition to BDNF itself as a potential now known as mitogen-activated protein kinase
378 Part III:Homeostatic Manipulators

(MAPK), phosphatidylinositol 3-kinase (PI3K) channel gene in PC12 cells is mediated through
and phospholipase C gamma (PLC-gamma) the PLC-gamma pathway (Choi etal.,2001).
pathways (Chao, 2003; Reichardt,2006).
The MAPK pathway is regulated by the activ- P 7 5 N T R S I G N A L I N G PAT H WAY
ity of the Ras protein, which serves as a transducer P75NTR is a member of the tumor necrosis
of signal from Trk to ERK proteins (Denhardt, factor (TNF) receptor superfamily. It consists
1996). The typical process starts from the activa- of an extracellular domain that includes four
tion of Ras by guanosine-5-triphosphate (GTP) cysteine-rich motifs, a single transmembrane
binding; following that, Ras recruits a serine domain, and a cytoplasmic domain that includes
kinase of the Raf family to the membrane, where a death domain similar to those present in
it is activated. This initiates another cascade in other members of the TNF family. Originally,
which Raf activates MEK, and then MEK phos- p75NTR was identified as a low-affinity receptor
phorylates and activates ERK. ERK is abundant for NGF, but it was subsequently shown to bind to
and multifunctional, regulating many different each of the neurotrophins (BDNF, NT-3, NT-4/5)
cellular activities through transcription factors with a similar affinity (Frade and Barde, 1998;
such as c-myc, CREB, and c-Fos (Cobb, 1999; Hempstead, 2006; Johnson etal.,1986).
Davis, 1993). By altering transcription factor P75NTR is a key signaling component for
levels and activity, MAPK regulates cell cycle, multiple ligands that regulate a wide array of
proliferation, and differentiation. Studies have biological responses, some of which are opposite
shown that MAPKs can mediate neuron survival (e.g., cell survival vs. programmed cell death),
and neurite outgrowth (Fukuda etal., 1995)and depending on the cellular and pathological
can regulate activity-dependent neuronal events context.
such as synaptic plasticity and LTP (English and To date, three signaling pathways have
Sweatt, 1997; Grewal etal.,1999). been elucidated following neurotrophin bind-
PI3K can be activated by three different ing to p75NTR. One major pathway is the Jun
mechanisms, either directly by phosphoryl- N-terminal kinase (JNK), also known as the
ated receptors, through docking proteins such stress-activated protein kinase signaling cas-
as the insulin receptor substrate family of pro- cade. Signaling through this cascade results in
teins (Myers and White, 1996), or by Ras protein activation of p53 and apoptosis, causing more
(Kodaki et al., 1994; Rodriguez-Viciana et al., neuronal cell death following injury (Coulson
1994). Upon activation, PI3K further stimulates etal., 2000; Reichardt, 2006). The crucial role of
the formation of second messenger phosphati- JNK pathway-mediated apoptosis in the p75NTR
dylinositol (3,4,5)-trisphosphate, which activates signaling pathway has been demonstrated in
the downstream effector Akt (also known as pro- many studies (Chen et al., 2009; Deshmukh
tein kinase B, which is a serine/threonine-specific et al., 1996). Effectors reported as involved in
protein kinase). Akt is best known for the JNK pathway-induced apoptosis include CDC42,
anti-apoptotic effects it generates by render- apoptosis signal regulated kinase 1, Rac guano-
ing downstream apoptogenic factors inactive in sine triphosphatase, P53, Bax, and Caspase (Chen
many cell systems (Kennedy etal., 1997; Zhuang et al., 2009; Harrington et al., 2002; Reichardt,
etal., 2011). In neurons, PI3K has been shown to 2006; Troy etal., 2002). Increased expression of
mediate cell survival, induce glutamate excito- p75NTR is a response to many forms of injury,
toxicity/sensitivity, and initiate neurite process including neural axotomy, mechanical damage,
outgrowth (Fryer etal.,2000). stroke ischemia, seizure, and so on. Several stud-
The Trk receptor can also activate ies have demonstrated that p75NTR is responsi-
PLC-gamma, which will result in the activation ble for cell death after injury in vivo. While the
of Ca2+ and protein kinaseregulated path- mechanism of how p75 induces apoptosis is not
ways and promote synaptic plasticity. Release completely understood, some studies have shown
of intracellular stores of calcium can have a link from proneurotrophin to p75-induced
many effects, including activation of Ca2+/ apoptosis. Following injury or seizure, neuro-
calmodulin-dependent protein kinases and an trophins were released in uncleaved precursor
increase in the production of cAMP; both have format-proneurotrophin. Antibodies specific to
powerful effects on neurons. Studies show that proneurotrophins prevented cell death accord-
after transient exposure to NGF, the induc- ingly (Harrington et al., 2004; Volosin et al.,
tion of the peripheral nerve type 1 sodium 2008). Further studies are needed to elucidate
Neurotrophic Regulation in Neurorestoration of theBrain 379

the pathways regulating proteolytic processing of Crosstalk occurs inside the p75NTR pathway.
proneurotrophin in the uncleaved versus mature For example, while many studies have shown that
form. In addition to regulating apoptosis and cell p75NTR/NF-B can enhance neuronal survival
death after injury, p75NTR has also been shown (Chen etal., 2009; Foehr etal., 2000; Hamanoue
to contribute to neurodegeneration. Studies have etal., 1999), research has also shown that the acti-
demonstrated that there is a high level of p75NTR vation of the p75NTR is the underlying mecha-
expression in the cholinergic neurons of the adult nism of neuronal damage by A peptides in AD.
basal forebrain, which is severely affected in AD. The p75NTR further activates p38 and JNK in a
Some studies indicated that beta amyloid (142), death domain-dependent manner, followed by
the major component of the hallmark plaque NF-B translocation, p53 activation, and apopto-
found in AD patients, is a proapoptotic ligand sis (Costantini etal.,2005).
for p75NTR and triggers p75-mediated cell death Even though both the p75NTR and TrkA
(Costantini et al., 2005). In addition, pro-NGF receptors lead to the activation of NF-B, they pro-
was also found to contribute to AD pathology ceed via distinctly different proximal-signaling
via the p75NTR pathway (Pedraza et al., 2005; intermediates and contribute to different cellu-
Podlesniy etal.,2006). lar outcomes. Blocking p75-mediated activation
Even though p75NTR mediates cell death of NF-B in PC12 cells significantly enhanced
and neurodegeneration, it also can promote cell apoptosis. In contrast, blocking NF-B induction
survival through different pathways. Nuclear via TrkA significantly inhibited the neurite for-
factor-kappa B (NF-B) cascade is the major mation process (Foehr etal.,2000).
signaling pathway induced by p75NTR activation
and contributes to neuronal survival (Hamanoue
etal., 1999; Middleton etal., 2000). In response FGF2
to ligand binding, p75NTR binds TNF receptor Fibroblast growth factor (FGF) is a superfam-
associated factor-6 proteins through the intra- ily with 23 members and 4 receptors identified
cellular domains and activates a kinase cascade to date. In the mid-1980s, FGF2 (also known as
that culminates in activation of IKK and IKK basic FGF) was the first FGF family member to
and subsequent phosphorylation of IB subunits, be isolated and cloned (Abraham et al., 1986a,
which targets them for ubiquitination and pro- 1986b). FGF2 is the most abundant member of
teosomal degradation. IB degradation releases FGF in the CNS and is widely expressed dur-
NF-B, which is a ubiquitously expressed tran- ing development and postnatally, to both neu-
scription factor. The released NF-B translocate rons and glial cells (Eckenstein et al., 1991;
to the nucleus where they stimulate specific gene Emoto et al., 1989). FGF2 has proven to be the
expression and regulate cell survival. most functionally versatile growth factor in the
A third pathway through the Rho family of CNS (Reuss and von Bohlen und Halbach, 2003;
small guanosine triphosphatases has been iden- Zechel etal., 2010)and is fundamentally involved
tified, which activates RhoA, thereby inhibiting in the development of the CNS, the promotion of
neurite outgrowth (Chen etal., 2009; Reichardt, axonal growth, the maintenance of plasticity in
2006; Yamashita et al., 1999). RhoA plays an adulthood, angiogenesis, and, most important,
important role in transducing extracellular sig- the stimulation of adult neurogenesis.
nals to changes in cytoskeletal proteins and Evidence has shown that FGF2 can modu-
causes growth cone repulsion and collapse (Wu late synaptic plasticity and axonal branching in
etal.,2005). vitro and in vivo. Early studies demonstrated that
Binding of each of the neurotrophins to FGF2 is capable of promoting neurite extension
p75NTR also evokes activation of cellular sphin- in PC12 cells (Rydel and Greene, 1987; Togari
gomyelinase, which results in increased ceramide et al., 1985) and rat cerebral cortical neurons
production (Dobrowsky etal., 1994). Ceramide is (Morrison et al., 1986) and axonal branching
a versatile molecule that can promote both apop- in hippocampal neurons (Aoyagi et al., 1994).
totic and prosurvival mechanisms by activating Application of FGF2 enhanced LTP in rat hip-
many different signaling pathways, including pocampal slices (Terlau and Seifert, 1990).
ERK, PI3K, JNK, and NF-B, as well as TrkA Intracerebroventricular injection of epidermal
(DeFreitas et al., 2001; MacPhee and Barker, growth factor and FGF2 promoted the genera-
1997; Muller et al., 1998; Song and Posse de tion of LTP in synapses in the dentate gyrus of
Chaves,2003). anaesthetized rats (Ishiyama etal.,1991).
380 Part III:Homeostatic Manipulators

In addition, studies have also documented through the surface glycoprotein CD200, which
the angiogenic properties of FGF2 in embryonic regulates microglial activation (Woodbury and
development (Leconte etal., 1998), as well as in Ikezu, 2013). Manipulation of FGF2/FGFR1
the adult myocardium after ischemia (Iwai-Kanai signaling has been a focus of therapeutic devel-
etal., 2002; Unger etal., 2000; Yanagisawa-Miwa opment for many neurodegenerative disorders,
et al., 1992). FGF2 and vascular endothelial such as AD, multiple sclerosis (Rottlaender etal.,
growth factor play synergistic roles in angio- 2011; Ruffini etal., 2001), PD (Baron etal., 2012),
genesis. They are released into the wound site and traumatic brain injury (Sun etal.,2009).
during haemostasis and promote the formation The intracellular signaling pathway of FGF2/
of new blood vessels, thus stimulating angiogen- FGFR1 activation has not been fully elucidated.
esis (Przybylski, 2009). In a brain injury model From available studies, we know that both
of chronic hypoxic-ischemia, the enhanced phosphotyrosine-dependent and -independent
endogenous angiogenesis and neurobehavioral mechanisms are involved, and common path-
functions were demonstrated to be mediated by ways such as MAPK, PI3K, and PLC-gamma,
upregulation of FGF2 (Seo etal.,2013). which regulate proliferation, differentiation, cell
Numerous studies have confirmed the impor- survival, and apoptosis, are also triggered under
tant role FGF2 plays in adult neurogenesis. In the different conditions.
adult CNS, FGF2 is expressed in the SVZ and the The most common pathway employed by
subgranular zone of the hippocampal dentate FGF2 is the MAPK pathway. The upstream
gyrus (Ernfors et al., 1990; Zheng et al., 2004). components of the signaling pathway, includ-
FGF2 has been extensively studied and found to ing fibroblast growth factor receptor substrate
be a potent modulator of proliferation and dif- 2 (FRS2) and the Src homology 2 domain, con-
ferentiation for multipotent neural progenitors. tain transforming protein (Shc). FGFR1 can
The ability of FGF2 to induce proliferation of phosphorylate FRS2 and Shc. The phosphoryl-
adult stem cells has been clearly demonstrated ated FRS2 continues to bind the adapter protein
in in vitro studies using adult mouse striatum growth factor receptor bound 2 (GRB2) and the
progenitors (Gritti etal., 1996)and hippocampal protein tyrosine phosphatase, nonreceptor type
progenitors (Gage etal., 1995; Ray etal., 1993)by 11. Shc and GRB2 form a complex with the gua-
maintaining these progenitors in the cell cycle nine nucleotide exchange factor Son of sevenless
and preventing them from further differentia- proteins (SOS). Further on, SOS activate the Ras/
tion. In vivo studies have also provided evidence Raf/MEK1/2/ERK1/2 signaling cascade, as dis-
of FGF2-induced neuronal precursor cell prolif- cussed in the TrK section. This cascade leads to
eration in the neurogenic regions of the adult rat phosphorylation of the target transcription fac-
brain (Kilpatrick and Bartlett, 1993; Rai et al., tor ELK1 (Katoh, 2006; Suyama etal., 2002; Yang
2007; Wagner etal.,1999). etal.,2008).
The FRS2/GRB2 complex might also trigger
F G F 2 S I G N A L I N G PAT H WAY the PI3K pathway through a connection with
FGFs elicit their diverse effects through acti- GRB2-associated binding protein 1 (GAB1). The
vation of cell surfacebound high affinity Trk assembly of the FRS2/GRB2/GAB1 complex leads
receptors FGFR1-4 (Friesel and Maciag, 1999; to activation of the PI3K pathway and down-
Jaye etal., 1992). The binding of FGFs to FGFRs stream effector proteins such as Akt (Hadari
is mediated in part by an additional interaction et al., 2001; Ong et al., 2001). FGF2 is a known
with heparin sulfate proteoglycans. This inter- inducer of epithelial-to-mesenchymal transition
action results in dimerization of the receptors, (EMT). FGF2 induces EMT via PI3K (Ko and
resulting in autophosphorylation and transpho- Kay, 2005; Lee and Kay, 2003; Lee and Kay, 2006).
sphorylation of tyrosine residues, which further FGF2/FGFR1 also can trigger the PLC-
activates the intracellular signaling pathways. gamma signaling pathway in the process of
Among FGF receptors, FGFR1 is widely hydrolyzing membrane phospholipids in cells.
expressed but is nevertheless confined to specific Upon activation, PLC-gamma in turn hydrolyzes
neuronal populations in the adult CNS (Asai phosphatidylinositol 4,5 bisphosphate to diacyl-
et al., 1993; Yazaki et al., 1994). FGF2/FGFR1 glycerol (DAG) and inositol trisphosphate (IP3).
signaling has been shown to regulate adult neu- DAG and IP3 are second messengers. DAG acti-
rogenesis in the SVZ of the adult rat (Mudo etal., vates protein kinase C delta; IP3 activates the IP3
2007). It also modulates inflammatory signaling receptor and induces the release of Ca(2+) from
Neurotrophic Regulation in Neurorestoration of theBrain 381

intracellular Ca(2+) storage into the cytoplasm and in vivo and can help functional recovery
(Kim etal.,2003). in different CNS disorders such as hypoxia-
ischemia (Moretto et al., 2009; Thauerer et al.,
NONADENINE-BASED PURINES 2012), stroke (Chang et al., 2008; Connell
Since neurotrophins such as NGF and BDNF et al., 2013; Rathbone et al., 2011), spinal cord
cannot cross the blood-brain barrier, their thera- injury (Jiang etal., 2003a; 2007, 2008a), seizure
peutic potential is limited. However, other strat- (Schmidt etal., 2000; 2005; Soares etal., 2004),
egies that are being pursued include developing and PD (Giuliani etal., 2012; Su etal.,2009).
drugs that mimic the action of growth factors on The purinergic system is emerging as a key
receptors, stimulating growth factor synthesis, player in the endogenous control of neural stem
and enhancing the effects of endogenous growth cells. Ulrich et al. (2012) discussed the impor-
factors. tance of adenine-based purinergic signaling
Our lab made the initial discovery of the molecules (ATP, uridine-5'-triphosphate, and
effects of extracellular guanosine on cells (Kim adenosine) in regulating the synchronized pro-
et al., 1991). Over the past two decades, we liferation, migration, differentiation, and death
have focused on the study of nonadenine-based of neural stem cells during brain and spinal
purines, especially guanosine. One of our studies cord development. In our lab, we discovered
showed that guanosine can cross the blood-brain that systemic administration of guanosine can
barrier. Exogenous guanosine (8 mg/kg body stimulate the proliferation of endogenous pro-
weight) entered the rat CNS within 7.5 minutes genitor/stem cells. In a traumatic spinal cord
of intraperitoneal administration and resulted injury rat model, we found that daily intraperito-
in a doubling of the guanosine concentration in neal administration of guanosine for two weeks
the brain (Jiang etal., 2008b). This unique char- enhanced functional improvement in correlation
acteristic of guanosine makes it advantageous in with an increase in myelination in the injured
the potential future application of CNS disorder cord. The functional improvement and remyeli-
treatment. nation observed after systemic administration
We discovered (Kim et al., 1991; Rathbone of guanosine is due to the effect of guanosine/
et al., 1992; 1999) and many others confirmed guanine on the proliferation of adult progenitor
(Bocklinger et al., 2004; Moretto et al., 2009; cells and their maturation into myelin-forming
Thauerer et al., 2012) that exogenous guano- cells (Jiang et al., 2008a). Furthermore, in a rat
sine exerts numerous neurotrophic effects. One model of Parkinsonism, we showed that guano-
mechanism of action is by stimulating synthesis sine treatment increased cellular proliferation in
and release of purines and trophic factors, such as the substantia nigra and SVZ, and ameliorated
NGF. In our 1995 study, we found that cultured symptoms. Proliferating cells in the SVZ were
neonatal mouse cortical astrocytes synthesized noted in nestin-positive adult neural progenitor/
NGF mRNA and released immunoreactive NGF stem cells. This finding indicated that guanosine
(ir-NGF) into the culture medium. The addition protected cells from apoptosis and stimulated
of 10 M guanosine to the cultures increased intrinsic adult progenitor/stem cells to become
ir-NGF release by six-fold after 24 hours and dopaminergic neurons in rats with proteasome
increased NGF mRNA six-fold after four hours inhibitor-induced PD (Su etal.,2009).
(Middlemiss et al., 1995b). Subsequent studies Guanosine treatment has some advantages
showed that extracellular guanosine promotes over autologous cell transplant and neuronal
neurite outgrowth in PC12 cells and synergis- growth factor techniques. First, as a naturally
tically enhances NGF-dependent neurite out- occurring compound, it is nontoxic, and, unlike
growth (Gysbers and Rathbone, 1992, 1996a). with cell transplantation, there is no immune
The enhancement of the neuritogenic effects of reaction. Another unique characteristic of
NGF by GTP and guanosine may have physi- guanosine is that it can cross the blood-brain
ological implications in sprouting and functional barrier (Jiang et al., 2008b). This evidence sug-
recovery after neuronal injury in the CNS due gests that the purinergic system may be a target
to the high levels of nucleosides and nucleotides for new strategies to treat neurodevelopmental
released from dead or injured cells (Gysbers and disorders and neurodegenerative diseases.
Rathbone, 1996a). Our studies showed that other nonadenine-
Extensive literature has demonstrated that based purine derivatives can also stimulate NGF
guanosine has neurotrophic effects both in vitro production. Neotrofin, leteprinim potassium
382 Part III:Homeostatic Manipulators

(AIT-082) is a novel, metabolically stable deriva- Numerous studies have also found that the
tive of the purine hypoxanthine. Addition of neuroprotective effect of guanosine on ischemia
AIT-082 to cultured PC12 cells significantly involves augmentation of glutamate uptake,
enhanced NGF-mediated neurite outgrowth which is modulated by BK channels and the
from PC12 cells (Middlemiss et al., 1995a). activation of the PI3K pathway. Moreover, neu-
Exposure of mouse astrocytes to AIT-082 stimu- roprotection caused by guanosine depends on
lated messenger RNA production for NGF, NT-3, the increased expression of phospho-Akt pro-
and bFGF in cultured mouse astrocytes (Glasky tein (Dal-Cim et al., 2011; Schmidt et al., 2008;
et al., 1995). Some preliminary experimental Tavares etal.,2005).
evidence indicates that AIT-082 effects on neu- We also found that guanosine can stimu-
rite outgrowth observed in vitro can also occur late endogenous stem/progenitor cell prolifera-
in vivo. In the latter, AIT-082 increased NGF tion in a number of models (Jiang etal., 2008a;
mRNA synthesis in the region of injury when Su etal., 2009). To further elucidate the mecha-
administered orally to rats following basal fore- nisms underlying guanosine-triggered neuro-
brain lesions induced by stereotaxic injection genesis, we used an in vitro neural stem cell line
of ibotenic acid (Glasky et al., 1997). AIT-082 model and found that guanosine can stimulate
administration to rats resulted in a small but neural stem cell proliferation partially through
significant advance in the onset of nerve sprout- the cAMP-CREB pathway (Su et al., 2013).
ing into denervated adjacent skin. This sprout- Guanosine might also exert its neurogenesis
ing process is NGF-dependent (Rathbone etal., effect by stimulating endogenous FGF2 (Su etal.,
1999). However, unlike treatment with exog- 2009) or BDNF (unpublished data) accumula-
enous NGF, AIT-082 treatment did not cause tion, or by working synergistically on common
hyperalgesia. These data not only indicate that signaling cascades downstream.
AIT-082 can stimulate neurite outgrowth in vivo In an in vitro model employing cultured rat
as in in vitro but also that AIT-082 may be thera- astrocytes, we found that the antiapoptotic effect
peutically useful since, unlike NGF, it does not of guanosine is mediated by PI3-kinase/Akt/
cause hyperalgesia. protein kinase B pathway activation (Di Iorio
etal., 2004). Utilizing an in vitro model of PD,
GUANOSINE SH AR ES Giuliani etal. (2012) found that guanosine pro-
COMMON SIGNALING tected SH-SY5Y neuroblastoma cells when they
TR ANSDUCTION CASCADES were exposed to neurotoxin 6-hydroxydopamine
W I T H N G F, B D N F, A N D F G F 2 (6-OHDA) and promoted their survival by reg-
Even though the detailed intracellular ulating the p-38 and JNK pathways. Moreover,
mechanism(s) involved in the effects of guano- guanosine also potentiated an early increase
sine on neuroprotection, neurogenesis, and in the phosphorylation of the anti-apoptotic
functional recovery of CNS disorders remains kinase Akt and increased the expression of
to be fully elucidated, knowledge gained from the anti-apoptotic Bcl-2 protein induced by
previous studies allows us some insight into 6-OHDA.
guanosines working network (Figure22.1). Guanosine has also been shown to contribute
Guanosine-activated cAMP-dependent to lipid homeostasis in the CNS. Cholesterol plays
mechanisms are thought to activate the MAPK a key role in the regulation of synaptic physiology
cascade and potentially other protein kinases and neurotransmission as well as in those mech-
(Rathbone et al., 1991; Tomaselli et al., 2005). anisms underlying synaptic plasticity (Koudinov
Within the MAPK pathway, guanosine has been and Koudinova, 2001). Moreover, cell choles-
found to activate and stimulate phosphorylation terol depletion has been linked to a reduction in
of ERK-1 and -2 in cultured astrocytes that may amyloid beta formation. One study showed that
act to promote NGF synthesis and release (Di guanosine was able to increase cholesterol efflux
Iorio etal., 2001; Rathbone etal., 1998; Schwartz from astrocytes and C6 rat glioma cells through
and Mishler, 1990). The MAPK cascade has been PI3K/ERK-1/2 pathways (Ballerini etal.,2006).
identified as the major pathway by which NGF The diverse effects of extracellular guanosine
induces growth and differentiation in PC12 cells on many cellular components of the CNS may
(Rathbone et al., 1999) and may, therefore, rep- be related to its uptake into cells but may also be
resent a convergent mechanism for NGF and due to its ability to release adenine-based purines
guanosine. from cells; however, it has also been suggested
Neurotrophic Regulation in Neurorestoration of theBrain 383

FIGURE 22.1: Guanosine may mediate its biological effects through growth factor pathways,
directly or indirectly, by sharing signaling transduction cascades. This schematic represents the pos-
sible mechanisms by which guanosine might mediate its biological effects. Guanosine, a small nonadenine purine
compound, may act through GPCR(s), or unknown receptor(s), to regulate signaling molecules (activating the
molecules as indicated by green arrow [], or inhibiting the molecule as indicated by green arrow end []). The
involvement of these molecules indicates that guanosine shares common signaling cascades with growth factors,
including NGF, BDNF, and FGF2. The three major signaling pathways (PLC-gamma, PI3K, MAPK) emanating
from receptor tyrosine kinases (RTKs) are shown here. RTKs are widely expressed transmembrane proteins that
act as receptors for growth factors, neurotrophic factors, and other extracellular signaling molecules. The Trk
receptor family and the FGF receptor family are two classes of RTKs. Guanosine was shown to activate the PI3K/
Akt and MAPK pathways. In the PI3K/Akt pathway, Akt has an anti-apoptotic effect through its effects on Bad and
caspases. In the MAPK pathway, activated Ras activates a cascade of kinases including Raf, MEK, and ERK (also
known as MAP kinases). ERKs stimulate many known effectors. Each of these pathways then exerts a number of
nuclear and nonnuclear actions with short- and long-term consequences for the cells. It was also demonstrated
that guanosine can induce glia cells to produce growth factors, which will further exert the biological effects.

that its mechanism of action may occur through 2011, a study using aDELFIA Eu-GTP binding
a specific G-protein coupled receptor. In 2002, assay further showed that the existing guano-
Traversa etal. showed that there is specific guano- sine binding site is a G-protein-coupled receptor
sine binding site in the rat brain membrane. In (Volpini etal.,2011).
384 Part III:Homeostatic Manipulators

TA R G E T I N G T H E C O N V E R G E N T the MAPK pathway on the CNS, a motor per-


MOLECULES ONTHE COMMON formance test was carried out in drosophila. The
SIGNALING TR ANSDUCTION use of small interfering RNA (siRNA) to decrease
CASCADE the MEK, ERK-1/2, and MSK1 drosophila homo-
As discussed previously, after CNS disorders, a logues resulted in increased motor performance
plethora of supportive (e.g., neurotrophic fac- and lifespan. Decreasing upstream MAPK path-
tors) and hostile molecules (such as semaphorins, way homologues suppressed ATXN1 eye defects
ephrins, myelin-secreted inhibitory glycopro- and improved motor and lifespan phenotypes. It
teins, and repulsive axon guidance molecules) was also found that ATXN1 levels were sensitive
are generated in the microenvironment (Chiquet, to S776 phosphorylation. Further study revealed
1989; Fan etal., 2013; Filbin, 1995). Like the sup- that the MAPK kinases were implicated and that
porting molecules, the nonpermissive inhibitory MSK1 was able to phosphorylate the consensus
cues also act through their respective receptors sequence associated with S776. Based on these
to affect the collapsing of growth cones via sev- previous findings, in 2013 Park etal. designed a
eral pathways (Hou et al., 2008). Many of these proof-of-principle strategy to determine whether
signaling pathways share common mechanisms the MAPK pathway could serve as a pharmaco-
in a variety of different CNS disorders; therefore, logical target for SCA1. Human cells expressing
identifying this pathways role in the disease will ATXN1 were treated with a PDI84352 (MEK1/2
help to identify novel pharmacological targets inhibitor), GW5704 (RAF1 inhibitor), and a
and design new modifiers for CNS disorders. Ro31-8220 (MSK1 inhibitor). Pharmacological
Many neurodegenerative disorders, such as inhibition of the MAPK pathway led to decreased
AD, PD, and polyglutamine diseases, share a ATXN1. Moreover, the addition of MAPK inhibi-
common pathogenic mechanism: the abnormal tors to cerebellar slices decreased ATXN1 levels
accumulation of disease-causing proteins due (Emamian et al., 2003; Jorgensen et al., 2009;
to either the mutant proteins resistance to deg- Park etal.,2013).
radation or overexpression of the wild-type pro- It has become evident that second messengers
tein. In 2013, a research group at Baylor College such as cAMP, PKA, calcium, and diacylglycerol
of Medicine in the United States published a can control ERK signaling via the small G pro-
strategy to identify therapeutic entry points for teins Ras and Rap (Grewal etal., 1999). In intra-
such neurodegenerative disorders by screening cellular signal transduction, cAMP facilitates
for genetic networks that influence the levels of the transfer of the effects of hormones such as
disease-driving proteins (Park etal., 2013). To test glucagon and adrenaline through the cell mem-
this principle, they used spinocerebellar ataxia brane. It is involved in the activation of protein
type 1 (SCA1), which is an inherited neurode- kinases and regulates the effects of adrenaline
generative disease characterized by a progres- and glucagon. cAMP also binds to and regulates
sive loss of cerebellar neurons. It is caused by the the function of ion channels. Numerous studies
expansion of a CAG triplet repeat located in the have shown the particularly beneficial effects of
N-terminal coding region of the disease-causing cAMP to promote axonal regeneration, includ-
gene ATXN1. Pathogenic ATXN1 contains an ing but not limited to spinal cord injury (Hannila
abnormally elongated stretch of the amino acid and Filbin, 2008; Kajana and Goshgarian, 2008;
glutamine. This causes the protein to sponta- Lau etal., 2013; Qiu etal., 2002; Whitaker etal.,
neously misfold and form aggregates (Donato 2008). Qiu et al. showed that by increasing the
etal.,2012). intracellular levels of cAMP and protein kinase
Studies (Emamian et al., 2003; Jorgensen Aactivity, neurons overcame myelin-based inhi-
et al., 2009; Park et al., 2013) have shown that bition. Many different methods have been studied
neurodegeneration in SCA1 parallels with the in an effort to increase intracellular cAMP:stim-
levels of the mutant ATXN1 protein, and over- ulating adenylate cyclase with forskolin (Kilmer
expression of wild type ATXN1 results in neuro- and Carlsen, 1984), using cell-permeable cAMP
degeneration. Genetic network analysis revealed analogues such as dibutyryl cAMP or Sp-cAMPS
that the MAPK cascade was the most enriched (Bhatt et al., 2004; Cai et al., 1999; Song et al.,
where 6/10 genes belonged to the canonical 1998), initiating an intracellular signaling cas-
MAPK pathway (ERK1, ERK2, MED2, MEK3, cade that prevents cAMP degradation by using
MEK6, and MSK1). ATXN1 is known to impair neurotrophins such as NGF and BDNF (Cai etal.,
motor performance; to determine the effects of 1999; Hannila and Filbin, 2008), and inhibiting
Neurotrophic Regulation in Neurorestoration of theBrain 385

PDE4 activity (the major source of phosphodi- exacerbate the condition (Binder and Scharfman,
esterase activity in the CNS) with an antibody 2004). Whether BDNF affects neurons positively
(Iona etal.,1998). or negatively depends on the intracellular signal-
As discussed in the previous section, the ing pathways it activates. For example, reduced
p75NTR signaling pathway can induce neuronal BDNF and TrK B receptor have been found in AD
death, reduce axonal growth, and decrease syn- patients, and beta amyloid (142), a pro-apoptotic
aptic function. It has been hypothesized that the ligand for p75NTR, triggered p75-mediated cell
induction of p75NTR may be part of a homeo- death in AD pathology. BDNF treatment not only
static program that removes defective neurons, enhances the Trk/Akt cell survival signaling but
axons, and synapses upon limited injury and also precludes the production of A, thus sup-
degeneration. Adownside of this function is the pressing the p75 neurodegeneration pathway.
exacerbated damage that p75NTR signaling pro- Despite the well-demonstrated beneficial
duces following severe lesions and in pronounced effects of growth factors, one practical issue with
neurodegeneration. Because these circumstances their application is that they cannot cross the
are prevalent in neural injury, stroke and neuro- blood-brain barrier and they rapidly degrade
degenerative diseases in humans, there is a strong in the circulation, which limits their thera-
rationale for inhibiting p75NTR cell death sign- peutic potential. Small molecules have no dif-
aling as a therapeutic approach for these condi- ficulty crossing the blood-brain barrier. If they
tions (Ibanez and Simi, 2012). Direct application can mimic the action of growth factors on their
of neurotrophin to push the balance of Trk versus receptors, stimulate growth factor synthesis,
p75 signaling toward Trk cell survival signaling is potentiate the effects of endogenous growth, or
an obvious approach (Yano and Chao, 2000). In even manipulate the activities of key cell signal-
addition, antibody or antagonist to p75NTR also ing molecules in a common mechanism in favor
prevents apoptosis (Wang et al., 2008). Further of restoration, they will represent a logical and
studies are needed to address the time window promising strategy.
for application. Astudy in myocardial-infarcted In this regard, the functions of adenine-based
mice may give us some hint. This study showed purines such as adenosine and ATP as neuro-
that reduced BDNF levels modulated the early transmitters and neuromodulators in the CNS
inflammatory and neovascularization responses, have garnered wide interest in the past few dec-
leading to improved survival and reduced cardiac ades (Rathbone etal., 1991; 1999; 2008). As our
remodeling at day 28 post-myocardial infarction lab discovered, and as has been confirmed by
(Halade etal.,2013). many others, nonadenosine-based purine nucle-
osides and nucleotides, particularly guanosine,
CONCLUDING REMARKS play an important role in CNS signaling and
As widely demonstrated, neurotrophic factors may exert neuroprotective and neurorestorative
play a pivotal role in the neurorestoration and effects in response to physiological and patho-
neuroregeneration of damaged CNS neurons, logical conditions (Rathbone etal., 1999; 2008).
not only by potentiating the effects of intrinsic Most important, we recently discovered that
growth-promoting programs but also by coun- guanosine stimulates proliferation of endogenous
teracting at least some of the inhibitory signal- progenitor/stem cells in the adult mammalian
ing within the CNS. Growth factors such as NGF, nervous system following chronic disorders. The
BDNF, and FGF2 have been widely applied in intracellular mechanisms involved in the effects
both preclinical and clinical studies, adminis- of guanosine have yet to be fully elucidated.
tered alone or in combination with other phar- We have suggested that guanosine might exert
macotherapeutic agents, which indicates their its effect through a G-protein coupled receptor.
fundamental role in neurorestoration. When Both the cAMP-independent and -dependent
contemplating the use of growth factors in the mechanisms have been involved (Gysbers and
treatment of CNS disorders, the particular dis- Rathbone, 1996b). Several in vitro studies have
ease must be taken into consideration. For exam- shown that the cAMP-dependent mechanism, the
ple, using BDNF in neurodegenerative diseases MAPK pathway, including ERK-1 and ERK-2 (Di
such as AD or PD will supply neurotrophic sup- Iorio etal., 2001; Rathbone etal., 1998; Schwartz
port and stimulate restoration (Lu et al., 2013; and Mishler, 1990), and the cAMP-CREB path-
Murer et al., 2001); however, in epilepsy and way (Su etal., 2013)are involved. Guanosine may
seizures, further BDNF administration will either directly stimulate endogenous growth
386 Part III:Homeostatic Manipulators

factor generation (including NGF, BDNF) or Aguado, F., Carmona, M.A., Pozas, E., Aguilo, A.,
crosstalk with the TrK pathway signaling cas- Martinez-Guijarro, F.J., Alcantara, S., Borrell,
cade through ERK and PI3K, thereby inhibiting V., Yuste, R., Ibanez, C.F., and Soriano, E. (2003).
p75 NTR signaling to reduce apoptosis. Other BDNF regulates spontaneous correlated activity at
mechanisms underlying the biological effects of early developmental stages by increasing synapto-
guanosine are still to be discovered. genesis and expression of the K+/Cl co-transporter
Despite the few but impressive clinical suc- KCC2. Development 130, 12671280.
cesses, multiple breakthroughs in animal models, Aloe, L., Rocco, M.L., Bianchi, P., and Manni, L.
and speedy progress in the development of basic (2012). Nerve growth factor:From the early dis-
science research, there is still a gap from bench coveries to the potential clinical use. J Transl
to bedside, and few successful therapies or drugs Med 10,239.
are available to treat CNS disease. Guanosine Aoyagi, A., Nishikawa, K., Saito, H., and Abe, K.
is an example of a molecule that is able to exert (1994). Characterization of basic fibroblast
biological effects by regulating common intracel- growth factor-mediated acceleration of axonal
branching in cultured rat hippocampal neurons.
lular signaling cascades. By doing so, it is able to
Brain Res 661, 117126.
promote cell proliferation, differentiation, sur-
Arnold, A.P., and Gorski, R.A. (1984). Gonadal ste-
vival, and neurogenesis. Identifying the common
roid induction of structural sex differences in the
pathways in CNS disorders will provide novel
central nervous system. Annu Rev Neurosci 7,
pharmacological targets and, more important,
413442.
will open new avenues for combination thera-
Asai, T., Wanaka, A., Kato, H., Masana, Y., Seo, M.,
pies. The combination therapies that have tar- and Tohyama, M. (1993). Differential expression
geted multiple barriers to neuronal functional of two members of FGF receptor gene family,
recovery have provided encouraging progress FGFR-1 and FGFR-2 mRNA, in the adult rat cen-
and represent better therapeutic interventions. tral nervous system. Brain Res Mol Brain Res 17,
Neurotrophic factors play a fundamental role 174178.
in building a permissive microenvironment for Asher, R.A., Morgenstern, D.A., Fidler, P.S., Adcock,
restoration. Intracellular signaling pathways of a K.H., Oohira, A., Braistead, J.E., Levine, J.M.,
variety of CNS disorders share a common sign- Margolis, R.U., Rogers, J.H., and Fawcett, J.W.
aling cascade. Research targeting the converging (2000). Neurocan is upregulated in injured brain
molecules on the common signaling transduc- and in cytokine-treated astrocytes. J Neurosci
tion cascade may lead to the development of a 20, 24272438.
new generation of therapeutics with better selec- Asher, R.A., Morgenstern, D.A., Shearer, M.C.,
tivity and potency. Adcock, K.H., Pesheva, P., and Fawcett, J.W.
(2002). Versican is upregulated in CNS injury
ACK NOWLEDGMENTS and is a product of oligodendrocyte lineage cells.
We thank Ms. Ann Kolkin for her helpful sugges- J Neurosci 22, 22252236.
tions and for her critical editorial review. Ballerini, P., Ciccarelli, R., Di Iorio, P., Buccella, S.,
DAlimonte, I., Giuliani, P., Masciulli, A., Nargi,
References E., Beraudi, A., Rathbone, M.P., et al. (2006).
Abraham, J.A., Mergia, A., Whang, J.L., Tumolo, A., Guanosine effect on cholesterol efflux and apoli-
Friedman, J., Hjerrild, K.A., Gospodarowicz, D., poprotein E expression in astrocytes. Purinergic
and Fiddes, J.C. (1986a). Nucleotide sequence Signal 2, 637649.
of a bovine clone encoding the angiogenic pro- Barde, Y.A., Edgar, D., and Thoenen, H. (1982).
tein, basic fibroblast growth factor. Science 233, Purification of a new neurotrophic factor from
545548. mammalian brain. EMBO J 1, 549553.
Abraham, J.A., Whang, J.L., Tumolo, A., Mergia, A., Baron, O., Ratzka, A., and Grothe, C. (2012).
and Fiddes, J.C. (1986b). Human basic fibroblast Fibroblast growth factor 2 regulates adequate
growth factor:Nucleotide sequence, genomic orga- nigrostriatal pathway formation in mice. J Comp
nization, and expression in mammalian cells. Cold Neurol 520, 39493961.
Spring Harb Symp Quant Biol 51 Pt 1, 657668. Bednar, M.M. (2008). The role of sildenafil in the
Acheson, A., Conover, J.C., Fandl, J.P., DeChiara, treatment of stroke. Curr Opin Investig Drugs 9,
T.M., Russell, M., Thadani, A., Squinto, S.P., 754759.
Yancopoulos, G.D., and Lindsay, R.M. (1995). A Bednar, M.M., and Perry, A. (2012). Neurorestoration
BDNF autocrine loop in adult sensory neurons therapeutics for neurodegenerative and psychi-
prevents cell death. Nature 374, 450453. atric disease. Neurol Res 34, 129142.
Neurotrophic Regulation in Neurorestoration of theBrain 387

Benarroch, E.E. (2005). Neuron-astrocyte interac- blocks inhibition of axonal regeneration by MAG
tions:partnership for normal function and dis- and myelin via a cAMP-dependent mechanism.
ease in the central nervous system. Mayo Clin Neuron 22, 89101.
Proc 80, 13261338. Cattaneo, A. (2010). Tanezumab, a recombinant
Benraiss, A., Chmielnicki, E., Lerner, K., Roh, D., and humanized mAb against nerve growth factor for
Goldman, S.A. (2001). Adenoviral brain-derived the treatment of acute and chronic pain. Curr
neurotrophic factor induces both neostriatal and Opin Mol Ther 12, 94106.
olfactory neuronal recruitment from endog- Chang, R., Algird, A., Bau, C., Rathbone, M.P., and
enous progenitor cells in the adult forebrain. J Jiang, S. (2008). Neuroprotective effects of gua-
Neurosci 21, 67186731. nosine on stroke models in vitro and in vivo.
Bertram, J.P., Rauch, M.F., Chang, K., and Lavik, E.B. Neurosci Lett 431, 101105.
(2010). Using polymer chemistry to modulate the Chao, M.V. (2003). Neurotrophins and their recep-
delivery of neurotrophic factors from degrad- tors: A convergence point for many signalling
able microspheres:delivery of BDNF. Pharm Res pathways. Nature Rev 4, 299309.
27,8291. Chaturvedi, R.K., Shukla, S., Seth, K., and Agrawal,
Bhatt, D.H., Otto, S.J., Depoister, B., and Fetcho, J.R. A.K. (2006). Nerve growth factor increases sur-
(2004). Cyclic AMP-induced repair of zebrafish vival of dopaminergic graft, rescue nigral dopa-
spinal circuits. Science 305, 254258. minergic neurons and restores functional deficits
Binder, D.K., and Scharfman, H.E. (2004). Brain- in rat model of Parkinsons disease. Neurosci
derived neurotrophic factor. Growth Factors 22, Lett 398,4449.
123131. Chen, L.W., Horng, L.Y., Wu, C.L., Sung, H.C., and
Bjorklund, A., Johansson, B., Stenevi, U., and Wu, R.T. (2012). Activating mitochondrial regu-
Svendgaard, N.A. (1975). Re-establishment of lator PGC-1alpha expression by astrocytic NGF
functional connections by regenerating central is a therapeutic strategy for Huntingtons dis-
adrenergic and cholinergic axons. Nature 253, ease. Neuropharmacology 63, 719732.
446448. Chen, M.S., Huber, A.B., van der Haar, M.E.,
Bjorklund, A., Katzman, R., Stenevi, U., and Frank, M., Schnell, L., Spillmann, A.A., Christ,
West, K.A. (1971). Development and growth F., and Schwab, M.E. (2000). Nogo-A is a
of axonal sprouts from noradrenaline and myelin-associated neurite outgrowth inhibitor
5-hydroxytryptamine neurones in the rat spinal and an antigen for monoclonal antibody IN-1.
cord. Brain Res 31,2133. Nature 403, 434439.
Bocklinger, K., Tomaselli, B., Heftberger, V., Chen, Y., Zeng, J., Cen, L., Wang, X., Yao, G., Wang,
Podhraski, V., Bandtlow, C., and Baier-Bitterlich, W., Qi, W., and Kong, K. (2009). Multiple roles
G. (2004). Purine nucleosides support the neurite of the p75 neurotrophin receptor in the nervous
outgrowth of primary rat cerebellar granule cells system. J Int Med Res 37, 281288.
after hypoxia. Eur J Cell Biol 83,5154. Cheng, S., Ma, M., Ma, Y., Wang, Z., Xu, G., and Liu,
Bovolenta, P., Wandosell, F., and Nieto-Sampedro, X. (2009). Combination therapy with intranasal
M. (1991). Neurite outgrowth over resting and NGF and electroacupuncture enhanced cell pro-
reactive astrocytes. Restor Neurol Neurosci 2, liferation and survival in rats after stroke. Neurol
221228. Res 31, 753758.
Bretzner, F., Plemel, J.R., Liu, J., Richter, M., Roskams, Chiquet, M. (1989). Neurite growth inhibition by
A.J., and Tetzlaff, W. (2010). Combination of CNS myelin proteins: A mechanism to confine
olfactory ensheathing cells with local versus sys- fiber tracts? Trends Neurosci 12,13.
temic cAMP treatment after a cervical rubrospi- Choi, D.Y., Toledo-Aral, J.J., Segal, R., and Halegoua,
nal tract injury. J Neurosci Res 88, 28332846. S. (2001). Sustained signaling by phospho-
Burnstock, G. (1972). Purinergic nerves. Pharmacol lipase C-gamma mediates nerve growth
Rev 24, 509581. factor-triggered gene expression. Mol Cell Biol
Burnstock, G. (2013). Purinergic signalling: 21, 26952705.
Pathophysiology and therapeutic potential. Keio Cobb, M.H. (1999). MAP kinase pathways. Prog
J Med 62,6373. Biophys Mol Biol 71, 479500.
Cacioppo, J., and Berntson, G. (2011). The Brain, Connell, B.J., Di Iorio, P., Sayeed, I., Ballerini, P.,
Homeostasis, and Health:Balancing Demands of Saleh, M.C., Giuliani, P., Saleh, T.M., Rathbone,
the Internal and External Milieu (Oxford:Oxford M.P., Su, C., and Jiang, S. (2013). Guanosine
University Press). protects against reperfusion injury in rat
Cai, D., Shen, Y., De Bellard, M., Tang, S., and Filbin, brains after ischemic stroke. J Neurosci Res 91,
M.T. (1999). Prior exposure to neurotrophins 262272.
388 Part III:Homeostatic Manipulators

Connor, B., and Dragunow, M. (1998). The role of for multiplex signalling. Biochem J 318 (Pt 3),
neuronal growth factors in neurodegenerative 729747.
disorders of the human brain. Brain Res Brain Deshmukh, M., Vasilakos, J., Deckwerth, T.L.,
Res Rev 27,139. Lampe, P.A., Shivers, B.D., and Johnson, E.M.,
Connor, B., Young, D., Yan, Q., Faull, R.L., Synek, B., Jr. (1996). Genetic and metabolic status of
and Dragunow, M. (1997). Brain-derived neuro- NGF-deprived sympathetic neurons saved by an
trophic factor is reduced in Alzheimers disease. inhibitor of ICE family proteases. J Cell Biol 135,
Brain Res Mol Brain Res 49,7181. 13411354.
Costantini, C., Rossi, F., Formaggio, E., Bernardoni, Di Iorio, P., Ballerini, P., Traversa, U., Nicoletti, F.,
R., Cecconi, D., and Della-Bianca, V. (2005). DAlimonte, I., Kleywegt, S., Werstiuk, E.S.,
Characterization of the signaling pathway Rathbone, M.P., Caciagli, F., and Ciccarelli, R.
downstream p75 neurotrophin receptor involved (2004). The antiapoptotic effect of guanosine is
in beta-amyloid peptide-dependent cell death. J mediated by the activation of the PI 3-kinase/
Mol Neurosci 25, 141156. AKT/PKB pathway in cultured rat astrocytes.
Coulson, E.J., Reid, K., Murray, S.S., Cheema, S.S., Glia 46, 356368.
and Bartlett, P.F. (2000). Role of neurotrophin Di Iorio, P., Caciagli, F., Giuliani, P., Ballerini, P.,
receptor p75NTR in mediating neuronal cell and Ciccarelli, R. (2001). Purine nucleosides
death following injury. Clin Exp Pharmacol protect injured neurons and stimulate neuro-
Physiol 27, 537541. nal regeneration by intracellular and membrane
Croll, S.D., Suri, C., Compton, D.L., Simmons, M.V., receptor-mediated mechanisms. Drug Develop
Yancopoulos, G.D., Lindsay, R.M., Wiegand, Res, 303315.
S.J., Rudge, J.S., and Scharfman, H.E. (1999). Dittrich, F., Ochs, G., Grosse-Wilde, A., Berweiler, U.,
Brain-derived neurotrophic factor transgenic Yan, Q., Miller, J.A., Toyka, K.V., and Sendtner,
mice exhibit passive avoidance deficits, increased M. (1996). Pharmacokinetics of intrathecally
seizure severity and in vitro hyperexcitabil- applied BDNF and effects on spinal motoneu-
ity in the hippocampus and entorhinal cortex. rons. Exp Neurol 141, 225239.
Neuroscience 93, 14911506. Dobrowsky, R.T., Werner, M.H., Castellino, A.M.,
Cunha, F. (1949). The Edwin Smith surgical papyrus. Chao, M.V., and Hannun, Y.A. (1994). Activation
Am J Surg 77, 793795. of the sphingomyelin cycle through the
Dal-Cim, T., Martins, W.C., Santos, A.R., and Tasca, low-affinity neurotrophin receptor. Science 265,
C.I. (2011). Guanosine is neuroprotective against 15961599.
oxygen/glucose deprivation in hippocampal Domeniconi, M., Cao, Z., Spencer, T., Sivasankaran, R.,
slices via large conductance Ca(2)+-activated Wang, K., Nikulina, E., Kimura, N., Cai, H., Deng,
K+ channels, phosphatidilinositol-3 kinase/pro- K., Gao, Y., et al. (2002). Myelin-associated gly-
tein kinase B pathway activation and glutamate coprotein interacts with the Nogo66 receptor to
uptake. Neuroscience 183, 212220. inhibit neurite outgrowth. Neuron 35, 283290.
David, S., and Aguayo, A.J. (1981). Axonal elonga- Donato, S.D., Mariotti, C., and Taroni, F. (2012).
tion into peripheral nervous system bridges Spinocerebellar ataxia type 1. Handb Clin
after central nervous system injury in adult rats. Neurol 103, 399421.
Science 214, 931933. Dreyfus, C.F. (1989). Effects of nerve growth factor
Davis, R.J. (1993). The mitogen-activated protein on cholinergic brain neurons. Trends Pharmacol
kinase signal transduction pathway. J Biol Chem Sci 10, 145149.
268, 1455314556. Dumont, R.J., Okonkwo, D.O., Verma, S.,
DeFreitas, M.F., McQuillen, P.S., and Shatz, C.J. Hurlbert, R.J., Boulos, P.T., Ellegala, D.B., and
(2001). A novel p75NTR signaling pathway pro- Dumont, A.S. (2001). Acute spinal cord injury,
motes survival, not death, of immunopurified part I: Pathophysiologic mechanisms. Clin
neocortical subplate neurons. J Neurosci 21, Neuropharmacol 24, 254264.
51215129. Duncan, I.D., Aguayo, A.J., Bunge, R.P., and Wood,
Demirtas-Tatlidede, A., Vahabzadeh-Hagh, A.M., P.M. (1981). Transplantation of rat Schwann cells
and Pascual-Leone, A. (2013). Can noninvasive grown in tissue culture into the mouse spinal
brain stimulation enhance cognition in neuro- cord. J Neurol Sci 49, 241252.
psychiatric disorders? Neuropharmacology 64, Dutta, S., Singh, G., Sreejith, S., Mamidi, M.K.,
566578. Husin, J.M., Datta, I., Pal, R., and Das, A.K.
Denhardt, D.T. (1996). Signal-transducing protein (2013). Cell therapy:The final frontier for treat-
phosphorylation cascades mediated by Ras/Rho ment of neurological diseases. CNS Neurosci
proteins in the mammalian cell: the potential Ther 19,511.
Neurotrophic Regulation in Neurorestoration of theBrain 389

Eckenstein, F., Woodward, W.R., and Nishi, R. of central nervous system architecture and
(1991). Differential localization and possible function, part 1: Foundations and historical
functions of aFGF and bFGF in the central and landmarks in contemporary stem cell biology.
peripheral nervous systems. Annals N.Y. Acad. Neurosurgery 64, 1539; discussion34.
Sci 638, 348360. Filbin, M.T. (1995). Myelin-associated glycopro-
Emamian, E.S., Kaytor, M.D., Duvick, L.A., Zu, T., tein:Arole in myelination and in the inhibition
Tousey, S.K., Zoghbi, H.Y., Clark, H.B., and Orr, of axonal regeneration? Curr Opin Neurobiol 5,
H.T. (2003). Serine 776 of ataxin-1 is critical for 588595.
polyglutamine-induced disease in SCA1 trans- Fitch, M.T., and Silver, J. (1997). Glial cell extracel-
genic mice. Neuron 38, 375387. lular matrix: Boundaries for axon growth in
Emoto, N., Gonzalez, A.M., Walicke, P.A., Wada, E., development and regeneration. Cell Tissue Res
Simmons, D.M., Shimasaki, S., and Baird, A. 290, 379384.
(1989). Basic fibroblast growth factor (FGF) in Foehr, E.D., Lin, X., OMahony, A., Geleziunas,
the central nervous system:identification of spe- R., Bradshaw, R.A., and Greene, W.C. (2000).
cific Loci of basic FGF expression in the rat brain. NF-kappa B signaling promotes both cell sur-
Growth Factors 2,2129. vival and neurite process formation in nerve
English, J.D., and Sweatt, J.D. (1997). A requirement growth factor-stimulated PC12 cells. J Neurosci
for the mitogen-activated protein kinase cascade 20, 75567563.
in hippocampal long term potentiation. J Biol Frade, J.M., and Barde, Y.A. (1998). Nerve growth
Chem 272, 1910319106. factor: Two receptors, multiple functions.
Ergul, A., Alhusban, A., and Fagan, S.C. (2012). Bioessays 20, 137145.
Angiogenesis:Aharmonized target for recovery Freire, M.A. (2012). Pathophysiology of neurodegen-
after stroke. Stroke 43, 22702274. eration following traumatic brain injury. West
Eriksdotter-Jonhagen, M., Nordberg, A., Amberla, Indian Med J 61, 751755.
K., Backman, L., Ebendal, T., Meyerson, B., Frielingsdorf, H., Simpson, D.R., Thal, L.J., and
Olson, L., Seiger, Shigeta, M., Theodorsson, E., Pizzo, D.P. (2007). Nerve growth factor promotes
et al. (1998). Intracerebroventricular infusion survival of new neurons in the adult hippocam-
of nerve growth factor in three patients with pus. Neurobiol Dis 26,4755.
Alzheimers disease. Dement Geriatr Cogn Friesel, R., and Maciag, T. (1999). Fibroblast growth
Disord 9, 246257. factor prototype release and fibroblast growth
Eriksdotter-Jonhagen, M., Linderoth, B., Lind, factor receptor signaling. Thromb Haemost 82,
G., Aladellie, L., Almkvist, O., Andreasen, N., 748754.
Blennow, K., Bogdanovic, N., Jelic, V., Kadir, A., Fryer, H.J., Wolf, D.H., Knox, R.J., Strittmatter, S.M.,
et al. (2012). Encapsulated cell biodelivery of Pennica, D., OLeary, R.M., Russell, D.S., and
nerve growth factor to the basal forebrain in Kalb, R.G. (2000). Brain-derived neurotrophic
patients with Alzheimers disease. Dement factor induces excitotoxic sensitivity in cultured
Geriatr Cogn Disord 33,1828. embryonic rat spinal motor neurons through
Eriksson, P.S., Perfilieva, E., Bjork-Eriksson, T., activation of the phosphatidylinositol 3-kinase
Alborn, A.M., Nordborg, C., Peterson, D.A., pathway. J Neurochem 74, 582595.
and Gage, F.H. (1998). Neurogenesis in the adult Fukuda, M., Gotoh, Y., Tachibana, T., Dell, K.,
human hippocampus. Nat Med 4, 13131317. Hattori, S., Yoneda, Y., and Nishida, E. (1995).
Ernfors, P., Lonnerberg, P., Ayer-LeLievre, C., and Induction of neurite outgrowth by MAP kinase
Persson, H. (1990). Developmental and regional in PC12 cells. Oncogene 11, 239244.
expression of basic fibroblast growth factor Gabathuler, R. (2010). Approaches to transport ther-
mRNA in the rat central nervous system. J apeutic drugs across the blood-brain barrier to
Neurosci Res 27,1015. treat brain diseases. Neurobiol Dis 37,4857.
Everall, I.P., and Kerwin, R. (1990). The role of nerve Gage, F.H., Ray, J., and Fisher, L.J. (1995). Isolation,
growth factor in Alzheimers disease. Psychol characterization, and use of stem cells from the
Med 20, 249251. CNS. Annu Rev Neurosci 18, 159192.
Fan, X.Y., Mothe, A.J., and Tator, C.H. (2013). Gall, C.M., Gold, S.J., Isackson, P.J., and Seroogy,
Ephrin-B3 decreases the survival of adult rat spi- K.B. (1992). Brain-derived neurotrophic factor
nal cord-derived neural stem/progenitor cells in and neurotrophin-3 mRNAs are expressed in
vitro and after transplantation into the injured ventral midbrain regions containing dopaminer-
rat spinal cord. Stem Cells Dev 22, 359373. gic neurons. Mol Cell Neurosci 3,5663.
Farin, A., Liu, C.Y., Elder, J.B., Langmoen, I.A., and Gral, C., Angelova, A., and Lesieur, S. (2013).
Apuzzo, M.L. (2009). The biological restoration From molecular to nanotechnology strategies
390 Part III:Homeostatic Manipulators

for delivery of neurotrophins: Emphasis on Gysbers, J.W., and Rathbone, M.P. (1996b). Neurite
brain-derived neurotrophic factor (BDNF). outgrowth in PC12 cells is enhanced by gua-
Pharmaceutics 5, 127167. nosine through both cAMP-dependent and
Giuliani, P., Romano, S., Ballerini, P., Ciccarelli, R., independent mechanisms. Neurosci Lett 220,
Petragnani, N., Cicchitti, S., Zuccarini, M., Jiang, 175178.
S., Rathbone, M.P., Caciagli, F., et al. (2012). Hadari, Y.R., Gotoh, N., Kouhara, H., Lax, I.,
Protective activity of guanosine in an in vitro and Schlessinger, J. (2001). Critical role for
model of Parkinsons disease. Panminerva Med the docking-protein FRS2 alpha in FGF
54,4351. receptor-mediated signal transduction pathways.
Glasky, A.J., Glasky, M.S., Ritzmann, R.F., and Proc Natl Acad Sci USA 98, 85788583.
Rathbone, M.P. (1997). AIT-082, a novel purine Halade, G.V., Ma, Y., Ramirez, T.A., Zhang, J., Dai,
derivative with neuroregenerative properties. Q., Hensler, J.G., Lopez, E.F., Ghasemi, O., Jin,
Expert Opin Investig Drugs 6, 14131417. Y.F., and Lindsey, M.L. (2013). Reduced BDNF
Glasky, A.J., Kirat, S., Middlemiss, P.J., Gysbers, attenuates inflammation and angiogenesis to
J.W., and Rathbone, M.P. (1995). A novel purine improve survival and cardiac function following
derivative AIT-082 increases synthesis of NGF, myocardial infarction in mice. Am J Physiol 305,
FGF-2 and NT-3 mRNA in astrocytes. Abstr Soc H1830H1842.
Neurosci,295. Hamanoue, M., Middleton, G., Wyatt, S., Jaffray, E.,
Goedert, M., Fine, A., Hunt, S.P., and Ullrich, A. Hay, R.T., and Davies, A.M. (1999). p75-mediated
(1986). Nerve growth factor mRNA in peripheral NF-kappaB activation enhances the survival
and central rat tissues and in the human central response of developing sensory neurons to nerve
nervous system: Lesion effects in the rat brain growth factor. Mol Cell Neurosci 14,2840.
and levels in Alzheimers disease. Brain Res Hannila, S.S., and Filbin, M.T. (2008). The role of
387,8592. cyclic AMP signaling in promoting axonal
Goldberger, M.E., and Murray, M. (1974). Restitution regeneration after spinal cord injury. Exp Neurol
of function and collateral sprouting in the cat 209, 321332.
spinal cord: The deafferented animal. J Comp Harbaugh, R.E. (1989). Nerve growth factor as a
Neurol 158,3753. potential treatment in Alzheimers disease.
Golden, E., Emiliano, A., Maudsley, S., Windham, Biomed Pharmacother 43, 483485.
B.G., Carlson, O.D., Egan, J.M., Driscoll, I., Harrington, A.W., Kim, J.Y., and Yoon, S.O. (2002).
Ferrucci, L., Martin, B., and Mattson, M.P. Activation of Rac GTPase by p75 is necessary for
(2010). Circulating brain-derived neurotrophic c-jun N-terminal kinase-mediated apoptosis. J
factor and indices of metabolic and cardiovascu- Neurosci 22, 156166.
lar health:Data from the Baltimore Longitudinal Harrington, A.W., Leiner, B., Blechschmitt, C.,
Study of Aging. PLoS One 5, e10099. Arevalo, J.C., Lee, R., Morl, K., Meyer, M.,
Grewal, S.S., York, R.D., and Stork, P.J. (1999). Hempstead, B.L., Yoon, S.O., and Giehl, K.M.
Extracellular-signal-regulated kinase signalling (2004). Secreted proNGF is a pathophysiologi-
in neurons. Curr Opin Neurobiol 9, 544553. cal death-inducing ligand after adult CNS injury.
Gritti, A., Parati, E.A., Cova, L., Frolichsthal, P., Proc Natl Acad Sci USA 101, 62266230.
Galli, R., Wanke, E., Faravelli, L., Morassutti, Hefti, F.F., Rosenthal, A., Walicke, P.A., Wyatt, S.,
D.J., Roisen, F., Nickel, D.D., et al. (1996). Vergara, G., Shelton, D.L., and Davies, A.M.
Multipotential stem cells from the adult mouse (2006). Novel class of pain drugs based on
brain proliferate and self-renew in response to antagonism of NGF. Trends Pharmacol Sci
basic fibroblast growth factor. J Neurosci 16, 27,8591.
10911100. Hempstead, B.L. (2006). Dissecting the diverse
Guo, Q., Li, S., and Su, B. (2007). Expression of oligo- actions of pro- and mature neurotrophins. Curr
dendrocyte myelin glycoprotein and its receptor Alzheimer Res 3,1924.
NgR after the injury of rat central nervous sys- Hou, S.T., Jiang, S.X., and Smith, R.A. (2008).
tem. Neurosci Lett 422, 103108. Permissive and repulsive cues and signalling
Gysbers, J.W., and Rathbone, M.P. (1992). Guanosine pathways of axonal outgrowth and regeneration.
enhances NGF-stimulated neurite outgrowth in Int Rev Cell Mol Biol 267, 125181.
PC12 cells. Neuroreport 3, 9971000. Howells, D.W., Porritt, M.J., Wong, J.Y., Batchelor,
Gysbers, J.W., and Rathbone, M.P. (1996a). GTP and P.E., Kalnins, R., Hughes, A.J., and Donnan,
guanosine synergistically enhance NGF-induced G.A. (2000). Reduced BDNF mRNA expression
neurite outgrowth from PC12 cells. Int J Dev in the Parkinsons disease substantia nigra. Exp
Neurosci 14,1934. Neurol 166, 127135.
Neurotrophic Regulation in Neurorestoration of theBrain 391

Huang, E.J., and Reichardt, L.F. (2001). Jiang, S., Khan, M.I., Wang, J., Middlemiss, P.J.,
Neurotrophins: Roles in neuronal development Werstiuk, E.S., Wickson, R., and Rathbone, M.P.
and function. Annu Rev Neurosci 24, 677736. (2003b). Enteric glia promote functional recov-
Ibanez, C.F., and Simi, A. (2012). p75 neurotrophin ery of CTM reflex after dorsal root transection.
receptor signaling in nervous system injury and Neuroreport 14, 13011304.
degeneration:Paradox and opportunity. Trends Jiang, S., Wang, J., Khan, M.I., Middlemiss, P.J.,
Neurosci 35, 431440. Salgado-Ceballos, H., Werstiuk, E.S., Wickson,
Imagama, S., Sakamoto, K., Tauchi, R., Shinjo, R., R., and Rathbone, M.P. (2003c). Enteric glia
Ohgomori, T., Ito, Z., Zhang, H., Nishida, Y., promote regeneration of transected dorsal root
Asami, N., Takeshita, S., et al. (2011). Keratan axons into spinal cord of adult rats. Exp Neurol
sulfate restricts neural plasticity after spinal cord 181,7983.
injury. J Neurosci 31, 1709117102. Johnson, D., Lanahan, A., Buck, C.R., Sehgal, A.,
Iona, S., Cuomo, M., Bushnik, T., Naro, F., Sette, C., Morgan, C., Mercer, E., Bothwell, M., and Chao,
Hess, M., Shelton, E.R., and Conti, M. (1998). M. (1986). Expression and structure of the
Characterization of the rolipram-sensitive, cyclic human NGF receptor. Cell 47, 545554.
AMP-specific phosphodiesterases:Identification Jorgensen, N.D., Andresen, J.M., Lagalwar, S.,
and differential expression of immunologically Armstrong, B., Stevens, S., Byam, C.E., Duvick,
distinct forms in the rat brain. Mol Pharmacol L.A., Lai, S., Jafar-Nejad, P., Zoghbi, H.Y., etal.
53,2332. (2009). Phosphorylation of ATXN1 at Ser776 in
Ishiyama, J., Saito, H., and Abe, K. (1991). Epidermal the cerebellum. J Neurochem 110, 675686.
growth factor and basic fibroblast growth factor Kajana, S., and Goshgarian, H.G. (2008). Spinal acti-
promote the generation of long-term potentia- vation of the cAMP-PKA pathway induces respi-
tion in the dentate gyrus of anaesthetized rats. ratory motor recovery following high cervical
Neurosci Res 12, 403411. spinal cord injury. Brain Res 1232, 206213.
Iwai-Kanai, E., Hasegawa, K., Fujita, M., Araki, Katoh, M. (2006). FGF signaling network in the
M., Yanazume, T., Adachi, S., and Sasayama, S. gastrointestinal tract (review). Int J Oncol 29,
(2002). Basic fibroblast growth factor protects 163168.
cardiac myocytes from iNOS-mediated apopto- Katzman, R., Bjorklund, A., Owman, C., Stenevi, U.,
sis. J Cell Physiol 190,5462. and West, K.A. (1971). Evidence for regenerative
Jaye, M., Schlessinger, J., and Dionne, C.A. (1992). axon sprouting of central catecholamine neurons
Fibroblast growth factor receptor tyrosine in the rat mesencephalon following electrolytic
kinases:Molecular analysis and signal transduc- lesions. Brain Res 25, 579596.
tion. Biochim Biophys Acta 1135, 185199. Kennedy, S.G., Wagner, A.J., Conzen, S.D., Jordan, J.,
Jiang, S., Ballerini, P., Buccella, S., Giuliani, P., Jiang, Bellacosa, A., Tsichlis, P.N., and Hay, N. (1997).
C., Huang, X., and Rathbone, M.P. (2008a). The PI 3-kinase/Akt signaling pathway delivers
Remyelination after chronic spinal cord injury an anti-apoptotic signal. Genes Dev 11, 701713.
is associated with proliferation of endogenous Kilmer, S.L., and Carlsen, R.C. (1984). Forskolin
adult progenitor cells after systemic administra- activation of adenylate cyclase in vivo stimulates
tion of guanosine. Purinergic Signal 4,6171. nerve regeneration. Nature 307, 455457.
Jiang, S., Bendjelloul, F., Ballerini, P., DAlimonte, Kilpatrick, T.J., and Bartlett, P.F. (1993). Cloning
I., Nargi, E., Jiang, C., Huang, X., and Rathbone, and growth of multipotential neural precur-
M.P. (2007). Guanosine reduces apoptosis and sors: Requirements for proliferation and differ-
inflammation associated with restoration of entiation. Neuron 10, 255265.
function in rats with acute spinal cord injury. Kim, H.J., Kim, J.H., Bae, S.C., Choi, J.Y., and
Purinergic Signal 3, 411421. Ryoo, H.M. (2003). The protein kinase C path-
Jiang, S., Fischione, G., Giuliani, P., Romano, S., way plays a central role in the fibroblast growth
Caciagli, F., and Di Iorio, P. (2008b). Metabolism factor-stimulated expression and transactivation
and distribution of guanosine given intraperi- activity of Runx2. J Biol Chem 278, 319326.
toneally: Implications for spinal cord injury. Kim, J.K., Rathbone, M.P., Middlemiss, P.J., Hughes,
Nucleosides Nucleotides Nucleic Acids 27, D.W., and Smith, R.W. (1991). Purinergic stimu-
673680. lation of astroblast proliferation:guanosine and
Jiang, S., Khan, M.I., Lu, Y., Wang, J., Buttigieg, J., its nucleotides stimulate cell division in chick
Werstiuk, E.S., Ciccarelli, R., Caciagli, F., and astroblasts. J Neurosci Res 28, 442455.
Rathbone, M.P. (2003a). Guanosine promotes Kivitz, A.J., Gimbel, J.S., Bramson, C., Nemeth,
myelination and functional recovery in chronic M.A., Keller, D.S., Brown, M.T., West, C.R.,
spinal injury. Neuroreport 14, 24632467. and Verburg, K.M. (2013). Efficacy and safety of
392 Part III:Homeostatic Manipulators

tanezumab versus naproxen in the treatment of Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai) 32,
chronic low back pain. Pain 154, 10091021. 413416.
Ko, M.K., and Kay, E.P. (2005). Regulatory role of Lin, J.C., Tsao, D., Barras, P., Bastarrachea, R.A.,
FGF-2 on type I collagen expression during Boyd, B., Chou, J., Rosete, R., Long, H., Forgie,
endothelial mesenchymal transformation. Invest A., Abdiche, Y., etal. (2008). Appetite enhance-
Ophthalmol Vis Sci 46, 44954503. ment and weight gain by peripheral administra-
Kodaki, T., Woscholski, R., Hallberg, B., Rodriguez- tion of TrkB agonists in non-human primates.
Viciana, P., Downward, J., and Parker, P.J. (1994). PLoS One 3,e1900.
The activation of phosphatidylinositol 3-kinase Linnarsson, S., Bjorklund, A., and Ernfors, P. (1997).
by Ras. Curr Biol 4, 798806. Learning deficit in BDNF mutant mice. Eur J
Kokaia, M., Ernfors, P., Kokaia, Z., Elmer, E., Neurosci 9, 25812587.
Jaenisch, R., and Lindvall, O. (1995). Suppressed Liu, R.J., Lee, F.S., Li, X.Y., Bambico, F., Duman, R.S.,
epileptogenesis in BDNF mutant mice. Exp and Aghajanian, G.K. (2012). Brain-derived neu-
Neurol 133, 215224. rotrophic factor Val66Met allele impairs basal
Koudinov, A.R., and Koudinova, N.V. (2001). and ketamine-stimulated synaptogenesis in pre-
Essential role for cholesterol in synaptic plas- frontal cortex. Biol Psychiatry 71, 9961005.
ticity and neuronal degeneration. FASEB J 15, Lobato, R.D. (2008). Historical vignette of Cajals
18581860. work Degeneration and regeneration of the
Lampe, K.J., Kern, D.S., Mahoney, M.J., and Bjugstad, nervous system with a reflection of the author.
K.B. (2011). The administration of BDNF and Neurocirugia (Astur) 19, 456468.
GDNF to the brain via PLGA microparticles pat- Lorentz, C.U., Parrish, D.C., Alston, E.N., Pellegrino,
terned within a degradable PEG-based hydro- M.J., Woodward, W.R., Hempstead, B.L., and
gel: Protein distribution and the glial response. Habecker, B.A. (2013). Sympathetic denerva-
J Biomed Mater Res A 96, 595607. tion of peri-infarct myocardium requires the p75
Lau, B.Y., Fogerson, S.M., Walsh, R.B., and Morgan, neurotrophin receptor. Exp Neurol 249, 111119.
J.R. (2013). Cyclic AMP promotes axon regen- Lu, B., Nagappan, G., Guan, X., Nathan, P.J., and
eration, lesion repair and neuronal survival in Wren, P. (2013). BDNF-based synaptic repair as
lampreys after spinal cord injury. Exp Neurol a disease-modifying strategy for neurodegenera-
250C,3142. tive diseases. Nature Rev Neurosci 14, 401416.
Leconte, I., Fox, J.C., Baldwin, H.S., Buck, C.A., and Luk, Y.O., Chen, W.Y., Wong, W.J., Hu, H.H., Hsu,
Swain, J.L. (1998). Adenoviral-mediated expres- L.C., Chern, C.M., Huang, K.J., and Law, S.L.
sion of antisense RNA to fibroblast growth fac- (2004). Treatment of focal cerebral ischemia with
tors disrupts murine vascular development. Dev liposomal nerve growth factor. Drug Deliv 11,
Dyn 213, 421430. 319324.
Lee, H.T., and Kay, E.P. (2003). FGF-2 induced reor- MacPhee, I.J., and Barker, P.A. (1997). Brain-derived
ganization and disruption of actin cytoskeleton neurotrophic factor binding to the p75 neu-
through PI 3-kinase, Rho, and Cdc42 in corneal rotrophin receptor reduces TrkA signaling
endothelial cells. Mol Vis 9, 624634. while increasing serine phosphorylation in the
Lee, J.G., and Kay, E.P. (2006). Cross-talk among TrkA intracellular domain. J Biol Chem 272,
Rho GTPases acting downstream of PI 3-kinase 2354723551.
induces mesenchymal transformation of cor- Mandel, R.J. (2010). CERE-110, an adeno-associated
neal endothelial cells mediated by FGF-2. Invest virus-based gene delivery vector expressing
Ophthalmol Vis Sci 47, 23582368. human nerve growth factor for the treatment
Leisman, D. (1990). The brain: A dynamic system of Alzheimers disease. Curr Opin Mol Ther 12,
tending toward homeostasis. Int J Neurosci 54, 240247.
119124. Mantyh, P.W., Koltzenburg, M., Mendell, L.M., Tive,
Levine, J.M. (1994). Increased expression of the NG2 L., and Shelton, D.L. (2011). Antagonism of nerve
chondroitin-sulfate proteoglycan after brain growth factor-TrkA signaling and the relief of
injury. J Neurosci 14, 47164730. pain. Anesthesiology 115, 189204.
Li, J., and Lepski, G. (2013). Cell transplantation for Massa, S.M., Yang, T., Xie, Y., Shi, J., Bilgen, M., Joyce,
spinal cord injury:Asystematic review. Biomed J.N., Nehama, D., Rajadas, J., and Longo, F.M.
Res Int 2013, 786475. (2010). Small molecule BDNF mimetics activate
Li, X.B., Liao, G.S., Huang, S.M., Shu, Y.Y., and Tang, TrkB signaling and prevent neuronal degenera-
S.X. (2000). NGF-Tf conjugate prevents degen- tion in rodents. J Clin Invest 120, 17741785.
eration of substantia nigra neurons in a mouse McKeon, R.J., Jurynec, M.J., and Buck, C.R. (1999).
model of Parkinsons disease. Sheng Wu Hua The chondroitin sulfate proteoglycans neurocan
Neurotrophic Regulation in Neurorestoration of theBrain 393

and phosphacan are expressed by reactive astro- and cross-talk with mitogenic signalling. EMBO
cytes in the chronic CNS glial scar. J Neurosci 19, J 17, 732742.
1077810788. Murer, M.G., Yan, Q., and Raisman-Vozari, R. (2001).
Menon, D.K. (1999). Cerebral protection in severe Brain-derived neurotrophic factor in the con-
brain injury:Physiological determinants of out- trol human brain, and in Alzheimers disease and
come and their optimisation. Br Med Bull 55, Parkinsons disease. Progress Neurobiol 63, 71124.
226258. Murray, K.D., Gall, C.M., Jones, E.G., and
Middlemiss, P.J., Glasky, A.J., Rathbone, M.P., Isackson, P.J. (1994). Differential regulation of
Werstuik, E., Hindley, S., and Gysbers, J. (1995a). brain-derived neurotrophic factor and type II
AIT-082, a unique purine derivative, enhances calcium/calmodulin-dependent protein kinase
nerve growth factor mediated neurite outgrowth messenger RNA expression in Alzheimers dis-
from PC12 cells. Neurosci Lett 199, 131134. ease. Neuroscience 60,3748.
Middlemiss, P.J., Gysbers, J.W., and Rathbone, M.P. Myers, M.G., Jr., and White, M.F. (1996). Insulin sig-
(1995b). Extracellular guanosine and guanosine- nal transduction and the IRS proteins. Annu Rev
5-triphosphate increase: NGF synthesis and Pharmacol Toxicol 36, 615658.
release from cultured mouse neopallial astro- Nagahara, A.H., and Tuszynski, M.H. (2011).
cytes. Brain Res 677, 152156. Potential therapeutic uses of BDNF in neuro-
Middleton, G., Hamanoue, M., Enokido, Y., Wyatt, logical and psychiatric disorders. Nat Rev Drug
S., Pennica, D., Jaffray, E., Hay, R.T., and Davies, Discov 10, 209219.
A.M. (2000). Cytokine-induced nuclear fac- Noureddini, M., Verdi, J., Mortazavi-Tabatabaei, S.A.,
tor kappa B activation promotes the survival of Sharif, S., Azimi, A., Keyhanvar, P., and
developing neurons. J Cell Biol 148, 325332. Shoae-Hassani, A. (2012). Human endometrial
Mironova, Y.A., and Giger, R.J. (2013). Where no stem cell neurogenesis in response to NGF and
synapses go: Gatekeepers of circuit remodel- bFGF. Cell Biol Int 36, 961966.
ing and synaptic strength. Trends Neurosci 36, Olson, L., Backlund, E.O., Ebendal, T., Freedman,
363373. R., Hamberger, B., Hansson, P., Hoffer, B.,
Mobley, W.C. (1989). Nerve growth factor in Lindblom, U., Meyerson, B., Stromberg, I., etal.
Alzheimers disease: To treat or not to treat? (1991). Intraputaminal infusion of nerve growth
Neurobiol Aging 10, 578580; discussion 588590. factor to support adrenal medullary autografts in
Moretto, M.B., Boff, B., Lavinsky, D., Netto, C.A., Parkinsons disease. One-year follow-up of first
Rocha, J.B., Souza, D.O., and Wofchuk, S.T. clinical trial. Arch Neurol 48, 373381.
(2009). Importance of schedule of adminis- Olson, L., Nordberg, A., von Holst, H., Backman, L.,
tration in the therapeutic efficacy of guano- Ebendal, T., Alafuzoff, I., Amberla, K., Hartvig,
sine: Early intervention after injury enhances P., Herlitz, A., Lilja, A., et al. (1992). Nerve
glutamate uptake in model of hypoxia-ischemia. growth factor affects 11C-nicotine binding,
J Mol Neurosci 38, 216219. blood flow, EEG, and verbal episodic memory
Morrison, R.S., Sharma, A., de Vellis, J., and in an Alzheimer patient (case report). J Neural
Bradshaw, R.A. (1986). Basic fibroblast growth Transm Park Dis Dement Sect 4,7995.
factor supports the survival of cerebral cortical Ong, S.H., Hadari, Y.R., Gotoh, N., Guy, G.R.,
neurons in primary culture. Proc Natl Acad Sci Schlessinger, J., and Lax, I. (2001). Stimulation
USA 83, 75377541. of phosphatidylinositol 3-kinase by fibroblast
Mu, J.S., Li, W.P., Yao, Z.B., and Zhou, X.F. (1999). growth factor receptors is mediated by coordi-
Deprivation of endogenous brain-derived neu- nated recruitment of multiple docking proteins.
rotrophic factor results in impairment of spatial Proc Natl Acad Sci USA 98, 60746079.
learning and memory in adult rats. Brain Res Park, J., Al-Ramahi, I., Tan, Q., Mollema, N.,
835, 259265. Diaz-Garcia, J.R., Gallego-Flores, T., Lu, H.C.,
Mudo, G., Belluardo, N., Mauro, A., and Fuxe, Lagalwar, S., Duvick, L., Kang, H., etal. (2013).
K. (2007). Acute intermittent nicotine treat- RAS-MAPK-MSK1 pathway modulates ataxin 1
ment induces fibroblast growth factor-2 in the protein levels and toxicity in SCA1. Nature 498,
subventricular zone of the adult rat brain and 325331.
enhances neuronal precursor cell proliferation. Pasterkamp, R.J., and Verhaagen, J. (2006).
Neuroscience 145, 470483. Semaphorins in axon regeneration: Developmental
Muller, G., Storz, P., Bourteele, S., Doppler, H., guidance molecules gone wrong? Philos Trans R
Pfizenmaier, K., Mischak, H., Philipp, A., Kaiser, Soc Lond B Biol Sci 361, 14991511.
C., and Kolch, W. (1998). Regulation of Raf-1 Pedraza, C.E., Podlesniy, P., Vidal, N., Arevalo, J.C.,
kinase by TNF via its second messenger ceramide Lee, R., Hempstead, B., Ferrer, I., Iglesias, M.,
394 Part III:Homeostatic Manipulators

and Espinet, C. (2005). Pro-NGF isolated from guanosine. Nucleosides Nucleotides Nucleic
the human brain affected by Alzheimers dis- Acids 27, 666672.
ease induces neuronal apoptosis mediated by Rathbone, M.P., Middlemiss, P., Andrew, C.,
p75NTR. Am J Pathol 166, 533543. Caciagli, F., Ciccarelli, R., Di Iorio, P., and
Pencea, V., Bingaman, K.D., Wiegand, S.J., and Huang, R. (1998). The trophic effects of purines
Luskin, M.B. (2001). Infusion of brain-derived and purinergic signaling in pathologic reactions
neurotrophic factor into the lateral ventricle of of astrocytes. Alzheimer Dis Assoc Disord 12
the adult rat leads to new neurons in the paren- Suppl 2, S36S45.
chyma of the striatum, septum, thalamus, and Rathbone, M.P., Middlemiss, P.J., DeLuca, B., and
hypothalamus. J Neurosci 21, 67066717. Jovetich, M. (1991). Extracellular guanosine
Pezet, S., Malcangio, M., Lever, I.J., Perkinton, M.S., increases astrocyte cAMP:Inhibition by adenos-
Thompson, S.W., Williams, R.J., and McMahon, ine A2 antagonists. Neuroreport 2, 661664.
S.B. (2002). Noxious stimulation induces Trk Rathbone, M.P., Middlemiss, P.J., Gysbers, J.W.,
receptor and downstream ERK phosphorylation in Andrew, C., Herman, M.A., Reed, J.K., Ciccarelli,
spinal dorsal horn. Mol Cell Neurosci 21, 684695. R., Di Iorio, P., and Caciagli, F. (1999). Trophic
Phillips, H.S., Hains, J.M., Armanini, M., Laramee, effects of purines in neurons and glial cells. Prog
G.R., Johnson, S.A., and Winslow, J.W. (1991). Neurobiol 59, 663690.
BDNF mRNA is decreased in the hippocampus Rathbone, M.P., Middlemiss, P.J., Kim, J.K., Gysbers,
of individuals with Alzheimers disease. Neuron J.W., DeForge, S.P., Smith, R.W., and Hughes,
7, 695702. D.W. (1992). Adenosine and its nucleotides
Phillips, H.S., Hains, J.M., Laramee, G.R., Rosenthal, stimulate proliferation of chick astrocytes and
A., and Winslow, J.W. (1990). Widespread human astrocytoma cells. Neurosci Res 13,117.
expression of BDNF but not NT3 by target areas Rathbone, M.P., Saleh, T.M., Connell, B.J., Chang, R.,
of basal forebrain cholinergic neurons. Science Su, C., Worley, B., Kim, M., and Jiang, S. (2011).
250, 290294. Systemic administration of guanosine promotes
Podlesniy, P., Kichev, A., Pedraza, C., Saurat, J., functional and histological improvement following
Encinas, M., Perez, B., Ferrer, I., and Espinet, C. an ischemic stroke in rats. Brain Res 1407,7989.
(2006). Pro-NGF from Alzheimers disease and Ray, J., Peterson, D.A., Schinstine, M., and Gage,
normal human brain displays distinctive abili- F.H. (1993). Proliferation, differentiation, and
ties to induce processing and nuclear translo- long-term culture of primary hippocampal neu-
cation of intracellular domain of p75NTR and rons. Proc Natl Acad Sci USA 90, 36023606.
apoptosis. Am J Pathol 169, 119131. Recio-Pinto, E., Rechler, M.M., and Ishii, D.N.
Przybylski, M. (2009). A review of the current (1986). Effects of insulin, insulin-like growth
research on the role of bFGF and VEGF in angio- factor-II, and nerve growth factor on neurite for-
genesis. J Wound Care 18, 516519. mation and survival in cultured sympathetic and
Qian, M.D., Zhang, J., Tan, X.Y., Wood, A., Gill, D., sensory neurons. J Neurosci 6, 12111219.
and Cho, S. (2006). Novel agonist monoclonal Reichardt, L.F. (2006). Neurotrophin-regulated sig-
antibodies activate TrkB receptors and demon- nalling pathways. Philos Trans R Soc Lond B Biol
strate potent neurotrophic activities. J Neurosci Sci 361, 15451564.
26, 93949403. Reuss, B., and von Bohlen und Halbach, O. (2003).
Qiu, J., Cai, D., Dai, H., McAtee, M., Hoffman, P.N., Fibroblast growth factors and their receptors in
Bregman, B.S., and Filbin, M.T. (2002). Spinal the central nervous system. Cell Tissue Res 313,
axon regeneration induced by elevation of cyclic 139157.
AMP. Neuron 34, 895903. Rodriguez-Viciana, P., Warne, P.H., Dhand, R.,
Rai, K.S., Hattiangady, B., and Shetty, A.K. (2007). Vanhaesebroeck, B., Gout, I., Fry, M.J.,
Enhanced production and dendritic growth of Waterfield, M.D., and Downward, J. (1994).
new dentate granule cells in the middle-aged Phosphatidylinositol-3-OH kinase as a direct
hippocampus following intracerebroventricular target of Ras. Nature 370, 527532.
FGF-2 infusions. Eur J Neurosci 26, 17651779. Rothwell, N.J., and Strijbos, P.J. (1995). Cytokines
Raiker, S.J., Lee, H., Baldwin, K.T., Duan, Y., Shrager, in neurodegeneration and repair. Int J Dev
P., and Giger, R.J. (2010). Oligodendrocyte-myelin Neurosci 13, 179185.
glycoprotein and Nogo negatively regulate Rottlaender, A., Villwock, H., Addicks, K., and
activity-dependent synaptic plasticity. J Neurosci Kuerten, S. (2011). Neuroprotective role of fibro-
30, 1243212445. blast growth factor-2 in experimental autoim-
Rathbone, M., Pilutti, L., Caciagli, F., and Jiang, S. mune encephalomyelitis. Immunology 133,
(2008). Neurotrophic effects of extracellular 370378.
Neurotrophic Regulation in Neurorestoration of theBrain 395

Ruffini, F., Furlan, R., Poliani, P.L., Brambilla, E., Soares, F.A., Schmidt, A.P., Farina, M., Frizzo, M.E.,
Marconi, P.C., Bergami, A., Desina, G., Glorioso, Tavares, R.G., Portela, L.V., Lara, D.R., and
J.C., Comi, G., and Martino, G. (2001). Fibroblast Souza, D.O. (2004). Anticonvulsant effect of
growth factor-II gene therapy reverts the clini- GMP depends on its conversion to guanosine.
cal course and the pathological signs of chronic Brain Res 1005, 182186.
experimental autoimmune encephalomyelitis in Song, H., Ming, G., He, Z., Lehmann, M.,
C57BL/6 mice. Gene Ther 8, 12071213. McKerracher, L., Tessier-Lavigne, M., and Poo,
Rydel, R.E., and Greene, L.A. (1987). Acidic and basic M. (1998). Conversion of neuronal growth cone
fibroblast growth factors promote stable neurite responses from repulsion to attraction by cyclic
outgrowth and neuronal differentiation in cul- nucleotides. Science 281, 15151518.
tures of PC12 cells. J Neurosci 7, 36393653. Song, M.S., and Posse de Chaves, E.I. (2003).
Sandvig, A., Berry, M., Barrett, L.B., Butt, A., Inhibition of rat sympathetic neuron apoptosis
and Logan, A. (2004). Myelin-, reactive glia-, by ceramide. Role of p75NTR in ceramide gen-
and scar-derived CNS axon growth inhibi- eration. Neuropharmacology 45, 11301150.
tors: Expression, receptor signaling, and corre- Su, C., Elfeki, N., Ballerini, P., DAlimonte, I., Bau, C.,
lation with axon regeneration. Glia 46, 225251. Ciccarelli, R., Caciagli, F., Gabriele, J., and Jiang, S.
Schmidt, A.P., Avila, T.T., and Souza, D.O. (2005). (2009). Guanosine improves motor behavior,
Intracerebroventricular guanine-based purines reduces apoptosis, and stimulates neurogenesis in
protect against seizures induced by quinolinic rats with parkinsonism. J Neurosci Res 87, 617625.
acid in mice. Neurochem Res 30,6973. Su, C., Wang, P., Jiang, C., Ballerini, P., Caciagli, F.,
Schmidt, A.P., Lara, D.R., de Faria Maraschin, J., da Rathbone, M.P., and Jiang, S. (2013). Guanosine
Silveira Perla, A., and Onofre Souza, D. (2000). promotes proliferation of neural stem cells
Guanosine and GMP prevent seizures induced through cAMP-CREB pathway. J Biol Regul
by quinolinic acid in mice. Brain Res 864,4043. Homeost Agents 27, 673680.
Schmidt, A.P., Tort, A.B., Souza, D.O., and Lara, Suliman, S., Hemmings, S.M., and Seedat, S. (2013).
D.R. (2008). Guanosine and its modula- Brain-derived neurotrophic factor (BDNF)
tory effects on the glutamatergic system. Eur protein levels in anxiety disorders: Systematic
Neuropsychopharmacol 18, 620622. review and meta-regression analysis. Front
Schroer, S., and Adamson, J. (2011). Acupuncture for Integr Neurosci7,55.
depression:Acritique of the evidence base. CNS Sun, D., Bullock, M.R., McGinn, M.J., Zhou, Z.,
Neurosci Ther 17, 398410. Altememi, N., Hagood, S., Hamm, R., and
Schwab, M.E. (2004). Nogo and axon regeneration. Colello, R.J. (2009). Basic fibroblast growth
Curr Opin Neurobiol 14, 118124. factor-enhanced neurogenesis contributes to
Schwartz, J.P., and Mishler, K. (1990). Beta-adrenergic cognitive recovery in rats following traumatic
receptor regulation, through cyclic AMP, of nerve brain injury. Exp Neurol 216,5665.
growth factor expression in rat cortical and cere- Suyama, K., Shapiro, I., Guttman, M., and Hazan,
bellar astrocytes. Cell Mol Neurobiol 10, 447457. R.B. (2002). A signaling pathway leading to
Seifert, G., and Steinhauser, C. (2013). Neuron- metastasis is controlled by N-cadherin and the
astrocyte signaling and epilepsy. Exp Neurol FGF receptor. Cancer Cell 2, 301314.
244,410. Takahashi, M., Hayashi, S., Kakita, A., Wakabayashi, K.,
Seiger, A., Nordberg, A., von Holst, H., Backman, L., Fukuda, M., Kameyama, S., Tanaka, R.,
Ebendal, T., Alafuzoff, I., Amberla, K., Hartvig, Takahashi, H., and Nawa, H. (1999). Patients
P., Herlitz, A., Lilja, A., etal. (1993). Intracranial with temporal lobe epilepsy show an increase in
infusion of purified nerve growth factor to an brain-derived neurotrophic factor protein and its
Alzheimer patient:The first attempt of a possible correlation with neuropeptide Y. Brain Res 818,
future treatment strategy. Behav Brain Res 57, 579582.
255261. Tanaka, J., Horiike, Y., Matsuzaki, M., Miyazaki, T.,
Seo, J.H., Yu, J.H., Suh, H., Kim, M.S., and Cho, S.R. Ellis-Davies, G.C., and Kasai, H. (2008). Protein
(2013). Fibroblast growth factor-2 induced by synthesis and neurotrophin-dependent struc-
enriched environment enhances angiogenesis tural plasticity of single dendritic spines. Science
and motor function in chronic hypoxic-ischemic 319, 16831687.
brain injury. PLoS One 8, e74405. Tartaglia,N.,Du,J.,Tyler,W.J.,Neale,E.,Pozzo-Miller,L.,
Shults, C.W., Kimber, T., and Altar, C.A. (1995). and Lu, B. (2001). Protein synthesis-dependent
BDNF attenuates the effects of intrastriatal and -independent regulation of hippocampal syn-
injection of 6-hydroxydopamine. Neuroreport 6, apses by brain-derived neurotrophic factor. J Biol
11091112. Chem 276, 3758537593.
396 Part III:Homeostatic Manipulators

Tavares, R.G., Schmidt, A.P., Abud, J., Tasca, C.I., Thomas, A., and Cristalli, G. (2011). Evidence for
and Souza, D.O. (2005). In vivo quinolinic acid the existence of a specific g protein-coupled recep-
increases synaptosomal glutamate release in tor activated by guanosine. ChemMedChem 6,
rats:Reversal by guanosine. Neurochem Res 30, 10741080.
439444. Wagner, J.P., Black, I.B., and DiCicco-Bloom, E.
Terlau, H., and Seifert, W. (1990). Fibroblast growth (1999). Stimulation of neonatal and adult brain
factor enhances long-term potentiation in the neurogenesis by subcutaneous injection of
hippocampal slice. Eur J Neurosci 2, 973977. basic fibroblast growth factor. J Neurosci 19,
Thauerer, B., Zur Nedden, S., and Baier-Bitterlich, G. 60066016.
(2012). Purine nucleosides: endogenous neuro- Wang, Y.Q., Bian, G.L., Bai, Y., Cao, R., and
protectants in hypoxic brain. J Neurochem 121, Chen, L.W. (2008). Identification and kainic
329342. acid-induced up-regulation of low-affinity p75
Togari, A., Dickens, G., Kuzuya, H., and Guroff, G. neurotrophin receptor (p75NTR) in the nigral
(1985). The effect of fibroblast growth factor on dopamine neurons of adult rats. Neurochem Int
PC12 cells. J Neurosci 5, 307316. 53,5662.
Tomaselli, B., Podhraski, V., Heftberger, V., Bock, Whitaker, C.M., Beaumont, E., Wells, M.J.,
G., and Baier-Bitterlich, G. (2005). Purine Magnuson, D.S., Hetman, M., and Onifer, S.M.
nucleoside-mediated protection of chemical (2008). Rolipram attenuates acute oligodendro-
hypoxia-induced neuronal injuries involves cyte death in the adult rat ventrolateral funiculus
p42/44 MAPK activation. Neurochem Int 46, following contusive cervical spinal cord injury.
513521. Neurosci Lett 438, 200204.
Tonchev, A.B. (2011). Brain ischemia, neurogenesis, Wiliams, R.R., and Bunge, M.B. (2012). Schwann cell
and neurotrophic receptor expression in pri- transplantation:Arepair strategy for spinal cord
mates. Arch Ital Biol 149, 225231. injury? Prog Brain Res 201, 295312.
Troy, C.M., Friedman, J.E., and Friedman, W.J. Woodbury, M.E., and Ikezu, T. (2013). Fibroblast
(2002). Mechanisms of p75-mediated death of Growth factor-2 signaling in neurogenesis and
hippocampal neurons: Role of caspases. J Biol neurodegeneration. J Neuroimmune Pharmacol
Chem 277, 3429534302. 9, 92101.
Tuszynski, M.H., Thal, L., Pay, M., Salmon, D.P., Wu, K.Y., Hengst, U., Cox, L.J., Macosko, E.Z.,
U, H.S., Bakay, R., Patel, P., Blesch, A., Vahlsing, Jeromin, A., Urquhart, E.R., and Jaffrey, S.R.
H.L., Ho, G., et al. (2005). A phase 1 clinical (2005). Local translation of RhoA regulates
trial of nerve growth factor gene therapy for growth cone collapse. Nature 436, 10201024.
Alzheimer disease. Nat Med 11, 551555. Xu, X.M., Guenard, V., Kleitman, N., Aebischer, P.,
Tyler, W.J., and Pozzo-Miller, L. (2003). Miniature and Bunge, M.B. (1995). A combination of BDNF
synaptic transmission and BDNF modulate den- and NT-3 promotes supraspinal axonal regenera-
dritic spine growth and form in rat CA1 neu- tion into Schwann cell grafts in adult rat thoracic
rones. J Physiol 553, 497509. spinal cord. Exp Neurol 134, 261272.
Ulrich, H., Abbracchio, M.P., and Burnstock, G. Yamada, H., Fredette, B., Shitara, K., Hagihara, K.,
(2012). Extrinsic purinergic regulation of neu- Miura, R., Ranscht, B., Stallcup, W.B., and
ral stem/progenitor cells: Implications for Yamaguchi, Y. (1997). The brain chondroitin
CNS development and repair. Stem Cell Rev 8, sulfate proteoglycan brevican associates with
755767. astrocytes ensheathing cerebellar glomeruli and
Unger, E.F., Goncalves, L., Epstein, S.E., Chew, E.Y., inhibits neurite outgrowth from granule neu-
Trapnell, C.B., Cannon, R.O., 3rd, and Quyyumi, rons. J Neurosci 17, 77847795.
A.A. (2000). Effects of a single intracoronary Yamashita, T., Tucker, K.L., and Barde, Y.A. (1999).
injection of basic fibroblast growth factor in sta- Neurotrophin binding to the p75 receptor modu-
ble angina pectoris. Am J Cardiol 85, 14141419. lates Rho activity and axonal outgrowth. Neuron
Volosin, M., Trotter, C., Cragnolini, A., Kenchappa, 24, 585593.
R.S., Light, M., Hempstead, B.L., Carter, B.D., Yanagisawa-Miwa, A., Uchida, Y., Nakamura, F.,
and Friedman, W.J. (2008). Induction of proneu- Tomaru, T., Kido, H., Kamijo, T., Sugimoto, T.,
rotrophins and activation of p75NTR-mediated Kaji, K., Utsuyama, M., Kurashima, C., et al.
apoptosis via neurotrophin receptor-interacting (1992). Salvage of infarcted myocardium by
factor in hippocampal neurons after seizures. J angiogenic action of basic fibroblast growth fac-
Neurosci 28, 98709879. tor. Science 257, 14011403.
Volpini, R., Marucci, G., Buccioni, M., Dal Ben, D., Yang, H., Xia, Y., Lu, S.Q., Soong, T.W., and Feng, Z.W.
Lambertucci, C., Lammi, C., Mishra, R.C., (2008). Basic fibroblast growth factor-induced
Neurotrophic Regulation in Neurorestoration of theBrain 397

neuronal differentiation of mouse bone mar- Zheng, W., Nowakowski, R.S., and Vaccarino, F.M.
row stromal cells requires FGFR-1, MAPK/ERK, (2004). Fibroblast growth factor 2 is required
and transcription factor AP-1. J Biol Chem 283, for maintaining the neural stem cell pool in the
52875295. mouse brain subventricular zone. Dev Neurosci
Yang, P., and Yang, Z. (2012). Enhancing intrinsic 26, 181196.
growth capacity promotes adult CNS regenera- Zhu, W., Cheng, S., Xu, G., Ma, M., Zhou, Z., Liu,
tion. J Neurol Sci 312,16. D., and Liu, X. (2011). Intranasal nerve growth
Yano, H., and Chao, M.V. (2000). Neurotrophin recep- factor enhances striatal neurogenesis in adult
tor structure and interactions. Pharmaceutica rats with focal cerebral ischemia. Drug Deliv 18,
acta Helvetiae 74, 253260. 338343.
Yazaki, N., Hosoi, Y., Kawabata, K., Miyake, A., Zhuang, Z., Zhao, X., Wu, Y., Huang, R., Zhu, L.,
Minami, M., Satoh, M., Ohta, M., Kawasaki, T., Zhang, Y., and Shi, J. (2011). The anti-apoptotic
and Itoh, N. (1994). Differential expression pat- effect of PI3K-Akt signaling pathway after sub-
terns of mRNAs for members of the fibroblast arachnoid hemorrhage in rats. Ann Clin Lab Sci
growth factor receptor family, FGFR-1-FGFR-4, 41, 364372.
in rat brain. J Neurosci Res 37, 445452. Zigova, T., Pencea, V., Wiegand, S.J., and Luskin,
Yoshimoto, Y., Lin, Q., Collier, T.J., Frim, D.M., M.B. (1998). Intraventricular administration of
Breakefield, X.O., and Bohn, M.C. (1995). BDNF increases the number of newly generated
Astrocytes retrovirally transduced with BDNF neurons in the adult olfactory bulb. Mol Cell
elicit behavioral improvement in a rat model of Neurosci 11, 234245.
Parkinsons disease. Brain Res 691,2536. Zuccato, C., Ciammola, A., Rigamonti, D., Leavitt,
Zechel, S., Werner, S., Unsicker, K., and von Bohlen B.R., Goffredo, D., Conti, L., MacDonald, M.E.,
und Halbach, O. (2010). Expression and func- Friedlander, R.M., Silani, V., Hayden, M.R., etal.
tions of fibroblast growth factor 2 (FGF-2) in (2001). Loss of huntingtin-mediated BDNF gene
hippocampal formation. Neuroscientist 16, transcription in Huntingtons disease. Science
357373. 293, 493498.
PARTIV

Homeostatic Therapies
for Disease and Dysfunction
23
Epilepsy
DETLEVBOISON

INTRODUCTION has become clear that comorbidities occur


Epilepsy is one of the most frequent neurological early in the disease and frequently precede the
conditions, affecting about 1% of the population emergence of epileptic seizures, often by years
or 70 million persons worldwide. It is a het- (Kanner, 2012; Rudzinski and Meador, 2013). It
erogeneous syndrome characterized by spon- is tempting to speculate that epileptic seizures
taneous recurrent seizures, which can be brief are merely one more symptom of a complex syn-
periods of altered consciousness or absences drome in which major homeostatic functions of
but can also generalize and involve motor func- the brain are disrupted.
tions leading to convulsions. Whereas seizures
are generated by excessive electrical discharges DISRUPTION OFNET WORK
in neuronal clusters, the cellular and molecular H O M E O S TA S I S I N E P I L E P S Y
basis of epilepsy is still largely unknown. It is Current epilepsy treatment is based on the pre-
now widely accepted that epilepsy is far more sumption that neuronal dysfunction is the
complex than merely a disruption of the balance primary cause of epilepsy. However, current
between neuronal excitation and inhibition, antiepileptic drugs (AEDs) that have neuronal
and in particular the contribution of inflam- targets and have been designed to suppress sei-
matory, glial, bioenergetic, and epigenetic zures as the major symptom found in epilepsy
changes to the pathophysiology of epilepsy has are ineffective in about one-third of all persons
gained much attention recently (Boison et al., with epilepsy; importantly, those drugs (i) fail
2011; Pan et al., 2005; Qureshi and Mehler, to treat the comorbidities of epilepsy and (ii)
2010; Ravizza et al., 2008; Seifert et al., 2010; fail to affect the underlying pathogenetic pro-
Vezzani et al., 2008; Williams-Karnesky et al., cesses (Brooks-Kayal et al., 2013; Sorensen and
2013; Williamson et al., 2005). Most forms of Kokaia, 2013). There is a substantial amount of
epilepsy are triggered by precipitating injuries, evidence implicating nonneuronal mechanisms
which lead to blood-brain barrier (BBB) disrup- as a major contributing factor to the develop-
tion, inflammatory responses, and microglial ment of epilepsy (i.e., epileptogenesis). Among
and macroglial activation. In particular, astro- those, inflammatory and immunomodulatory
gliosis and disruption of key astrocyte depend- processes, as well as the integrity of the BBB,
ent homeostatic functions such as aquaporin are involved, and glial dysfunction is therefore
expression, ion buffering, and adenosine equi- a major player in the disruption of homeostatic
librium play important roles in epileptogenesis. functions in epilepsy (Figure 23.1; Boison, 2012b;
Those mechanisms certainly play a role in post- Coulter and Eid, 2012; Steinhauser et al., 2012).
traumatic epileptogenesis, where disruption of The identification of nonneuronal mechanisms
the BBB and astrogliosis are significant factors. contributing to the syndrome of epilepsy offers
Consequently, epilepsy presents as a disorder of new hopes to treat not only the hallmark seizures
complex network dysfunction, which is not only of epilepsy but also the accompanying comor-
characterized by recurrent seizures but also by bidities and to halt those processes that lead to
comorbidities ranging from sleep dysfunction the development of epilepsy. The next sections
to cognitive impairment, depression, and psychi- describe key homeostatic functions that are dis-
atric problems (Hermann etal., 2008; LaFrance rupted in clinical epilepsy as well as in animal
et al., 2008; Lin et al., 2012). Intriguingly, it models of the disease.
FIGURE23.1: Disruption of network homeostasis in epilepsy. Astrocytes play a key role in the mainte-
nance of network homeostasis in the brain. Astrocytic endfeet couple to the vasculature, whereas distinct processes
contribute to the tripartite synapse. Through neurovascular coupling, astrocytes can directly link vascular signals
to neuronal activity, whereas neuronal activity can directly influence vascular responses. A selection of major
homeostatic regulators is shown, with pathological alterations in epilepsy shown in red. Water and ion homeosta-
sis depend largely on the water channel aquaporin 4 (AQ4, in pink), the potassium channel Kir4.1 (in green), the
sodium potassium ATPase (in orange), and the sodium potassium chloride cotransporter (in violet). In epilepsy,
delocalization of AQ4 impairs potassium uptake through Kir4.1, contributing to a rise in synaptic K+. Astrocytes
control synaptic glutamate via reuptake through the glutamate transporter GLT-1 (in blue), through intracellular
conversion of glutamate into glutamine via glutamine synthetase (GS, in blue), and through the Ca 2+-dependent
release of glutamate as gliotransmitter. In epilepsy, decreased GLT-1 dependent glutamate uptake, decreased met-
abolic clearance through GS, and increased astroglial release of glutamate all contribute to an increase in synaptic
glutamate. Astrogliosis and associated overexpression of adenosine kinase (ADK, in red) in the epileptic brain
lead to increased metabolic clearance of adenosine (ADO) and resulting adenosine deficiency. Disruption of the
blood-brain barrier leads to the extravasation of albumin and TGF, which trigger astrogliosis and compromise
the buffering capacity of astrocytes. Growth factors such as brain derived neurotrophic factor (BDNF) lead to the
activation of transcriptional programs in neurons and via transactivation of the TrkB receptor (in green) with the
adenosine A 2AR (in orange) can trigger astrogliosis and overexpression of ADK. Inflammatory mediators, such as
interleukin 1 (IL-1) or tumor necrosis factor alpha (TNF) can compromise GLT-1-dependent glutamate uptake
and can trigger the astroglial release of glutamate. Mitochondria play a major role in the regulation of ATP and
may release reactive oxygen species (ROS) in epilepsy. See the text for more details.
Epilepsy 403

Ion and Water Homeostasis of more than 80% of synaptic glutamate and
In the brain, the homeostasis of water and ions therefore play an essential role in terminat-
is intricately linked and controlled by channels, ing excitatory neurotransmission (Anderson
which are permeable to water and/or ions, such and Swanson, 2000). The homeostasis of gluta-
as K+, Na+, Ca2+, and Cl. Apart from tightly con- mate, but also of its metabolic product GABA,
trolled ion fluxes through neuronal membranes, depends on the glutamate-glutamine cycle in
glial cells exert a major control over water and ion which astrocytes convert the transported gluta-
homeostasis (Seifert etal., 2010; Steinhauser etal., mate into glutamine via glutamine synthetase.
2012). Remarkably, a major part of the energy Glutamine is transported back into neurons,
expenditure of the brain is devoted to the con- where it is transformed back into glutamate
trol of ion fluxes. Aquaporin 4 (AQP4) is a glial (or into GABA following decarboxylation of
water transport channel, which is altered as a glutamate). Any disruption of astrocyte func-
consequence of brain injury (Guo etal., 2006)and tion in epilepsy is therefore expected to affect
directly implicated in epileptogenesis (Binder glial glutamate homeostasis. Virus-induced
etal., 2012). The channel is normally localized to gliosis was recently used to trigger a reduc-
both the perivascular endfeet and within peri- tion in the expression of astroglial glutamine
synaptic processes of astrocytes, where it per- synthetase; as a consequence, adjacent neu-
mits the bidirectional exchange of water between rons displayed a specific deficit in inhibitory
the extracellular space and the blood (Nagelhus synaptic function, however, in the absence of
etal., 2004; Nielsen etal., 1997; Rash etal., 1998). changes in excitatory function (Ortinski et al.,
However, AQP4 redistributed primarily to peri- 2010). Those non-cell-autonomous deficits in
synaptic processes in human mesial temporal lobe inhibition were mimicked by blockade of the
epilepsy and therefore might be a contributing glutamate-glutamine cycle and reversed by
factor of hyperexcitability through dysregulation exogenous glutamine. This study directly dem-
of water and K+ homeostasis (Eid etal., 2005). In onstrated that astrogliosis per se could alter
support of this view, AQP4 knockout mice have neuronal excitability via disruption of astroglial
an increased volume of extracellular space and are glutamate homeostasis. In line with those find-
less susceptible to pentylenetetrazole-induced sei- ings, epileptogenic tissue resected from patients
zures (Binder et al., 2004a; Binder et al., 2004b). with temporal lobe epilepsy (TLE) was charac-
Glial water flow is tightly linked to K+ transport terized by a significant reduction in glutamine
via the inward rectifying K+ channel, Kir4.1, which synthetase as a likely explanation for increased
colocalizes with AQP4 on the astrocyte membrane extracellular glutamate in TLE (Eid etal., 2004).
(Hsu etal., 2011). Likewise, elevation of extracel- Glutamine synthetase deficiency might be a pos-
lular K+ to millimolar concentrations influences sible cause for TLE since the sustained microin-
neuronal excitability and exacerbates epileptiform fusion of a glutamine synthetase inhibitor into
activity (Feng and Durand, 2006). The importance the hippocampus of rats triggered recurrent
of the maintenance of K+ clearance mechanisms is seizures that continued for several weeks and
indicated by findings that polymorphisms in the resulted in neuropathological features reminis-
gene for Kir4.1 are associated with human epi- cent of TLE (Eid et al., 2008). Thus astrocytes
lepsy and that a glial specific deletion of Kir4.1 in directly control the homeostasis of critical neu-
mice reduced K+ clearance from the synaptic cleft; rotransmitters via specific uptake and intracel-
deficiencies in the homeostasis of K+ clearance lular metabolic mechanisms. However, glial cells
therefore constitute a feasible mechanism for the can also directly affect the homeostasis of neuro-
seizure phenotype in human patients (Haj-Yasein transmitters and neuromodulators through gli-
et al., 2011; Heinemann et al., 2000). Together otransmission, as evaluated in the next section.
these findings suggest an intricate interplay
between water and potassium homeostasis and a Glial Dysfunction and
direct involvement of water and ion homeostasis Gliotransmitter Homeostasis
in the pathogenesis of epilepsy. Hippocampal sclerosis is a characteristic patho-
logical hallmark of TLE (Malmgren and Thom,
Glutamate and Glutamine 2012), and surgical resections of epileptogenic
Homeostasis brain tissue have demonstrated that onset
Glutamate transporters located in astrocytes zones for chronic temporal lobederived and
are responsible for the reuptake and clearance posttraumatic seizures correlate with gliotic
404 Part IV:Homeostatic Therapies

scarring. Astrocytes influence the pathogenesis Adenosine Homeostasis


and pathophysiology of epilepsy by the homeo- Epileptic seizures trigger an increase in extracel-
static control of synaptic transmission via the lular adenosine, and it is this seizure-induced
release of gliotransmitters such as glutamate, increase in adenosine that normally terminates
adenosine 5-triphosphate (ATP), and d-serine a seizure and leads to postictal brain shutdown;
(Haydon and Carmignoto, 2006) in addition consequently, adenosine is an endogenous anti-
to the reuptake of neurotransmitters such as convulsant of the brain (Boison, 2012b). It affects
glutamate (Coulter and Eid, 2012) or neuro- neuronal excitability via pre- and postsynaptic
modulators such as adenosine (Boison, 2012b). adenosine receptors, which contribute to the
An astrocytic basis of epilepsy was proposed control of neuronal Ca2+ and K+ fluxes; in addi-
based on findings suggesting that prolonged tion, it provides important homeostatic control
episodes of neuronal depolarization evoked by over mitochondrial bioenergetics and epige-
the astrocytic release of glutamate contributes netic functions through interference with DNA
to epileptiform discharges (Tian et al., 2005). methylation (Boison, 2013; Masino et al., 2011;
Astrocytes play important upstream homeo- Williams-Karnesky et al., 2013). Adenosine
static roles in controlling uptake, degradation, prevents seizures via activation of adenosine A1
and recycling of neurotransmitters. In addi- receptors, which couple to inhibitory G proteins.
tion, glial dysfunction in the BBB, as well as Deletion of the A1R in mice causes lethal status
neuroimmunological functions governed by epilepticus (SE) following traumatic brain injury
glia, have been implicated not only in seizure or exposure to an excitotoxin (Fedele etal., 2006;
generation (i.e., ictogenesis) but most impor- Kochanek etal., 2006), whereas therapeutic aden-
tantly in the pathophysiological processes that osine augmentation effectively stops seizures
lead to the development of epilepsy (i.e., epi- (Boison, 2012a). A crucial role for A1 receptors
leptogenesis). Because glia communicate with in seizure control are supported by clinical find-
each other and assume a role that is upstream of ings of A1R variants that have been associated
neuronal function, perturbations of glial home- with the development of posttraumatic epilepsy
ostasis can affect entire neuronal networks. (Wagner et al., 2010). Synaptic levels of adeno-
Those network effects of glia might indeed be a sine are largely controlled by metabolic clearance
reason why neuronal networks in epilepsy syn- through the astrocyte-based enzyme adenosine
chronize; similarly, fluctuations in homeostatic kinase (ADK), which is a phosphotransferase
functions of glia might explain why seizures are converting adenosine into adenosine monophos-
sporadic. phate (AMP; Boison, 2013; Figure 23.2). ADK is

FIGURE 23.2: Adenosine homeostasis:functions of adenosine and therapeutic adenosine aug-


mentation. Red: ATP is the major source of adenosine, whereas the metabolic clearance of adenosine is largely
mediated by adenosine kinase, which in the adult brain is primarily located in astrocytes. Green:Adenosine exerts
neuroprotection and seizure suppression via the activation of neuronal A1Rs; in addition adenosine has procogni-
tive and antipsychotic properties via activation of neuronal A 2ARs. Blue: Adenosine plays an epigenetic role via
the regulation of DNA methylation, a mechanism that is antiepileptogenic. Violet: Several strategies have been
developed to therapeutically augment adenosine signaling in the brain. Those include systemic ADK inhibitors,
systemic A1R agonists, stem cell therapy, and gene therapy.
Epilepsy 405

upregulated and causes adenosine deficiency in the initial trigger for BBB dysfunction, which
epileptogenic sclerotic tissue in a variety of rodent became apparent as soon as one day after the
models of epilepsy, as well as in human specimens event. Thus BBB disruption as evaluated by MRI
resected from patients with TLE and hippocam- might constitute a valuable biomarker and target
pal sclerosis (Aronica etal., 2011; Li etal., 2008). for the prediction and prevention of epileptogen-
Acausal role for overexpressed ADK in epilepsy esis following an insult to thebrain.
is suggested by findings that transgenic or viral
overexpression of ADK was sufficient to trigger Growth Factors
recurrent electrographic seizures in mice (Li Epileptic seizures can induce wide-ranging
etal., 2008; Shen etal., 2014). In addition, homeo- changes in growth factors, neurotrophins, and
static functions of the adenosine system appear to transcription factors (Grabenstatter etal., 2012).
play a crucial role in epileptogenesis. Both trans- Akey role in epileptogenesis has been ascribed to
genic animals with forebrain-selective reduction brain-derived neurotrophic factor (BDNF) and
of ADK and recipients of adenosine-releasing its receptor TrkB (Grabenstatter etal., 2012). In
cells or devices showed a significant attenuation neurons, BDNF activates the JAK/STAT pathway,
of the development of epilepsy (Li et al., 2008; cAMP response element binding protein, induc-
Williams-Karnesky et al., 2013). These findings ible cAMP early repressor, and early growth
indicate that astroglial ADK is a promising target response factors that induce a shift in the expres-
for the prediction and prevention of seizures in sion of specific subunits of the GABA AR, as well
epilepsy. as the expression levels of N-methyl-D-aspartate
receptors (Kim et al., 2012; Lund et al., 2008;
Blood-Brain Barrier Roberts et al., 2006). In astrocytes, activation
A breakdown of the BBB has been intricately of TrkB has been linked to the development of
linked to epileptogenesis (Heinemann et al., astrogliosis, a mechanism that is also influenced
2012)and plays an instrumental role in the devel- by transactivation of the TrkB receptor through
opment of pharmacoresistance in chronic epi- the adenosine A2AR (Brambilla et al., 2003).
lepsy (Loscher and Potschka, 2002). Clinical data Therefore, an injury- or seizure-induced surge
document BBB disruption in patients with post- in adenosine (Clark et al., 1997) could trigger
traumatic epilepsy (Tomkins et al., 2008) and and potentiate pathogenetic astroglial changes
suggest an epileptogenic role of BBB disruption through increased TrkB signaling. Growth factor
in patients with brain tumors (Marchi et al., dependent mechanisms that contribute to epilep-
2007). BBB disruption has been associated with togenesis via the disruption of glial homeostatic
the extravasation of albumin and transforming functions are a new area of research that requires
growth factor beta (TGF-) signaling (Cacheaux increased attention.
etal., 2009; Ivens etal., 2007), which contributes
to the transformation of astrocytes into their Immunomodulatory and
reactive form, a process that also involves the Inflammatory Responses
initiation of a transcriptional program leading Immune and inflammatory processes play cru-
to further astrocyte activation, inflammation, cial roles in the development of epilepsy, and
and reduced clearance capacity for glutamate anti-inflammatory therapies might have antie-
and K+ (Cacheaux et al., 2009; David et al., pileptogenic properties (Aronica et al., 2012).
2009). The reduced buffering capacity of trans- Importantly, components of the proinflamma-
formed astrocytes for glutamate and K+ appears tory interleukin-1/Toll-like receptor (IL-1R/
to be most critical during repetitive activa- TLR) signaling pathway are overexpressed in
tion. Experimental blockade of TGF- signal- surgically resected specimen from a human with
ing following BBB disruption decreased those TLE (Ravizza etal., 2008), whereas experimental
transcriptional responses and prevented epi- activation of the IL-1R/TLR pathway has been
leptogenesis (Cacheaux etal., 2009; David etal., linked to the precipitation and recurrence of
2009). More recently, BBB disruption following seizures (Vezzani et al., 2011a). In addition, the
kainic acidinduced epileptogenesis in rats was activation of the IL-1R/TLR pathway by endog-
monitored longitudinally via MRI. Those MRI enous ligands or by viral or bacterial mimetics
data showed BBB leakage at 1 day and 6 weeks can increase excitability of the brain by inducing
after SE in the hippocampus, entorhinal cortex, rapid posttranslational changes in voltage- and
amygdala, and piriform cortex (van Vliet et al., ligand-gated ion channels. Among the endog-
2014). In this case kainic acidinduced SE was enous ligands are proinflammatory cytokines,
406 Part IV:Homeostatic Therapies

such as interleukin-1 (IL-1), or danger signals, from seizures or injuries to the brain but also
such as High Mobility Group Box 1 (HMGB1), contributing to epileptogenesis (Waldbaum and
which can be released from injured or activated Patel, 2010). Mitochondria can produce reactive
cells (Vezzani etal., 2011b). HMGB1 is the endog- oxygen species and therefore are uniquely vul-
enous ligand of TLR4 and normally bound to nerable to oxidative stress, which can severely
chromatin. However, following either cell dam- affect the production of ATP, the stability of mito-
age or neuronal hyperexcitability, this factor chondrial DNA, and synaptic glutamate homeo-
can be released into the extracellular space. The stasis, which can all affect neuronal excitability
proepileptogenic role of HMGB1 is supported by and seizure susceptibility. Therefore, improving
recent data showing that blockade of the TLR4 mitochondrial bioenergetics may offer the dual
pathway significantly delays the onset of seizures benefits of restoring metabolic dysfunction and
(Maroso etal., 2010). Likewise, engineered mice adenosine homeostasis and is a potential strategy
with defects in IL-1R/TLR signaling are intrin- to affect not only seizures but also the pathoge-
sically resistant to seizures. Together, inflamma- netic processes involved in the development of
tory processes and disruption of the BBB might epilepsy.
support chronic hyperexcitability of the brain via
compromised homeostatic functions dependent Epigenetics
on glial mechanisms on multiple levels. The methylation hypothesis of epileptogenesis
suggests that seizures by themselves can induce
Mitochondrial Bioenergetics epigenetic chromatin modifications and thereby
Mitochondria are characterized by important aggravate the epileptogenic condition (Kobow
homeostatic functions that influence neuronal and Blumcke, 2011). Epigenetic modifications,
excitability, including production of ATP, fatty which alter gene transcription without modify-
acid oxidation, control of apoptosis and necrosis, ing the underlying DNA sequence, are highly
regulation of amino acid cycling, neurotransmit- plastic and can respond rapidly in response to
ter biosynthesis, and regulation of cytosolic Ca2+ environmental cues, an important endogenous
homeostasis. Mitochondrial oxidative phospho- mechanism for temporally and spatially control-
rylation is the main pathway for the generation ling gene expression. Changes in histone acetyla-
of ATP, which in turn is intricately related to tion and methylation, as well as changes in DNA
adenosine. Uncoupling of mitochondrial oxida- methylation, once thought to occur only in divid-
tive phosphorylation leads to an increase in aden- ing cells, have been shown to also occur in mature
osine, which is independent of ATP (Doolette, cells in the central nervous system (Feng et al.,
1997). Energy depletion in general is marked 2010; Ma et al., 2009). Tellingly, these changes
by the accumulation of AMP, a direct precur- occur regularly and rapidly. Methylation of
sor of adenosine. At the same time, increased DNA in the central nervous system has attracted
AMP signals an impending energy crisis and increasing attention recently, with new research
activates the serine/threonine AMP-activated showing activity-induced proliferation of neural
protein kinase (AMPK; Zong etal., 2002), which precursor cells via active DNA demethylation
in turn interrupts ATP-consuming reactions, (Ma et al., 2009). Altered DNA methylation in
activates ATP-generating pathways (Hardie, the brain has also been implicated in psychiatric
2004), and promotes mitochondrial biogenesis and neurological conditions, including epilepsy
(Reznick and Shulman, 2006). Therefore enhanc- (Kobow etal., 2009; Ma etal., 2009). DNA meth-
ers of mitochondrial biogenesis or other strate- ylation requires the donation of a methyl group
gies that enhance ATP increase adenosine via from S-adenosylmethionine, a process that is
increased availability of the adenosine precursor facilitated by DNA methyltransferase enzymes.
ATP (Masino and Geiger, 2008). Based on those The resulting product, S-adenosylhomocysteine
mechanisms, it can be hypothesized that when- (SAH) is then further converted into adenosine
ever mitochondrial function is altered (in both and homocysteine by SAH hydrolase. Critically,
directions), an increased production of adeno- the equilibrium constant of the SAH hydrolase
sine can be expected. Certain inherited epilepsies enzyme lies in the direction of SAH forma-
are associated with mitochondrial dysfunction; tion (Kredich and Martin, 1977); therefore, the
however, little is known about its role in acquired reaction will proceed only when adenosine and
epilepsies such as TLE. Recent lines of evidence homocysteine are constantly removed (Boison
suggest that mitochondrial oxidative stress and et al., 2002; Kredich and Martin, 1977). In
dysfunction are key factors not only resulting the adult brain, removal of adenosine occurs
Epilepsy 407

Epileptogenic condition Adenosine therapy


S-adenosylmethionine
DNMT DNMT
Methylated DNA Methylated DNA

S-adenosylhomocysteine S-adenosylhomocysteine
Epileptogenesis
Homocysteine Antiepileptogenesis
Adenosine (ADO) (HCY)
Adenosine (ADO)
Adenosine kinase (ADK)
Seizures AMP Seizure suppression

FIGURE23.3: Epigenetic role of adenosine. Red:Adenosine is an obligatory end product of transmethyla-


tion reactions. If adenosine is not constantly removed by adenosine kinase (ADK), its levels increase and block
DNA methylation. In the chronic epileptic brain, ADK is increased, and this increase in the metabolic clearance of
adenosine is thought to drive DNA methylation. Correspondingly, the chronically epileptic brain is characterized
by hypermethylated DNA. DNA hypermethylation and reduced adenosine are both thought to drive epileptogen-
esis and disease progression in epilepsy. Green:Adenosine augmentation therapy restores adenosine and thereby
not only exerts seizure suppression but also blocks DNA methylation, leading to a reversal of the hypermethylated
state found in chronic epilepsy. Through this epigenetic mechanism, adenosine augmentation therapy is thought
to prevent epileptogenesis. Blue:Reactions dependent on DNA methyltransferases (DNMT).

largely via the astrocyte-based enzyme ADK, different levels. The synergistic modulation of
which is overexpressed in epilepsy (Boison, multiple known and unknown pathways may
2013; Fredholm, 2012; Pignataro et al., 2008). offer new hope to treat epileptic seizures as well
If metabolic clearance of adenosine through as associated comorbidities comprehensively
ADK is impaired, SAH levels rise (Boison etal., and to directly prevent those processes that lead
2002). SAH in turn is known to inhibit DNA to the development of epilepsy and an aggrava-
methyltransferases through product inhibition tion and progression of the epileptic phenotype.
(James et al., 2002). Since disruption of adeno- Interestingly, older AEDs such as valproate, car-
sine homeostasis and adenosine deficiency has bamazepine, and phenytoin, which are relatively
been implicated in epileptogenesis, local thera- unspecific and may act on different targets and
peutic adenosine augmentation is an effective mechanisms, are still the clinical mainstay for
strategy to acutely suppress seizures in modeled epilepsy therapy, whereas newer AEDs, which are
epilepsy (Boison, 2009). In addition, possible epi- cleaner and more specific to better character-
genetic effects of adenosine augmentation in the ized mechanisms and pathways, did not lead to a
treatment of epilepsy, including the potential to major overall improvement in the therapy of the
modulate DNA methylation status, have recently epilepsies (Vajda, 2007). The following sections
been investigated in our laboratory, making use report recent progress in severalareas.
of adenosine-releasing silk-based brain implants,
which provided long-term suppression of epilep- Restoration ofGlutamate
togenesis (Figure 23.3; see also next chapter and Homeostasis
Williams-Karnesky etal.,2013). Although few studies have directly addressed
glutamatergic therapies in animal models of
Reconstruction ofNetwork epilepsy, four strategies might have the potential
Homeostasis inthe Therapy to reconstruct glutamate homeostasis in epilepsy.
ofEpilepsy First, -lactam antibiotics increase glutamate
The mechanisms discussed in the preceding sec- transporter expression on astrocytes (GLT-1)
tions offer novel opportunities for the treatment in vivo and thereby increase the clearance rate
and prevention of epilepsy via the therapeutic of glutamate from the synaptic cleft (Rothstein
manipulation of homeostatic control mecha- et al., 2005). In an in vitro study, the -lactam
nisms. Those approaches differ from conventional antibiotic ceftriaxone (10M for 2days) increased
single-target or defined-target symptomatic glutamate uptake in primary human astrocytes
therapies by affecting entire networks on several through NFB-dependent activation of the
408 Part IV:Homeostatic Therapies

major astroglial glutamate reuptake transporter electrographic seizures (Boison, 2012b); conse-
excitatory amino acid transporter 2 (EAAT2; Lee quently, adenosine augmentation therapies are a
etal., 2008). Second, daily administration of lev- direct rational therapeutic approach and highly
etiracetam (50 mg/kg, i.p) for 20days beginning effective in suppressing and preventing seizures
one day after a traumatic brain injury triggered in rodent models of epilepsy (Figure 23.2; Boison,
by controlled cortical impact in rats reversed 2009). To avoid widespread systemic side effects of
the TBI-induced decrease in regional glutamate adenosine, local or brain-specific treatment strat-
transporter expression, indicating that at least egies are required. Four successful approaches
one AED has the potential to restore glutamate have been demonstrated: (i) Silk-based brain
homeostasis (Zou et al., 2013). Third, epilepti- implants engineered to release defined doses of
form activity was supported by the maintenance adenosine with known kinetics prevented kindled
of glutamine transport into neurons (Tani etal., seizures in the rat. Importantly, a transient dose
2010), whereas intact glutamine synthetase func- of adenosine delivered during kindling robustly
tion appears to be crucial for seizure prevention attenuated epilepsy development, indicating a
by driving the intracellular metabolic clearance potential antiepileptogenic effect of therapeutic
of extracellular glutamate (Ortinski etal., 2010). adenosine augmentation (Szybala et al., 2009).
Therapies aimed at reconstructing glutamine (ii) Stem cells engineered to release adenosine
synthetase function generally appear to have sci- were used as infrahippocampal cell grafts and
entific merit; however, the concept that reduced shown to prevent acute and induced seizures, to
glutamine synthetase expression and activity attenuate development of astrogliosis, to prevent
are required for seizure generation has recently overexpression of ADK, and to prevent develop-
been challenged based on findings suggesting ment of spontaneous recurrent epileptic seizures
normal expression levels but different distribu- in mice and rats (Li etal., 2008; Li etal., 2007).
tion of the enzyme (Papageorgiou et al., 2011). (iii) Agene therapy designed to selectively target
Fourth, astrocytes affect neuronal function by ADK in astrocytes based on RNA interference
the uptake and release of glutamate; decreased technology almost completely abrogated any
uptake or increased release of glutamate might spontaneous seizure activity in epileptic Adk-tg
contribute to the initiation of focal ictal dis- mice (Theofilas etal., 2011). (iv) Finally, a high-fat
charges (Gomez-Gonzalo et al., 2010; Halassa low-carbohydrate ketogenic diet (see later discus-
etal., 2010). New findings suggest that astroglial sion) suppressed seizures in mice via increased
A2ARs, through direct antagonistic interaction activation of adenosine A1 receptors (Masino
with the astroglial glutamate transporter GLT-1, et al., 2011). Based on the potent antiictogenic
play major roles in setting the tone of extracel- and antiepileptogenic properties, the translation
lular glutamate (Matos etal., 2012a; Matos etal., into clinical practice is likely. Possible hurdles are
2012b). Aprimary role of astrocytic A 2ARs might the development of local adenosine augmentation
be to sense synaptic transmission (which gener- therapies, which might best be achieved by gene
ates ATP adenosine signals) in order to adjust therapy; however, dietary alternatives provide a
extracellular levels of glutamate by modulation readily available alternative.
of transporter activity (Matos et al., 2012b) or
by controlling the vesicular release of glutamate Restoration ofthe DNA Methylome
(Li etal., 2001; Nishizaki, 2004; Nishizaki etal., According to the notion seizures beget sei-
2002), thereby controlling and adjusting neu- zures, the seizures in epilepsy often increase in
ronal excitability. Thus therapeutic enhancement frequency and severity during the course of the
of astroglial glutamate uptake or attenuation disease. Several causes for the progressive course
of the astrocytic release of glutamate could be of epilepsy have been proposed, including epi-
achieved pharmacologically by blocking A 2ARs genetic modifications that lead to changes in
and thereby may constitute alternative therapeu- neuronal cell activity. While hypermethylation
tic strategies for the treatment of epilepsy that of DNA in the central nervous system is associ-
still need to be developed and tested in preclini- ated with epilepsy (Kobow and Blumcke, 2011;
cal and clinical studies. Qureshi and Mehler, 2010), there has been a lack
of evidence directly demonstrating a causative
Adenosine Augmentation role for increased DNA methylation in epilepsy
As mentioned, adenosine deficiency is a patholog- progression. We therefore explored the role
ical hallmark of epilepsy and sufficient to trigger of adenosine as a potential therapeutic agent
Epilepsy 409

capable of reversing epigenetic changes associ- Drug Transporters in


ated with epilepsy (Williams-Karnesky et al., the Blood-Brain Barrier
2013). We confirmed increased DNA methyla- The expression of transporters for AEDs and
tion in the hippocampus of epileptic rats; hyper- enzymes such as cytochrome P450 that metabo-
methylation of the epileptogenic hippocampus lize AEDs in glial cells of the BBB determine
was associated with increased metabolic clear- whether antiepileptic treatments can reach
ance of adenosine through increased expression the epileptogenic brain areas. Expression of
of astroglial ADK (Figure 23.3). Importantly, these multidrug transporters, which include
the transient delivery of adenosine to the brain ATP-binding cassette proteins and the human
via an engineered silk-based brain implant cor- P-glycoprotein, is severely disturbed in the epi-
rected the epilepsy-associated changes in DNA leptic brain and might contribute to pharmacore-
methylation and prevented disease progression sistance in intractable epilepsies; overexpression
in these animals long term. These findings dem- of either the transporters or of AED-metabolizing
onstrate that changes in DNA methylation pat- enzymes in the BBB has therefore been linked
terns are a key determinant of the progression to pharmacoresistant epilepsy (Ghosh et al.,
of epilepsy and that adenosine augmentation 2010; Loscher, 2007; Loscher and Delanty, 2009;
therapies may reverse DNA hypermethylation Loscher et al., 2011; Luna-Tortos et al., 2008).
and break the cycle of increasing seizure sever- Association studies investigating drug trans-
ity (Williams-Karnesky et al., 2013). Our work porter gene polymorphisms within the context
suggests a biphasic response of the DNA meth- of pharmacoresistance in epilepsy have yielded
ylome in response to an epileptogenesis trig- conflicting results: whereas pharmacoresist-
gering insult: acute DNA hypomethylation as ance to carbamazepine has been associated
a presumably epileptogenesis-triggering event with single nucleotide polymorphisms (SNPs)
followed by chronic DNA hypermethylation pre- in ATP-binding cassette transporters, polymor-
sumably required to maintain the epileptic state phisms in the human multidrug resistance gene
and to promote disease progression. Importantly could not be associated with pharmacoresistance
this biphasic response can be directly related in epilepsy. Overexpression of P-glycoprotein in
to biphasic expression changes of ADK dur- epilepsy depends on a cyclooxygenase 2 medi-
ing the course of epileptogenesis (Boison, 2008; ated signaling pathway (van Vliet et al., 2010).
Li et al., 2008; Williams-Karnesky et al., 2013). It was shown that cyclooxygenase 2 blockade
Acute DNA hypomethylation has also been with anti-inflammatory drugs normalized
found within 24 hours after a traumatic brain P-glycoprotein expression in rats and improved
injury and has been associated with microglial brain penetration of phenytoin (van Vliet etal.,
activation (Zhang et al., 2007). Those epige- 2010). In contrast to direct transporter inhibi-
netic changes might become future diagnostic tion, this therapeutic strategy might have transla-
tools to predict outcome following a potential tional value to preserve physiological functions,
epileptogenesis-triggering event, such as trau- while specifically normalizing the pathological
matic brain injury (Conley and Alexander, 2011). transporter functions (Potschka,2010).
Adenosine augmentation via ADK inhibitors
might be a therapeutically viable option to pre- Anti-Inflammatory and
vent epileptogenesis (Boison, 2013). ADK inhibi- Immunomodulatory Therapies
tors are highly effective in raising hippocampal As outlined, brain insults, such as SE or trau-
adenosine (Pak etal., 1994)and have been used in matic brain injury, trigger inflammatory
preclinical drug development for the treatment of processes thought to play a crucial role in epi-
epilepsy and chronic pain (McGaraughty et al., leptogenesis (Vezzani et al., 2011a). Therefore
2005). However, drug development efforts have anti-inflammatory therapies are potentially
largely been abandoned due to major side effects antiepileptogenic. In line with this notion, the
of chronic drug use leading to cardiovascular cyclooxygenase 2 inhibitor celecoxib, admin-
depression and liver toxicity (Boison, 2013). If, istered chronically for a duration of 2 weeks
however, a transient therapy with an ADK inhibi- beginning 24 hours following an SE prevented
tor has long-lasting antiepileptogenic effects, we hippocampal neuronal injury and reduced the
may already have suitable drugs that could be incidence and frequency of spontaneous recurrent
repurposed for the indication antiepileptogenesis seizures in rats (Jung etal., 2006). IL-1 appears
(Boison,2013). to be a key player in the onset of injury-induced
410 Part IV:Homeostatic Therapies

inflammation (Vezzani and Baram, 2007). occurring during childhood (Kossoff and Rho,
Consequently, pretreatment of rodents with an 2009; McQuarrie and Keith, 1927). Importantly,
IL-1 receptor antagonist prior to an SE signifi- ketogenic diet therapy is effective in epilepsies
cantly reduced SE onset and BBB damage (Marchi that are refractory to conventional AED treat-
etal., 2009); however, pretreatment regimens will ment (Mackay et al., 2005; Sirven et al., 1999).
also affect the quality and quantity of the precipi- Moreover, the literature reports on patients
tating injury level, and therefore any interpreta- treated with a ketogenic diet that remained sei-
tions regarding antiepileptogenic outcome must zure free after diet reversal, suggesting poten-
be treated with caution. More recently, the selec- tial antiepileptogenic effects of the diet (Kossoff
tive interleukin-converting enzyme/caspase-1 and Rho, 2009). Last, ketogenic diet therapy not
inhibitor VX-765 was used to block the biosyn- only affects epileptic seizures but also normal-
thesis of IL-1; this approach led to a reduction of izes behavioral symptoms thought to be comor-
chronic epileptic activity in mice (Maroso etal., bid with epilepsy (Masino et al., 2013). Given
2011). Rapamycin is an immunosuppressive drug those characteristics, ketogenic diet therapy is
acting on the serine/threonine protein kinase a prototypic homeostatic therapy. This notion
mammalian target of rapamycin (mTOR), which is supported by several beneficial mechanisms
regulates many forms of synaptic plasticity in the thought to be activated by ketogenic diet therapy.
adult brain and has been linked with plasticity Ketogenic diet therapy forces the brain to use
changes during epileptogenesis (Cao etal., 2009). ketones in lieu of glucose as the primary energy
mTOR signaling is also involved in the expres- source, and it is those metabolic changes leading
sion of glutamate transporter 1 in astrocytes (Wu to disruption of glutamatergic synaptic transmis-
et al., 2010). Thus dysregulation of mTOR sign- sion, inhibition of glycolysis, increase in adeno-
aling in astrocytes might critically affect major sine, and activation of ATP-sensitive potassium
homeostatic systems of the brain thought to be of channels that are thought to underlie the thera-
relevance for epileptogenesis. In a rodent model peutic effects of this type of dietary intervention
of epilepsy, the mTOR pathway was found to be (Bough, 2008; Bough etal., 2006; Kalapos, 2007;
upregulated following kainic acidinduced SE Lutas and Yellen, 2013; Ma et al., 2007; Masino
and was linked to chronic epileptogenesis (Zeng et al., 2011; Yellen, 2008). A large body of evi-
et al., 2009). If mTOR activation plays a role in dence supports the notion that a ketogenic diet
epileptogenesis, then mTOR blockade should leads to increased adenosine signaling in the
have antiepileptogenic effects. Consequently, brain (Masino and Geiger, 2008, 2009; Masino
rapamycin blocked SE-induced mossy fiber et al., 2012; Masino et al., 2009) most likely via
sprouting and the frequency of spontaneous sei- reduced expression of ADK (Masino etal., 2011).
zures in two independent studies (Buckmaster Reduction of ADK expression seems to be a gen-
et al., 2009; Zeng et al., 2009). Since seizures eral mechanism to shift the brain into a protective
were recorded only during the treatment period mode (Pignataro et al., 2008). As an additional
(Zeng et al., 2009) it remains to be determined mechanism, activation of the metabolically sensi-
whether the observed therapeutic effects were tive ATP-sensitive potassium channel was shown
anti-ictogenic or antiepileptogenic. Based on the to be instrumental in the resistance to behavioral
studies discussed previously, anti-inflammatory and electrographic seizures in vivo, an effect that
therapies are certainly promising candidates for was dependent on Bcl-associated death protein
antiepileptogenesis. To be effective, those thera- modifications that reduce glucose metabolism
pies should be initiated as soon as possible fol- (Gimenez-Cassina etal., 2012). Metabolic thera-
lowing a precipitating event and maintained for pies can also effect gene expression as a possible
approximately 2 weeks, a time window during explanation for antiepileptogenic effects of the
which inflammatory and immunomodulatory diet. It was shown that the glycolytic inhibitor
processpeak. 2-deoxy-d-glucose potently reduced the pro-
gression of kindling epileptogenesis and blocked
Dietary Interventions seizure-induced increases in BDNF and its recep-
Mounting evidence suggests that diet plays a tor, TrkB. This effect was found to depend on the
major role in setting the homeostatic stage in neuron-restrictive silencer factor, a transcrip-
the brain. High-fat/low-carbohydrate ketogenic tion factor that recruits the reduced nicotina-
diets have been in clinical use for over 80years mide adenine dinucleotide (NADH)-binding
to treat refractory epilepsies, particularly those C-terminal binding protein, a corepressor
Epilepsy 411

generating a repressive chromatin environment Porkka-Heiskanen and Kalinchuk, 2011)and are


around the BDNF promoter (Garriga-Canut increased by bacterial endotoxins (Ramakers
etal., 2006). Together, these findings suggest that et al., 2011). Consequently, any lifestyle-related
a ketogenic diet exerts antiepileptic effects by a changes in adenosine homeostasis will have a
combination of acute mechanisms and genomic direct impact on the susceptibility of epilep-
mechanisms, which may lead to long-term altera- tic seizures. Conversely, methylxanthines such
tions of neuronal circuitry. Additional dietary as caffeine or theophylline, well-characterized
components such as branched-chain amino acids antagonists of adenosine receptors, have in gen-
(Novarino et al., 2012), certain oils (Bandero eral acute proconvulsant activities; however, the
etal., 2013; Koutroumanidou etal., 2013; Shawki chronic use of methylxanthines leads to adaptive
etal., 2013), or medium-chain triglycerides (Liu increases in the adenosine A1 receptor and thereby
and Wang, 2013)all have potential for the meta- enhances the antiepileptic activity of endogenous
bolic therapy of epilepsy and are readily translat- adenosine (Boison, 2010). Overall, it is estimated
able. In particular, medium-chain triglycerides that up to 80% of all persons with epilepsy are
with an uneven number of carbon atoms, such habitual consumers of caffeine. While chronic
as triheptanoin, have shown significant promise caffeine use might be beneficial to alleviate cog-
in enforcing a ketone-based metabolism in the nitive side effects of AEDs and might increase the
brain with proven antiepileptic efficacy in several capacity of endogenous adenosinedependent
rodent models of epilepsy (Hadera et al., 2013; seizure-control mechanisms, caffeine might also
Kim etal.,2013). play a crucial role in sudden unexpected death
in epilepsy (SUDEP; Shen et al., 2010). On one
Lifestyle hand, caffeine can prevent respiratory arrest and
Lifestyle choices can have a significant impact SUDEP (Shen etal., 2010), but SUDEP in people
on brain function. Physical exercise is known usually occurs at night during sleep when caf-
to increase neurogenesis in the adult brain (van feine levels are lowest. It needs to be determined
Praag et al., 1999), a mechanism thought to in careful clinical studies whether daytime caf-
protect the brain from hippocampal degenera- feine use may increase nighttime SUDEP risk
tion and that has been demonstrated to enhance and whether caffeine in general is of any benefit
cognitive function (Miltiadous et al., 2013). for epilepsy and its comorbidities.
Pathological neurogenesis, as triggered by epi-
leptic seizures, in contrast, is thought to aggra- Age andGender
vate the epileptogenic cascade. Several lines of The incidence of epilepsy peaks in the juvenile
evidence implicate newly generated neurons in (<5 years of age) and in the elderly populations
structural and functional network abnormali- (>60years of age; Annegers etal., 1999). However,
ties in the epileptic brain, such as aberrant mossy treatment outcome may be age dependent. In gen-
fiber reorganization, persistence of immature eral, epilepsy with an age of onset <12years has a
dentate granular cell structure, and the abnormal lower recurrence risk following discontinuation
migration of newborn neurons to ectopic sites in of medication than that of an older age of onset
the dentate gyrus (Parent and Lowenstein, 2002). (Berg and Shinnar, 1991, 1994). In contrast, the
Whereas the significance of exercise-induced probability of attaining and maintaining remis-
neurogenesis on seizure propensity is unknown, sion after medication withdrawal is a function
exercise-induced neurogenesis is certainly of of age of onset and duration of the seizure disor-
value to ameliorate cognitive comorbidities of der, without a special role for puberty (Shinnar
epilepsy. While clinical studies indicate that per- and Moshe, 1991). Children with higher meta-
sons with epilepsy have in general reduced their bolic rates usually require higher drug dosages,
physical activity levels as a result of their condi- as measured in milligram drug per kilogram
tion (Jalava and Sillanpaa, 1997), clinical studies body weight, than adults. Whereas ketogenic
evaluating potential benefits of exercise for epi- diets traditionally have mostly been used for the
lepsy are surprisingly lacking. treatment of childhood epilepsies, a recent study
Several lifestyle choices influence adenosine suggests that ketogenic diet therapy is equally
signaling in the brain; intense physical exercise effective in adult and adolescent patients (Nei
has been shown to increase adenosine in the etal.,2014).
brain (Dworak etal., 2007). Adenosine levels in Hormones have a marked effect on neuronal
the brain also depend on sleep (Huang etal., 2011; excitability. Estrogen reduces the concentration
412 Part IV:Homeostatic Therapies

of GABA and reduces the number and chloride dietary and lifestyle interventions might hold
flux of GABA A receptors; in addition, estrogen promise to affect network homeostasis as a novel
activates N-methyl-D-aspartate receptors in the conceptual strategy to treat and prevent epilepsy
hippocampus (Morrell, 2002). Thereby estrogen on the network level. Adenosine emerges as a pro-
exerts potent proconvulsive effects that may trig- totype homeostatic network regulator with the
ger seizures, when seizure thresholds are patho- proven capability to control network activity both
logically lowered. Conversely, progesterone has through receptor-dependent as well as through
opposite effects on GABAergic and glutamatergic receptor-independent epigenetic and bioener-
neurotransmission, resulting in anticonvulsant getic mechanisms (Boison, 2013). Consequently,
effects. For those reasons, catamenial, menstrual therapeutic adenosine augmentation has been
cycleassociated seizure patterns, arise in women demonstrated to suppress epileptic seizures (Li
with seizures more likely to occur in the premen- etal., 2008; Li etal., 2007), and to prevent disease
strual and ovulatory phases (i.e., at times when progression and epileptogenesis (Li et al., 2008;
estrogen levels are high and progesterone levels Williams-Karnesky etal., 2013), but also to pre-
are low; Hattemer etal., 2006). AED elimination vent psychosis and improve cognition (Shen etal.,
can depend on both sex and age. Fertile women 2012) without any known adverse effects. The
often have faster drug metabolism rates than robust antipsychotic and procognitive effect of
age-matched men, whereas the rate of metabo- adenosine in mice (Shen etal., 2012)suggests that
lism decreases with age in both sexes. An efficacy therapeutic adenosine augmentation might com-
analysis of newer AEDs by gender, however, did bine anticonvulsant with cognition-enhancing
not show any major differences in efficacy and effects. In preclinical toxicity studies of intrathecal
tolerability with the exception of zonisamide, adenosine in dogs, no side effects were observed
which was better tolerated by women (Morrell, with intrathecal adenosine infused chronically
1996; Yerby, 2000a, 2000b). for 26days (Chiari etal., 1999). Likewise, intrath-
ecal adenosine was tested in humans in escalating
doses of up to 2 mg without any adverse effects
CONCLUSIONS AND (Eisenach etal., 2002a, 2002b). Several therapeu-
OUTLOOK tic adenosine augmentation strategies ranging
The excitability of the brain is determined by from gene therapy to dietary intervention are
neurons, which in turn are connected to net- currently in preclinical development. It is pos-
works, which in turn are broadly controlled by sible that adenosine-augmenting therapies will
the homeostatic environment of their surround- be introduced into the clinic as a novel class of
ings, which to a large degree is under the control homeostatic network therapy within the next
of glial cells. It becomes clear that complex neu- 10years. Challenges will include developing strat-
rological syndromes, such as epilepsy, which are egies to confine adenosines action to identifiable
not only defined by a dominant symptom (i.e., a target areas and cells; the advent of cell-type spe-
seizure) but also by a growing number of associ- cific gene therapy vectors might offer a promising
ated comorbidities, can best be explained by the strategy to manipulate adenosine homeostasis in
disruption of network homeostasis. Disruption a localized and cell-type selective manner.
of network homeostasis implies the simultane-
ous and concordant dysregulation of several ACK NOWLEDGMENTS
molecular pathways, which in turn can affect The author is indebted to outstanding experi-
each other and lead to a progression of the dis- mental work from his research team. The
ease in the sense that seizures beget seizures. authors work is funded through grants from the
This old concept has a lot of truth and can best National Institutes of Health (R01 NS065957, R01
be explained by the self-reinforcing interplay of MH083973, R01 NS061844), the US Department of
several homeostatic systems that become pro- the Army (W81XWH-12-1-0283), Citizens United
gressively dysregulated during disease progres- for Research in Epilepsy, the Parkinsons Northwest
sion. Conventional AEDs with a target-centric Group, and the Legacy Hospital Foundations.
mode of action are unlikely to affect network
homeostasis or prevent the progressive maladap- References
tive changes occurring during epileptogenesis, Anderson, C.M., and Swanson, R.A. (2000).
which can be considered a lifelong process of dis- Astrocyte glutamate transport: Review of prop-
ease progression. Novel therapeutic interventions erties, regulation, and physiological functions.
based on adenosine, epigenetic mechanisms, or Glia 32,114.
Epilepsy 413

Annegers, J.F., Dubinsky, S., Coan, S.P., Newmark, Boison, D., Masino, M.A., and Geiger, J.D. (2011).
M.E., and Roht, L. (1999). The incidence of epi- Homeostatic bioenergetic network regulationa
lepsy and unprovoked seizures in multieth- novel concept to avoid pharmacoresistance in
nic, urban health maintenance organizations. epilepsy. Expert Opin Drug Discov 6, 713724.
Epilepsia 40, 502506. Boison, D., Scheurer, L., Zumsteg, V., Rlicke, T.,
Aronica, E., Ravizza, T., Zurolo, E., and Vezzani, A. Litynski, P., Fowler, B., Brandner, S., and Mohler,
(2012). Astrocyte immune responses and epi- H. (2002). Neonatal hepatic steatosis by disrup-
lepsy. Glia 60, 12581268. tion of the adenosine kinase gene. Proc Natl
Aronica, E., Zurolo, E., Iyer, A., de Groot, M., Anink, Acad Sci USA 99, 69856990.
J., Carbonell, C., van Vliet, E.A., Baayen, J.C., Bough, K. (2008). Energy metabolism as part of the
Boison, D., and Gorter, J.A. (2011). Upregulation anticonvulsant mechanism of the ketogenic diet.
of adenosine kinase in astrocytes in experimen- Epilepsia 49 Suppl 8,9193.
tal and human temporal lobe epilepsy. Epilepsia Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F.,
52, 16451655. Gawryluk, J.W., Greene, J.G., Shaw, R., Smith, Y.,
Bandero, C.R., Salvadori, M.G., Gomes, A.T., Geiger, J.D., and Dingledine, R.J. (2006).
Dal Ri, N.M., Furian, A.F., Oliveira, M.S., Mitochondrial biogenesis in the anticonvulsant
Rambo, L.M., Scorza, F.A., Cysneiros, R.M., mechanism of the ketogenic diet. Ann Neurol 60,
Emanuelli, T., et al. (2013). Fish oil attenuates 223235.
methylmalonate-induced seizures. Epilepsy Res Brambilla, R., Cottini, L., Fumagalli, M., Ceruti, S.,
105,6976. and Abbracchio, M.P. (2003). Blockade of A2A
Berg, A.T., and Shinnar, S. (1991). The risk of sei- adenosine receptors prevents basic fibroblast
zure recurrence following a first unprovoked growth factor-induced reactive astrogliosis in rat
seizure: A quantitative review. Neurology 41, striatal primary astrocytes. Glia 43, 190194.
965972. Brooks-Kayal, A.R., Bath, K.G., Berg, A.T.,
Berg, A.T., and Shinnar, S. (1994). Relapse fol- Galanopoulou, A.S., Holmes, G.L., Jensen, F.E.,
lowing discontinuation of antiepileptic Kanner, A.M., OBrien, T.J., Whittemore, V.H.,
drugs:Ameta-analysis. Neurology 44, 601608. Winawer, M.R., et al. (2013). Issues related to
Binder, D.K., Nagelhus, E.A., and Ottersen, O.P. symptomatic and disease-modifying treatments
(2012). Aquaporin-4 and epilepsy. Glia 60, affecting cognitive and neuropsychiatric comor-
12031214. bidities of epilepsy. Epilepsia 54 Suppl 4,4460.
Binder, D.K., Oshio, K., Ma, T., Verkman, A.S., and Buckmaster, P.S., Ingram, E.A., and Wen, X. (2009).
Manley, G.T. (2004a). Increased seizure thresh- Inhibition of the mammalian target of rapamy-
old in mice lacking aquaporin-4 water channels. cin signaling pathway suppresses dentate granule
Neuroreport 15, 259262. cell axon sprouting in a rodent model of tempo-
Binder, D.K., Papadopoulos, M.C., Haggie, P.M., ral lobe epilepsy. J Neurosci 29, 82598269.
and Verkman, A.S. (2004b). In vivo measure- Cacheaux, L.P., Ivens, S., David, Y., Lakhter, A.J.,
ment of brain extracellular space diffusion by Bar-Klein, G., Shapira, M., Heinemann, U.,
cortical surface photobleaching. J Neurosci 24, Friedman, A., and Kaufer, D. (2009). Transcriptome
80498056. profiling reveals TGF-beta signaling involvement
Boison, D. (2008). The adenosine kinase hypothesis in epileptogenesis. J Neurosci 29, 89278935.
of epileptogenesis. Prog Neurobio 84, 249262. Cao, R., Li, A., and Cho, H.Y. (2009). mTOR signal-
Boison, D. (2009). Adenosine augmentation thera- ing in epileptogenesis:Too much of a good thing?
pies (AATs) for epilepsy: Prospect of cell and J Neurosci 29, 1237212373.
gene therapies. Epilepsy Res 85, 131141. Chiari, A., Yaksh, T.L., Myers, R.R., Provencher, J.,
Boison, D. (2010). Methylxanthines, seizures and Moore, L., Lee, C.S., and Eisenach, J.C. (1999).
excitotoxicity. Handb Exp Pharmacol 200, Preclinical toxicity screening of intrathecal
251266. adenosine in rats and dogs. Anesthesiology 91,
Boison, D. (2012a). Adenosine augmentation therapy 824832.
for epilepsy. In:Jaspers Basic Mechanisms of the Clark, R.S., Carcillo, J.A., Kochanek, P.M., Obrist,
Epilepsies, J.L. Noebels, M. Avoli, M.A. Rogawski, W.D., Jackson, E.K., Mi, Z., Wisneiwski,
R.W. Olsen, and A.V. Delgado-Escueta, eds. S.R., Bell, M.J., and Marion, D.W. (1997).
(Oxford:Oxford University Press), pp. 11501160. Cerebrospinal fluid adenosine concentration
Boison, D. (2012b). Adenosine dysfunction in epi- and uncoupling of cerebral blood flow and oxi-
lepsy. Glia 60, 12341243. dative metabolism after severe head injury in
Boison, D. (2013). Adenosine kinase:Exploitation for humans. Neurosurgery 41, 12841292; discus-
therapeutic gain. Pharmacol Rev 65, 906943. sion 12921293.
414 Part IV:Homeostatic Therapies

Conley, Y.P., and Alexander, S. (2011). Genomic, tran- Feng, Z., and Durand, D.M. (2006). Effects of
scriptomic, and epigenomic approaches to recov- potassium concentration on firing patterns of
ery after acquired brain injury. PM R 3, S5258. low-calcium epileptiform activity in anesthe-
Coulter, D.A., and Eid, T. (2012). Astrocytic regula- tized rat hippocampus: Inducing of persistent
tion of glutamate homeostasis in epilepsy. Glia spike activity. Epilepsia 47, 727736.
60, 12151226. Fredholm, B.B. (2012). Rethinking the purinergic
David, Y., Cacheaux, L.P., Ivens, S., Lapilover, E., neuron-glia connection. Proc Natl Acad Sci USA
Heinemann, U., Kaufer, D., and Friedman, A. 109, 59135914.
(2009). Astrocytic dysfunction in epileptogene- Garriga-Canut, M., Schoenike, B., Qazi, R.,
sis:Consequence of altered potassium and gluta- Bergendahl, K., Daley, T.J., Pfender, R.M.,
mate homeostasis? J Neurosci 29, 1058810599. Morrison, J.F., Ockuly, J., Stafstrom, C., Sutula,
Doolette, D.J. (1997). Mechanism of adenosine accu- T., et al. (2006). 2-Deoxy-D-glucose reduces
mulation in the hippocampal slice during energy epilepsy progression by NRSF-CtBP-dependent
deprivation. Neurochem Intl 30, 211223. metabolic regulation of chromatin structure. Nat
Dworak, M., Diel, P., Voss, S., Hollmann, W., Neurosci 9, 13821387.
and Struder, H.K. (2007). Intense exercise Ghosh, C., Gonzalez-Martinez, J., Hossain, M.,
increases adenosine concentrations in rat Cucullo, L., Fazio, V., Janigro, D., and Marchi, N.
brain:Implications for a homeostatic sleep drive. (2010). Pattern of P450 expression at the human
Neuroscience 150, 789795. blood-brain barrier:Roles of epileptic condition
Eid, T., Ghosh, A., Wang, Y., Beckstrom, H., Zaveri, and laminar flow. Epilepsia 51, 14081417.
H.P., Lee, T.S., Lai, J.C., Malthankar-Phatak, Gimenez-Cassina, A., Martinez-Francois, J.R.,
G.H., and de Lanerolle, N.C. (2008). Recurrent Fisher, J.K., Szlyk, B., Polak, K., Wiwczar, J.,
seizures and brain pathology after inhibition Tanner, G.R., Lutas, A., Yellen, G., and Danial,
of glutamine synthetase in the hippocampus in N.N. (2012). BAD-dependent regulation of fuel
rats. Brain 131, 20612070. metabolism and K(ATP) channel activity con-
Eid, T., Lee, T.S., Thomas, M.J., Amiry-Moghaddam, fers resistance to epileptic seizures. Neuron 74,
M., Bjornsen, L.P., Spencer, D.D., Agre, P., 719730.
Ottersen, O.P., and de Lanerolle, N.C. (2005). Gomez-Gonzalo, M., Losi, G., Chiavegato, A., Zonta,
Loss of perivascular aquaporin 4 may under- M., Cammarota, M., Brondi, M., Vetri, F., Uva,
lie deficient water and K+ homeostasis in the L., Pozzan, T., de Curtis, M., et al. (2010). An
human epileptogenic hippocampus. Proc Natl excitatory loop with astrocytes contributes to
Acad Sci USA 102, 11931198. drive neurons to seizure threshold. PLoS Biol 8,
Eid, T., Thomas, M.J., Spencer, D.D., Runden-Pran, e1000352.
E., Lai, J.C., Malthankar, G.V., Kim, J.H., Grabenstatter, H.L., Russek, S.J., and Brooks-Kayal,
Danbolt, N.C., Ottersen, O.P., and de Lanerolle, A.R. (2012). Molecular pathways controlling
N.C. (2004). Loss of glutamine synthetase in the inhibitory receptor expression. Epilepsia 53
human epileptogenic hippocampus: Possible Suppl 9,7178.
mechanism for raised extracellular glutamate in Guo, Q., Sayeed, I., Baronne, L.M., Hoffman,
mesial temporal lobe epilepsy. Lancet 363,2837. S.W., Guennoun, R., and Stein, D.G. (2006).
Eisenach, J.C., Hood, D.D., and Curry, R. (2002a). Progesterone administration modulates AQP4
Phase I safety assessment of intrathecal injec- expression and edema after traumatic brain
tion of an American formulation of adenosine in injury in male rats. Exp Neurol 198, 469478.
humans. Anesthesiology 96,2428. Hadera,M.G.,Smeland,O.B.,McDonald,T.S.,Tan,K.N.,
Eisenach, J.C., Hood, D.D., and Curry, R. (2002b). Sonnewald, U., and Borges, K. (2013). Triheptanoin
Preliminary efficacy assessment of intrathecal partially restores levels of tricarboxylic acid cycle
injection of an American formulation of adenos- intermediates in the mouse pilocarpine model of
ine in humans. Anesthesiology 96,2934. epilepsy. J Neurochem 129, 107119.
Fedele, D.E., Li, T., Lan, J.Q., Fredholm, B.B., and Haj-Yasein, N.N., Jensen, V., Vindedal, G.F.,
Boison, D. (2006). Adenosine A1 receptors are Gundersen, G.A., Klungland, A., Ottersen, O.P.,
crucial in keeping an epileptic focus localized. Hvalby, O., and Nagelhus, E.A. (2011). Evidence
Exp Neurol 200, 184190. that compromised K+ spatial buffering con-
Feng, J., Zhou, Y., Campbell, S.L., Le, T., Li, E., Sweatt, tributes to the epileptogenic effect of mutations
J.D., Silva, A.J., and Fan, G. (2010). Dnmt1 and in the human Kir4.1 gene (KCNJ10). Glia 59,
Dnmt3a maintain DNA methylation and regu- 16351642.
late synaptic function in adult forebrain neurons. Halassa, M.M., Dal Maschio, M., Beltramo, R.,
Nat Neurosci 13, 423430. Haydon, P.G., Benfenati, F., and Fellin, T. (2010).
Epilepsy 415

Integrated brain circuits: Neuron-astrocyte recurrent seizures following pilocarpine-induced


interaction in sleep-related rhythmogenesis. status epilepticus. Neurobiol Dis 23, 237246.
ScientificWorldJournal 10, 16341645. Kalapos, M.P. (2007). Possible mechanism for the
Hardie, D.G. (2004). The AMP-activated protein effect of ketogenic diet in cases of uncontrolled
kinase pathwaynew players upstream and seizures:The reconsideration of acetone theory.
downstream. J Cell Sci 117, 54795487. Med Hypotheses 68, 13821388.
Hattemer, K., Knake, S., Reis, J., Oertel, W.H., Kanner, A.M. (2012). Psychiatric comorbidities and
Rosenow, F., and Hamer, H.M. (2006). Cyclical epilepsy:Is it the old story of the chicken and the
excitability of the motor cortex in patients with egg? Ann Neurol 72, 153155.
catamenial epilepsy: A transcranial magnetic Kim, J.H., Roberts, D.S., Hu, Y., Lau, G.C.,
stimulation study. Seizure 15, 653657. Brooks-Kayal, A.R., Farb, D.H., and Russek, S.J.
Haydon, P.G., and Carmignoto, G. (2006). Astrocyte (2012). Brain-derived neurotrophic factor uses
control of synaptic transmission and neurovas- CREB and Egr3 to regulate NMDA receptor levels
cular coupling. Physiol Rev 86, 10091031. in cortical neurons. J Neurochem 120, 210219.
Heinemann, U., Gabriel, S., Jauch, R., Schulze, K., Kim, T.H., Borges, K., Petrou, S., and Reid, C.A.
Kivi, A., Eilers, A., Kovacs, R., and Lehmann, T.N. (2013). Triheptanoin reduces seizure susceptibil-
(2000). Alterations of glial cell function in ity in a syndrome-specific mouse model of gener-
temporal lobe epilepsy. Epilepsia 41 Suppl 6, alized epilepsy. Epilepsy Res 103, 101105.
S185S189. Kobow, K., and Blumcke, I. (2011). The methylation
Heinemann, U., Kaufer, D., and Friedman, A. (2012). hypothesis: Do epigenetic chromatin modifica-
Blood-brain barrier dysfunction, TGF-beta sig- tions play a role in epileptogenesis? Epilepsia 52
naling and astrocyte dysfunction in epilepsy. Suppl 4,1519.
Glia 60, 12511257. Kobow, K., Jeske, I., Hildebrandt, M., Hauke, J.,
Hermann, B., Seidenberg, M., and Jones, J. (2008). Hahnen, E., Buslei, R., Buchfelder, M., Weigel,
The neurobehavioural comorbidities of epi- D., Stefan, H., Kasper, B., etal. (2009). Increased
lepsy:Can a natural history be developed? Lancet reelin promoter methylation is associated with
Neurol 7, 151160. granule cell dispersion in human temporal lobe
Hsu, M.S., Seldin, M., Lee, D.J., Seifert, G., epilepsy. J Neuropathol Exp Neurol 68, 356364.
Steinhauser, C., and Binder, D.K. (2011). Kochanek, P.M., Vagni, V.A., Janesko, K.L.,
Laminar-specific and developmental expres- Washington, C.B., Crumrine, P.K., Garman,
sion of aquaporin-4 in the mouse hippocampus. R.H., Jenkins, L.W., Clark, R.S., Homanics, G.E.,
Neuroscience 178,2132. Dixon, C.E., et al. (2006). Adenosine A1 recep-
Huang, Z.L., Urade, Y., and Hayaishi, O. (2011). The tor knockout mice develop lethal status epilepti-
role of adenosine in the regulation of sleep. Curr cus after experimental traumatic brain injury. J
Top Med Chem 11, 10471057. Cereb Blood Flow Metab 26, 565575.
Ivens, S., Kaufer, D., Flores, L.P., Bechmann, I., Kossoff, E.H., and Rho, J.M. (2009). Ketogenic
Zumsteg, D., Tomkins, O., Seiffert, E., diets:Evidence for short- and long-term efficacy.
Heinemann, U., and Friedman, A. (2007). Neurotherapeutics 6, 406414.
TGF-beta receptor-mediated albumin uptake Koutroumanidou, E., Kimbaris, A., Kortsaris, A.,
into astrocytes is involved in neocortical epilep- Bezirtzoglou, E., Polissiou, M., Charalabopoulos,
togenesis. Brain 130, 535547. K., and Pagonopoulou, O. (2013). Increased
Jalava, M., and Sillanpaa, M. (1997). Physical activ- seizure latency and decreased severity of
ity, health-related fitness, and health experience pentylenetetrazol-induced seizures in mice after
in adults with childhood-onset epilepsy:Acon- essential oil administration. Epilepsy Res Treat
trolled study. Epilepsia 38, 424429. 2013, 532657.
James, S.J., Melnyk, S., Pogribna, M., Pogribny, Kredich, N.M., and Martin, D.V. Jr. (1977). Role of
I.P., and Caudill, M.A. (2002). Elevation in S-adenosylhomocysteine in adenosinemediated
S-adenosylhomocysteine and DNA hypometh- toxicity in cultured mouse T lymphoma cells.
ylation: Potential epigenetic mechanism for Cell 12, 931938.
homocysteine-related pathology. J Nutr 132, LaFrance, W.C. Jr., Kanner, A.M., and Hermann, B.
2361S2366S. (2008). Psychiatric comorbidities in epilepsy. Int
Jung, K.H., Chu, K., Lee, S.T., Kim, J., Sinn, D.I., Rev Neurobiol 83, 347383.
Kim, J.M., Park, D.K., Lee, J.J., Kim, S.U., Kim, Lee, S.G., Su, Z.Z., Emdad, L., Gupta, P., Sarkar, D.,
M., et al. (2006). Cyclooxygenase-2 inhibitor, Borjabad, A., Volsky, D.J., and Fisher, P.B. (2008).
celecoxib, inhibits the altered hippocampal Mechanism of ceftriaxone induction of excit-
neurogenesis with attenuation of spontaneous atory amino acid transporter-2 expression and
416 Part IV:Homeostatic Therapies

glutamate uptake in primary human astrocytes. epigenetic DNA demethylation and adult neuro-
J Biol Chem 283, 1311613123. genesis. Science 323, 10741077.
Li, T., Ren, G., Lusardi, T., Wilz, A., Lan, J.Q., Ma, W., Berg, J., and Yellen, G. (2007). Ketogenic
Iwasato, T., Itohara, S., Simon, R.P., and Boison, diet metabolites reduce firing in central neu-
D. (2008). Adenosine kinase is a target for the rons by opening K(ATP) channels. J Neurosci 27,
prediction and prevention of epileptogenesis in 36183625.
mice. J Clin Inv 118, 571582. Mackay, M.T., Bicknell-Royle, J., Nation, J.,
Li, T., Steinbeck, J.A., Lusardi, T., Koch, P., Lan, Humphrey, M., and Harvey, A.S. (2005). The
J.Q., Wilz, A., Segschneider, M., Simon, R.P., ketogenic diet in refractory childhood epilepsy.
Brustle, O., and Boison, D. (2007). Suppression J Paediatr Child Health 41, 353357.
of kindling epileptogenesis by adenosine releas- Malmgren, K., and Thom, M. (2012). Hippocampal
ing stem cell-derived brain implants. Brain 130, sclerosisorigins and imaging. Epilepsia 53
12761288. Suppl 4,1933.
Li, X.X., Nomura, T., Aihara, H., and Nishizaki, T. Marchi, N., Angelov, L., Masaryk, T., Fazio, V.,
(2001). Adenosine enhances glial glutamate Granata, T., Hernandez, N., Hallene, K.,
efflux via A2a adenosine receptors. Life Sci 68, Diglaw, T., Franic, L., Najm, I., et al. (2007).
13431350. Seizure-promoting effect of blood-brain barrier
Lin, J.J., Mula, M., and Hermann, B.P. (2012). disruption. Epilepsia 48, 732742.
Uncovering the neurobehavioural comorbidi- Marchi, N., Fan, Q., Ghosh, C., Fazio, V., Bertolini,
ties of epilepsy over the lifespan. Lancet 380, F., Betto, G., Batra, A., Carlton, E., Najm, I.,
11801192. Granata, T., etal. (2009). Antagonism of periph-
Liu, Y.M., and Wang, H.S. (2013). Medium-chain eral inflammation reduces the severity of status
triglyceride ketogenic diet, an effective treatment epilepticus. Neurobiol Dis 33, 171181.
for drug-resistant epilepsy and a comparison Maroso, M., Balosso, S., Ravizza, T., Iori, V.,
with other ketogenic diets. Biomed J 36,915. Wright, C.I., French, J., and Vezzani, A. (2011).
Loscher, W. (2007). Drug transporters in the epilep- Interleukin-1beta biosynthesis inhibition reduces
tic brain. Epilepsia 48 Suppl 1,813. acute seizures and drug resistant chronic epileptic
Loscher, W., and Delanty, N. (2009). MDR1/ABCB1 activity in mice. Neurotherapeutics 8, 304315.
polymorphisms and multidrug resistance in epi- Maroso, M., Balosso, S., Ravizza, T., Liu, J., Aronica, E.,
lepsy:In and out of fashion. Pharmacogenomics Iyer, A.M., Rossetti, C., Molteni, M., Casalgrandi,
10, 711713. M., Manfredi, A.A., etal. (2010). Toll-like recep-
Loscher, W., Luna-Tortos, C., Romermann, K., and tor 4 and high-mobility group box-1 are involved
Fedrowitz, M. (2011). Do ATP-binding cassette in ictogenesis and can be targeted to reduce sei-
transporters cause pharmacoresistance in epi- zures. Nat Med 16, 413419.
lepsy? Problems and approaches in determin- Masino, S.A., and Geiger, J.D. (2008). Are purines
ing which antiepileptic drugs are affected. Curr mediators of the anticonvulsant/neuroprotec-
Pharm Des 17, 28082828. tive effects of ketogenic diets? Trends Neurosci
Loscher, W., and Potschka, H. (2002). Role of multi- 31, 273278.
drug transporters in pharmacoresistance to anti- Masino, S.A., and Geiger, J.D. (2009). The ketogenic
epileptic drugs. J Pharmacol Exp Ther 301,714. diet and epilepsy:Is adenosine the missing link?
Luna-Tortos, C., Fedrowitz, M., and Loscher, W. (2008). Epilepsia 50, 332333.
Several major antiepileptic drugs are substrates Masino, S.A., Kawamura, M.Jr., Cote, J.L., Williams,
for human P-glycoprotein. Neuropharmacology R.B., and Ruskin, D.N. (2013). Adenosine
55, 13641375. and autism: A spectrum of opportunities.
Lund, I.V., Hu, Y., Raol, Y.H., Benham, R.S., Faris, Neuropharmacology 68, 116121.
R., Russek, S.J., and Brooks-Kayal, A.R. (2008). Masino, S.A., Kawamura, M., Ruskin, D.N., Geiger,
BDNF selectively regulates GABAA receptor J.D., and Boison, D. (2012). Purines and neuronal
transcription by activation of the JAK/STAT excitability:Links to the ketogenic diet. Epilepsy
pathway. Sci Signal 1,ra9. Res 100, 229238.
Lutas, A., and Yellen, G. (2013). The ketogenic Masino, S.A., Kawamura, M., Wasser, C.A., Pomeroy,
diet: Metabolic influences on brain excitability L.T., and Ruskin, D.N. (2009). Adenosine, keto-
and epilepsy. Trends Neurosci 36,3240. genic diet and epilepsy:The emerging therapeu-
Ma, D.K., Jang, M.H., Guo, J.U., Kitabatake, Y., tic relationship between metabolism and brain
Chang, M.L., Pow-Anpongkul, N., Flavell, activity. Curr Neuropharmacol 7, 257268.
R.A., Lu, B., Ming, G.L., and Song, H. (2009). Masino, S.A., Li, T., Theofilas, P., Sandau, U.S.,
Neuronal activity-induced Gadd45b promotes Ruskin, D.N., Fredholm, B.B., Geiger, J.D.,
Epilepsy 417

Aronica, E., and Boison, D. (2011). A ketogenic Kodama, N., Takahashi, E., etal. (2002). A new
diet suppresses seizures in mice through adenos- neuromodulatory pathway with a glial contri-
ine A1 receptors. J Clin Inv 121, 26792683. bution mediated via A(2a) adenosine receptors.
Matos, M., Augusto, E., Machado, N.J., dos Glia 39, 133147.
Santos-Rodrigues, A., Cunha, R.A., and Agostinho, Novarino, G., El-Fishawy, P., Kayserili, H., Meguid,
P. (2012a). Astrocytic adenosine A2A receptors N.A., Scott, E.M., Schroth, J., Silhavy, J.L., Kara,
control the amyloid-beta peptide-induced decrease M., Khalil, R.O., Ben-Omran, T., et al. (2012).
of glutamate uptake. J Alzheimers Dis 31, 555567. Mutations in BCKD-kinase lead to a potentially
Matos, M., Augusto, E., Santos-Rodrigues, A.D., treatable form of autism with epilepsy. Science
Schwarzschild, M.A., Chen, J.F., Cunha, R.A., 338, 394397.
and Agostinho, P. (2012b). Adenosine A2A Ortinski, P.I., Dong, J., Mungenast, A., Yue, C.,
receptors modulate glutamate uptake in cultured Takano, H., Watson, D.J., Haydon, P.G., and
astrocytes and gliosomes. Glia 60, 702716. Coulter, D.A. (2010). Selective induction of astro-
McGaraughty, S., Cowart, M., Jarvis, M.F., and cytic gliosis generates deficits in neuronal inhibi-
Berman, R.F. (2005). Anticonvulsant and anti- tion. Nat Neurosci 13, 584591.
nociceptive actions of novel adenosine kinase Pak, M.A., Haas, H.L., Decking, U.K.M., and
inhibitors. Curr Top Med Chem 5,4358. Schrader, J. (1994). Inhibition of adenosine
McQuarrie, I., and Keith, H.M. (1927). Epilepsy in kinase increases endogenous adenosine and
children:Relationship of variations in the defree depresses neuronal activity in hippocampal
of ketonuria to occurrence of convulsions in slices. Neuropharmacology 33, 10491053.
epileptic children on ketogenic diets. Am J Dis Pan, J.W., Kim, J.H., Cohen-Gadol, A., Pan, C.,
Child 34, 10131029. Spencer, D.D., and Hetherington, H.P. (2005).
Miltiadous, P., Kouroupi, G., Stamatakis, A., Regional energetic dysfunction in hippocampal
Koutsoudaki, P.N., Matsas, R., and Stylianopoulou, epilepsy. Acta Neurol Scand 111, 218224.
F. (2013). Subventricular zone-derived neural stem Papageorgiou, I.E., Gabriel, S., Fetani, A.F., Kann,
cell grafts protect against hippocampal degenera- O., and Heinemann, U. (2011). Redistribution
tion and restore cognitive function in the mouse of astrocytic glutamine synthetase in the hip-
following intrahippocampal kainic acid adminis- pocampus of chronic epileptic rats. Glia 59,
tration. Stem Cells Transl Med 2, 185198. 17061718.
Morrell, M.J. (1996). The new antiepileptic drugs Parent, J.M., and Lowenstein, D.H. (2002).
and women:Efficacy, reproductive health, preg- Seizure-induced neurogenesis: Are more new
nancy, and fetal outcome. Epilepsia 37 Suppl 6, neurons good for an adult brain? Prog Brain Res
S34S44. 135, 121131.
Morrell, M.J. (2002). Epilepsy in women. Am Fam Pignataro, G., Maysami, S., Studer, F.E., Wilz,
Physician 66, 14891494. A., Simon, R.P., and Boison, D. (2008).
Nagelhus, E.A., Mathiisen, T.M., and Ottersen, O.P. Downregulation of hippocampal adenosine
(2004). Aquaporin-4 in the central nervous sys- kinase after focal ischemia as potential endoge-
tem: Cellular and subcellular distribution and nous neuroprotective mechanism. J Cereb Blood
coexpression with KIR4.1. Neuroscience 129, Flow Metab 28,1723.
905913. Porkka-Heiskanen, T., and Kalinchuk, A.V. (2011).
Nei, M., Ngo, L., Sirven, J.I., and Sperling, M.R. Adenosine, energy metabolism and sleep homeo-
(2014). Ketogenic diet in adolescents and adults stasis. Sleep Med Rev 15, 123135.
with epilepsy. Seizure 23, 439442. Potschka, H. (2010). Modulating P-glycoprotein reg-
Nielsen, S., Nagelhus, E.A., Amiry-Moghaddam, M., ulation: Future perspectives for pharmacoresis-
Bourque, C., Agre, P., and Ottersen, O.P. (1997). tant epilepsies? Epilepsia 51, 13331347.
Specialized membrane domains for water trans- Qureshi, I.A., and Mehler, M.F. (2010). Epigenetic
port in glial cells:High-resolution immunogold mechanisms underlying human epileptic dis-
cytochemistry of aquaporin-4 in rat brain. J orders and the process of epileptogenesis.
Neurosci 17, 171180. Neurobiol Dis 39,5360.
Nishizaki, T. (2004). ATP- and adenosine-mediated Ramakers, B.P., Riksen, N.P., van den Broek, P.,
signaling in the central nervous system: Franke, B., Peters, W.H., van der Hoeven, J.G.,
Adenosine stimulates glutamate release from Smits, P., and Pickkers, P. (2011). Circulating
astrocytes via A2a adenosine receptors. J adenosine increases during human experimental
Pharmacol Sci 94, 100102. endotoxemia but blockade of its receptor does
Nishizaki, T., Nagai, K., Nomura, T., Tada, H., not influence the immune response and subse-
Kanno, T., Tozaki, H., Li, X.X., Kondoh, T., quent organ injury. Crit Care 15,R3.
418 Part IV:Homeostatic Therapies

Rash, J.E., Yasumura, T., Hudson, C.S., Agre, P., and Sirven, J., Whedon, B., Caplan, D., Liporace, J.,
Nielsen, S. (1998). Direct immunogold labeling Glosser, D., ODwyer, J., and Sperling, M.R.
of aquaporin-4 in square arrays of astrocyte and (1999). The ketogenic diet for intractable epi-
ependymocyte plasma membranes in rat brain lepsy in adults:Preliminary results. Epilepsia 40,
and spinal cord. Proc Natl Acad Sci USA 95, 17211726.
1198111986. Sorensen, A.T., and Kokaia, M. (2013). Novel
Ravizza, T., Gagliardi, B., Noe, F., Boer, K., Aronica, approaches to epilepsy treatment. Epilepsia
E., and Vezzani, A. (2008). Innate and adaptive 54,110.
immunity during epileptogenesis and spontane- Steinhauser, C., Seifert, G., and Bedner, P. (2012).
ous seizures:Evidence from experimental mod- Astrocyte dysfunction in temporal lobe epi-
els and human temporal lobe epilepsy. Neurobiol lepsy: K+ channels and gap junction coupling.
Dis 29, 142160. Glia 60, 11921202.
Reznick, R.M., and Shulman, G.I. (2006). The role of Szybala, C., Pritchard, E.M., Wilz, A., Kaplan, D.L.,
AMP-activated protein kinase in mitochondrial and Boison, D. (2009). Antiepileptic effects of
biogenesis. J Physiol 574,3339. silk-polymer based adenosine release in kindled
Roberts, D.S., Hu, Y., Lund, I.V., Brooks-Kayal, A.R., rats. Exp Neurol 219, 126135.
and Russek, S.J. (2006). Brain-derived neuro- Tani, H., Dulla, C.G., Huguenard, J.R., and Reimer,
trophic factor (BDNF)-induced synthesis of R.J. (2010). Glutamine is required for persistent
early growth response factor 3 (Egr3) controls epileptiform activity in the disinhibited neocor-
the levels of type AGABA receptor alpha 4 sub- tical brain slice. J Neurosci 30, 12881300.
units in hippocampal neurons. J Biol Chem 281, Theofilas, P., Brar, S., Stewart, K.-A., Shen, H.-Y.,
2943129435. Sandau, U.S., Poulsen, D.J., and Boison, D.
Rothstein, J.D., Patel, S., Regan, M.R., Haenggeli, (2011). Adenosine kinase as a target for thera-
C., Huang, Y.H., Bergles, D.E., Jin, L., Dykes peutic antisense strategies in epilepsy. Epilepsia
Hoberg, M., Vidensky, S., Chung, D.S., et al. 52, 589601.
(2005). Beta-lactam antibiotics offer neuropro- Tian, G.F., Azmi, H., Takano, T., Xu, Q.W., Peng,
tection by increasing glutamate transporter W.G., Lin, J., Oberheim, N., Lou, N.H., Wang,
expression. Nature 433,7377. X.H., Zielke, H.R., et al. (2005). An astrocytic
Rudzinski, L.A., and Meador, K.J. (2013). Epilepsy basis of epilepsy. Nature Medicine 11, 973981.
and neuropsychological comorbidities. Tomkins, O., Shelef, I., Kaizerman, I., Eliushin, A.,
Continuum (Minneap Minn) 19, 682696. Afawi, Z., Misk, A., Gidon, M., Cohen, A.,
Seifert, G., Carmignoto, G., and Steinhauser, C. Zumsteg, D., and Friedman, A. (2008). Blood-brain
(2010). Astrocyte dysfunction in epilepsy. Brain barrier disruption in post-traumatic epilepsy. J
Res Rev 63, 212221. Neurol Neurosurg Psychiatry 79, 774777.
Shawki, M., El Wakeel, L., Shatla, R., El-Saeed, G., Vajda, F.J.E. (2007). Pharmacotherapy of epi-
Ibrahim, S., and Badary, O. (2013). The clinical lepsy: New armamentarium, new issues. J Clin
outcome of adjuvant therapy with black seed oil Neurosci 14, 813823.
on intractable paediatric seizures:Apilot study. van Praag, H., Kempermann, G., and Gage, F.H.
Epileptic Disord 15, 295301. (1999). Running increases cell proliferation and
Shen, H.Y., Li, T., and Boison, D. (2010). A novel neurogenesis in the adult mouse dentate gyrus.
mouse model for sudden unexpected death in Nat Neurosci 2, 266270.
epilepsy (SUDEP): Role of impaired adenosine van Vliet, E.A., Otte, W.M., Gorter, J.A., Dijkhuizen,
clearance. Epilepsia 51, 465468. R.M., and Wadman, W.J. (2014). Longitudinal
Shen, H.Y., Singer, P., Lytle, N., Wei, C.J., Lan, J.Q., assessment of blood-brain barrier leakage dur-
Williams-Karnesky, R.L., Chen, J.F., Yee, B.K., and ing epileptogenesis in rats: A quantitative MRI
Boison, D. (2012). Adenosine augmentation ame- study. Neurobiol Dis 63,7484.
liorates psychotic and cognitive endophenotypes van Vliet, E.A., Zibell, G., Pekcec, A., Schlichtiger,
of schizophrenia. J Clin Invest 122, 25672577. J., Edelbroek, P.M., Holtman, L., Aronica, E.,
Shen, H.-Y., Sun, H., Hanthorn, M.M., Zhi, Z., Lan, Gorter, J.A., and Potschka, H. (2010). COX-2
J.-Q., Poulsen, D.J., Wang, R., and Boison, D. inhibition controls P-glycoprotein expression
(2014). Overexpression of adenosine kinase in and promotes brain delivery of phenytoin in
cortical astrocytes generates focal neocortical chronic epileptic rats. Neuropharmacology 58,
epilepsy in mice. J Neurosurg 120, 628638. 404412.
Shinnar, S., and Moshe, S.L. (1991). Age specificity of Vezzani, A., and Baram, T.Z. (2007). New roles for
seizure expression in genetic epilepsies. Epilepsy interleukin-1 Beta in the mechanisms of epi-
Res Suppl 4,6985. lepsy. Epilepsy Curr 7,4550.
Epilepsy 419

Vezzani, A., French, J., Bartfai, T., and Baram, T.Z. regulates glutamate transporter 1 in astrocytes.
(2011a). The role of inflammation in epilepsy. Nat Biochem Biophys Res Commun 393, 514518.
Rev Neurol 7,3140. Yellen, G. (2008). Ketone bodies, glycolysis, and
Vezzani, A., Maroso, M., Balosso, S., Sanchez, M.A., KATP channels in the mechanism of the keto-
and Bartfai, T. (2011b). IL-1 receptor/Toll-like genic diet. Epilepsia 49 Suppl 8,8082.
receptor signaling in infection, inflammation, Yerby, M.S. (2000a). Quality of life, epilepsy
stress and neurodegeneration couples hyperex- advances, and the evolving role of anticonvul-
citability and seizures. Brain Behav Immun 25, sants in women with epilepsy. Neurology 55,
12811289. S21S31; discussion S54S28.
Vezzani, A., Ravizza, T., Balosso, S., and Aronica, E. Yerby, M.S. (2000b). Special considerations for
(2008). Glia as a source of cytokines:Implications women with epilepsy. Pharmacotherapy 20,
for neuronal excitability and survival. Epilepsia 159S170S.
49 Suppl 2,2432. Zeng, L.H., Rensing, N.R., and Wong, M. (2009).
Wagner, A.K., Miller, M.A., Scanlon, J., Ren, The mammalian target of rapamycin signal-
D., Kochanek, P.M., and Conley, Y.P. (2010). ing pathway mediates epileptogenesis in a
Adenosine A1 receptor gene variants associated model of temporal lobe epilepsy. J Neurosci 29,
with post-traumatic seizures after severe TBI. 69646972.
Epilepsy Res 90, 259272. Zhang, Z.Y., Zhang, Z., Fauser, U., and Schluesener,
Waldbaum, S., and Patel, M. (2010). Mitochondria, H.J. (2007). Global hypomethylation defines a
oxidative stress, and temporal lobe epilepsy. sub-population of reactive microglia/macro-
Epilepsy Res 88,2345. phages in experimental traumatic brain injury.
Williams-Karnesky, R.L., Sandau, U.S., Lusardi, Neurosci Lett 429,16.
T.A., Lytle, N.K., Farrell, J.M., Pritchard, E.M., Zong, H., Ren, J.M., Young, L.H., Pypaert, M., Mu,
Kaplan, D.L., and Boison, D. (2013). Epigenetic J., Birnbaum, M.J., and Shulman, G.I. (2002).
changes induced by adenosine augmentation AMP kinase is required for mitochondrial bio-
therapy prevent epileptogenesis. J Clin Inv 123, genesis in skeletal muscle in response to chronic
35523563. energy deprivation. Proc Natl Acad Sci USA 99,
Williamson, A., Patrylo, P.R., Pan, J., Spencer, D.D., 1598315987.
and Hetherington, H. (2005). Correlations Zou, H., Brayer, S.W., Hurwitz, M., Niyonkuru,
between granule cell physiology and bioenerget- C., Fowler, L.E., and Wagner, A.K. (2013).
ics in human temporal lobe epilepsy. Brain 128, Neuroprotective, neuroplastic, and neurobe-
11991208. havioral effects of daily treatment with leveti-
Wu, X., Kihara, T., Akaike, A., Niidome, T., and racetam in experimental traumatic brain injury.
Sugimoto, H. (2010). PI3K/Akt/mTOR signaling Neurorehabil Neural Repair 27, 878888.
24
Traumatic BrainInjury
R A J G . K U M A R A N D A M Y K . WA G N E R

A C U T E PAT H O P H Y S I O L O G Y months after injury and result in chronic symp-


A N D A C U T E H O M E O S TAT I C toms or deficits.
DISRUPTION
Excitotoxicity
Primary Injury Mechanisms: Excitatory amino acids (EAA) such as glutamate
Contusions, Axonal Stretch/ and aspartate are important in normal excita-
Disruption tory brain function, yet in excess they can result
Traumatic brain injury (TBI) is characterized by in excitotoxicity, a phenomenon that initiates a
both a primary and a secondary injury. A pri- whole host of secondary-injury mechanisms that
mary brain injury is classified as a focal or dif- can lead to cell death. After TBI, excessive EAA
fuse injury. Focal injuries can be categorized as concentrations have been documented in cer-
lesions, contusions, lacerations, and other cir- ebrospinal fluid (CSF; Wagner et al., 2005) and
cumscribed intracranial abnormalities caused pericontusional microdialysate fluid (Bullock
by collision forces that result in tissue compres- etal., 1998; Werner and Engelhard, 2007). EAA
sion below the cranium at the site of impact and/ levels become elevated as a result of mechanical
or of tissue opposite to the impact (Andriessen axon damage and subsequent influx of EAAs
et al., 2010). The rapid acceleration/decelera- from axons. Excessive glutamate levels may
tion injury that results in diffuse axonal injury over activate specific ion channels, especially
(DAI) is a common mechanism that results the N-methyl-D-aspartate (NMDA) channel
from high-speed motor-vehicle accidents and (Bullock et al., 1998), resulting in an uncon-
creates damaging shear, tensile, and compres- trolled influx of sodium and calcium, along with
sive forces within the brain tissue (Gentry etal., associated potassium efflux, which leads to acute
1988). Naturally, the severity of DAI depends neuronal swelling and cell death. Intracellular
largely on the duration, magnitude, and direc- calcium may activate lipid peroxidases, proteases,
tion of the angular acceleration, as well as the and phospholipases that in turn facilitate the pro-
associated impact (Gennarelli, 1993). With severe duction of intracellular free radicals, which then
DAI, there can be more significant damage than leads to cell membrane and DNA damage. Also,
superficial axonal injuries. The deeper white potassium efflux will result in rapid astrocytic
matter structures, as well as midline structures swelling as cells absorb excess potassium in an
like the corpus callosum, may be also affected attempt to maintain ionic homeostasis (Bullock
(Halliday, 1999). Secondary injury is the broad et al., 1995). It is postulated that this process
characterization of multifactorial biochemical may cause rapid cytotoxic edema, a primary
cascades that lead to cellular disruption and, in factor leading to elevated intracranial pressure
severe cases, cell death of central nervous system following severe TBI. Furthermore, excessive
(CNS) tissue. These secondary-injury manifesta- EAA levels have also been implicated with mito-
tions of TBI can have a delayed clinical presenta- chondrial damage. Healthy mitochondria play
tion and result in cerebral hypotension, hypoxia, an integral role in regulating intracellular Ca2+
ischemia, excitotoxicity, hormone abnormalities, homeostasis via various mechanisms (Gunter
disruption of ion homeostasis, and CNS inflam- et al., 1994). However, excessive Ca2+ exposure
mation (Chesnut etal., 1993). These components that accompanies elevated glutamate levels dis-
of secondary injury are complex, and some can rupts brain mitochondrial electron transfer and
persist for hours to days, while others persist for energy production due to excess mitochondrial
Traumatic BrainInjury 421

calcium levels and increased reactive oxygen spe- Jeong et al., 2013). Proinflammatory cytokines
cies (ROS) production, which leads to oxidative (e.g., IL-1 and IL-6) are elevated after injury
damage, disturbance of synaptic homeostasis, through synthesis by peripheral cells and resi-
and apoptosis (Sullivan et al., 1999; White and dent glia in the CNS. These cytokines can initi-
Reynolds, 1996; Xiong etal., 1997). Mitochondrial ate inflammatory pathways and lead to increased
dysfunction also leads to energy failure after TBI, neurotoxicity via oxidative injury (Feuerstein
decreased adenosine triphosphate production, etal., 1998). Additionally, there is evidence that
and decreased oxygen consumption by 40% to cytokines can play a dual neuroprotective role
50% (Maas et al., 2008). Alterations in mito- through angiogenic, neurotrophic, and other
chondrial integrity, specifically mitochondrial mechanisms known to attenuate CNS damage
permeability transition pore translocation, are (Jeong et al., 2013; Morganti-Kossmann et al.,
a fundamental component of disrupted calcium 2002). While evidence is convincing regard-
homeostasis that results in subsequent apoptosis. ing the existence of an elevated inflammatory
The importance of this translocation with regard response acutely following TBI (Bell etal., 1997;
to neuroprotection is evidenced by the neuropro- Buttram etal., 2007; Hensler etal., 2002; Maier
tective effects of pharmaceutical agents that tar- et al., 2001; Shiozaki et al., 2005; Singhal et al.,
get the mitochondrial transition pore in animal 2002; Wang etal., 2013), the balance and nuanced
models (Mazzeo etal.,2009). role of the relatively detrimental versus neuropro-
In addition to EAA such as glutamate, dopa- tective role of various inflammatory markers is
mine (DA) is also upregulated following TBI still largely unclear. Some studies have suggested
through increased tyrosine hydroxylase, the a dose-related effect (Wang etal., 2013). That is,
rate-limiting enzyme involved with DA synthesis there are potential benefits of cytokines and other
(Kobori etal., 2006; Yan etal., 2001). DA facili- inflammatory markers when under the control of
tates the development of acute cellular pathway physiological regulatory mechanisms; however,
dysfunction, including Na+/K+ ATPase, cell extremely high levels may be harmful. There
metabolism, Ca 2+ release, and the NMDA recep- are also time-dependent effects of inflammatory
tor through cAMP-regulated phosphoprotein markers following injury. A recent TBI clinical
32 kDa and protein phosphatase-1 (Bales et al., study (Johnson et al., 2013) identified activated
2009; Greengard etal., 1999; Nishi etal., 2002). microglia and phagocytic macrophages in exten-
Additionally, high synaptic DA levels can be rap- sive regions of the corpus callosum up to 18years
idly oxidized to form reactive DA semiquinones following a single TBI. Work from our group
(Hastings, 1995). Oxidized DA can generate suggests that a chronically sustained elevations
hydrogen peroxide and superoxide, which can in IL-1, IL-6, IL-8, IL-10, and TNF in the first
lead to significant oxidative stress (Bales et al., 3months after injury increases the risk for worse
2009). Interestingly, DAergic fibers can also mod- global outcomes at 6 and 12months post-injury
ulate striatal glutamate excitotoxicity (Chapman (Kumar etal., 2014). Posttraumatic inflammation
et al., 1989; Filloux and Wamsley, 1991), and is an important clinical consideration following
depleting DAergic projections into the striatum injury, and clinicians and researchers should fur-
prior to ischemic injury can be neuroprotective ther examine the balance between the neuropro-
(Globus etal.,1987). tective versus the detrimental effects of elevated
inflammatory states.
Inflammation
The CNS historically has been considered an BBB Disruption and Reactive
immunologically privileged organ due to the Astrocytosis
physical separation of the brain from the periph- Experimental animal models of TBI demonstrate
ery with the blood-brain barrier (BBB). In TBI, that BBB permeability is increased following
BBB integrity is acutely compromised. TBI is injury (Shapira etal., 1993). The BBB breakdown
characterized by a significant inflammatory is understood to be biphasic in nature, with rapid
response that includes not only CNS-mediated deterioration within the first few hours followed
inflammatory markers but also peripheral by subsequent improvement in integrity start-
inflammation (Schmidt etal., 2005). Among the ing 3 to 7 days after injury (Shlosberg et al.,
key mediators of the inflammatory response are 2010). In human TBI, BBB permeability usually
proinflammatory cytokines, chemokines, and returns to normal within days to weeks after
cell adhesion molecules (Ghirnikar et al., 1998; injury (Kirchhoff etal., 2006; Stahel etal., 2001).
422 Part IV:Homeostatic Therapies

BBB deterioration initiates soon after the trau- studies show that ischemia is a significant prob-
matic insult through shearing of endothelial cells lem in the first 24 hours after injury, and the
located in brain microvessels (Rodrguez-Baeza volume of ischemic brain is correlated with
et al., 2003). This phenomenon affects normal worse global outcomes at 6months (Coles etal.,
BBB regulation, cerebral blood flow (CBF), and 2004). Among the principle causes of cerebral
metabolic processes that can result in more ischemia are systemic arterial hypotension, ele-
serious secondary complications (Shlosberg vated intracranial pressure, edema, blood carbon
et al., 2010), such as ischemia that induces tis- dioxide concentrations, focal tissue compres-
sue hypoxia (Fischer etal., 2002). Other mecha- sion, and microvascular disease (Hekmatpanah
nisms of BBB disruption and ischemia following and Hekmatpanah, 1985; Pfenninger etal., 1989;
TBI may include cerebral vasospasm (Lee etal., Sarabia et al., 1988; Obrist et al., 1984). Since
1997), cerebral pressure autoregulatory dysfunc- the brain is so susceptible to ischemic events, it
tion (Rangel-Castilla etal., 2008), and coagulop- is vital to maintain the CBF and cerebral oxy-
athy (Nekludov etal., 2007). The resultant effects gen metabolism during acute care manage-
of BBB breakdown influence other downstream ment. Correspondingly, the The Guidelines for
secondary-injury cascades, including edema, the Management of Severe Head Injury (Brain
inflammation, and cell death. The BBB is a com- Trauma Foundation, 2007) for acute TBI are
ponent of the larger neurovascular unit that particularly focused on treating hypotension
also includes astrocytes, pericytes, microglia, and hypoxia, as it is estimated that nearly 90% of
neurons, and the extracellular matrix (Shlosberg those who die following TBI show some evidence
etal., 2010). Physiologically, these units perform of ischemia (Greve and Zink,2009).
many homeostatic functions that influence neu-
ronal survival. Specifically, astrocytes maintain Potential Acute Neuroprotectants
extracellular ion concentrations, influence glu- The purine ribonucleoside adenosine can have
tamate clearance, and produce inflammatory a beneficial influence on secondary-injury cas-
cytokines and chemokines (Chen and Swanson, cades in TBI (Headrick et al., 1994), including
2003). Reactive astrocytes can play a damaging improved Ca2+ homeostasis, attenuated EAA
role after CNS injury by facilitating the develop- release, and reduced oxidative stress. Adenosine
ment of glial scarring (Fawcett and Asher, 1999). controls many functions through coupling of
However, experimental TBI work by Myer and either inhibitory (A1, A3) or stimulatory (A 2a, A 2b)
colleagues (2006) showed that reactive astrocyto- G-protein receptors and the spatial distributions
sis also helps preserve neural tissue and reduces of these receptors within the brain (Fredholm
excess inflammation following moderate injury. etal., 2005). Adenosine binding to either A1 or A 2
receptors affect glutamate and DA neurotrans-
Ischemia and OxidativeStress mission (Boison, 2008). One animal study mod-
TBI also initiates the development of oxidative eling status epilepticus found that A1-receptor
stress, which refers to the generation of ROS. It activation was a key mediator in reducing neu-
is well established that ROS can lead to oxidative ronal cell loss and convulsions (Fedele et al.,
stress and resultant damage to lipids, proteins, 2006). Another animal study found that A 2
and nucleic acids (Halliwell, 1991). Further, ROS receptor inhibitors can have potent neuroprotec-
have been associated with TBI-related patho- tive properties by preventing apoptosis and syn-
genesis, including tissue loss (Shao etal., 2006), aptotoxicity (Silva etal.,2007).
reduced synaptic integrity (Ansari et al., 2008), Neurotrophic factors, including brain-derived
and cytoskeletal and mitochondrial damage neurotrophin factor (BDNF) and nerve-growth
(Singh etal., 2007). Several animal studies have factor, have important roles in cell differentia-
shown evidence for the neuroprotective proper- tion, growth, proliferation, and CNS cell survival
ties of antioxidant agents, such as propofol and (Hetman etal., 1999; Sofroniew etal., 2001). After
erythropoietin, in both lowering the extent of TBI, neurotrophins in both CSF (Chiaretti etal.,
oxidative injury (Ozturk et al., 2005, 2008) and 2008)and serum (Kalish and Phillips, 2010)are
reducing neuronal apoptosis (Liao etal.,2008). depressed in association with the severity of
The brain is vulnerable to the development of injury. There is some evidence that mesenchymal
cerebral ischemia as a part of secondary injury. stem cells have the capacity to upregulate neuro-
Soon after impact, CBF regulation and cerebral trophic factors and have shown some ability to
oxygen metabolism are impaired. Imaging enhance recovery in the acute stages following
Traumatic BrainInjury 423

TBI (Kim et al., 2010). However, there is some (Wagner et al., 2011b). Specifically, individuals
recent biomarker evidence that neurotrophins with persistently higher levels in the first week
are not neuroprotective early after injury, par- were at risk for poor outcome at 6 and 12months;
ticularly for older individuals with severe TBI however, the group whose BCL-2 levels started
(Failla etal., 2015a; Failla etal., 2015b). near control levels in the first few days after injury
Furthermore, acute TBI triggers a significant but showed a delayed rise in BCL-2 had the best
stress response that is not limited to the CNS. recovery, suggesting a prosurvival upregulatory
Work from our lab suggests that endogenous mechanism after severe TBI. Genetic variation
serum estradiol levels are highly associated with in BCL-2 may also influence outcome after TBI
mortality (Wagner et al., 2011a), though higher (Hoh etal.,2010).
estradiol/testosterone ratios in CSF may be neuro-
protective (Garringer etal., 2013). Accumulating Acute Pathophysiology
evidence in trauma and critically ill populations Integrative Summary
suggests that, in humans, serum estradiol is asso- Acute TBI is characterized by direct disruption of
ciated with a stress response that carries a higher brain tissue homeostasis, including alterations in
risk for mortality (May etal., 2008; Spratt etal., CBF, cerebral vasospasm, massive excitotoxicity
2006, 2008). Work from our lab also shows cortisol and oxidative stress, inflammation, and edema.
is elevated in the CSF (Santarsieri etal., 2014)and Experimental studies have provided the field
serum (Wagner etal., 2011a) acutely after injury. with a better understanding of acute TBI and
Clinical data show that elevated CSF cortisol is the secondary-injury cascade. However, as Maas
associated with poor outcomes (Santarsieri etal., and colleagues (2010) explain, clean experimen-
2014). Interestingly, glucocorticoid and corticos- tal conditions do not seem to transfer into the
teroid drugs, which were once commonly used dirty clinical situation. Clinical TBI represents
in TBI medical management, have fallen out of a complex spectrum and heterogeneous range
favor due to convincing evidence from clinical of pathologies that make a single golden bul-
trials that their use leads to increased mortal- let neurotherapy impractical (Beauchamp etal.,
ity rates (Alderson and Roberts, 2005; Edwards 2008). Due to patient heterogeneity, it is difficult
et al., 2005; Roberts et al., 2004). Importantly, to gauge whether targeted pathophysiological
estradiol and cortisol levels are dependent on processes associated with a given neurotherapy
progesterone, and temporal elevations in serum have the same effect in all individuals. Also then,
estradiol and serum/CSF cortisol after TBI a one-size fits all approach to clinical interven-
are likely the product of adrenal progesterone tions is not likely to be successful. Some com-
production. Due to the well-established neu- pounds thought to be neuroprotective may not
roprotective properties of progesterone in exper- be beneficial for all individuals after TBI. Thus
imental models (Stein, 2011), clinical research there is a continued need for well-designed clini-
efforts have targeted progesterone therapies cal characterization studies to further evaluate
(Wright et al., 2007). However, recent phase III heterogeneity involved with the acute pathophys-
multisite clinical trials have failed to show a ben- iological events associated with secondary injury
eficial effect when progesterone was given early in humans. This information should be used to
after moderate to severe TBI (Skolnick et al., guide individualized treatment approaches and
2014; Wright etal., 2014). Additional evaluation timelines as well as to understand how pleio-
of how progesterone treatment impacts homeo- tropic treatments or treatment combinations may
static disruption of physiological cortisol and confer therapeutic benefit.
estradiol production after severe TBI may aid
in identifying whether some subpopulations do T B I PAT H O L O G Y A N D
derive benefit from progesterone as a neuropro- BIOSUSCEPTIBILITY
tective treatment. TOCHRONIC CONDITIONS
Another consideration with acute TBI neu-
roprotection is the protein beta-cell lymphoma 2 Common Mechanisms ofInnate
(BCL-2). This anti-apoptotic factor works to pre- Homeostatic Heterogeneity
vent the mitochondrial cytochrome C release and To date, few TBI preclinical and clinical trials
subsequent caspase-mediated cell death. Data incorporate factors such as sex, age, baseline
show that acute CSF levels of BCL-2 have strong behavioral function, and variable standard care
prognostic potential as a marker of cell death therapeutics in their trial design. A few studies
424 Part IV:Homeostatic Therapies

that have specifically examined sex effects have gene (SLC6A4) variation, a common depression
observed significant sex-specific differences in risk allele, and the development of posttraumatic
treatment response. For example, both experi- depression (PTD; Failla etal., 2013). Specifically,
mental and clinical studies show women may not after TBI, a history of premorbid mood disor-
benefit from a treatment such as hypothermia as der and SLC6A4 genotype differentially influ-
much as men with similar injuries (Bayir etal., ences PTD risk. As an additional methylation
2004; Suzuki etal., 2003; Wagner etal., 2005). TBI site exists for SLC6A4 S-allele carriers compared
populations are also heterogeneous with respect to L-carriers (Failla etal., 2013), we hypothesize
to baseline prognostic potential for recovery and that injury-specific differences in SLC6A4 meth-
risk for complications. Factors such as genetics, ylation may be one contributor to differences in
propensity for autoimmune responses, brain PTD risk over time. The precise role of epigenetic
plasticity, and atrophy are highly specific to indi- modifications in injury and recovery, as well as
viduals and play an integral role in homeostatic how injury-induced epigenetic modifications
restoration and susceptibility to common post- drive homeostatic disruption and normalization,
traumatic chronic conditions such as epilepsy, is still largely unknown. However, rehabilitation
cognitive dysfunction, depression, migraine, and strategies after TBI may be potent modifiers of
neuroendocrine dysfunction. This section exam- the epigenetic remarking process and an impor-
ines these common TBI phenotypes in the con- tant mechanism by which rehabilitation strate-
text of innate patient heterogeneity. gies impact recovery.

Genetics/Epigenetics Autoimmunity
Increasing evidence supports the role of genetic Another source of homeostatic heterogene-
variation in influencing pathophysiological ity is autoimmunity. Antibodies specific for
mechanisms and outcome (Bonneh-Barkay etal., self-antigens are involved in normal physiologi-
2010; Diamond et al., 2014; Failla et al., 2013; cal conditions. These immunoglobulins facili-
Hoh etal., 2010; Miller etal., 2010; AK Wagner tate removal of dead cells through phagocytosis
et al., 2010). An overall understudied area to in a process known as opsonization. After TBI,
date in brain injury, genetics, could influence due to BBB disruption, antibodies are prevalent
secondary-injury mediators (Garringer et al., in greater quantities in the brain (Baloyannis
2013) and processes (Conley et al., 2014), and and Gonatas, 1979) and resident brain cells are
factors such as sex could interact with genetics exposed to greater quantities of autoantibod-
to moderate acute secondary-injury cascades, ies (Soares etal., 1995). Damaged CNS cells can
such as with acute DA dysfunction (Wagner release antigens that enter the blood circulation
etal.,2007). following BBB breach that can trigger autoanti-
The emerging field of epigenetics, which body production. While theoretically contrib-
refers to genetic control by factors other than an uting to heterogeneity with secondary injury
individuals DNA sequence, can influence how and recovery, the complexity and consequences
genes turn on and off to determine which pro- (positive or negative) of autoantibody produc-
teins are transcribed. Specifically, initial work tion following TBI is still unclear. One study,
suggests injury-induced epigenetic changes in using an experimental injury model, identified
the brain may affect TBI-related pathology and a neuroprotective role for circulating immuno-
contribute to heterogeneity in recovery and bio- globulin G autoantibodies to bind to dying neu-
susceptibility to complications after TBI, (Gao rons that were proximal to the injury site after
etal., 2006; Lundberg etal., 2009; Schober etal., TBI (Stein et al., 2002). Also, there is evidence
2012; Wang etal., 2011; Zhang etal., 2007). Two that S100b can be released from brain astrocytes
primary epigenetics mechanisms relevant to and contribute to an autoimmune response,
TBI include histone posttranslational modifica- and S100b has been shown to be a reliable bio-
tions and DNA methylation (Jaenisch and Bird, marker after sports-related concussions (Marchi
2003); injury-induced changes in these post- et al., 2013). A recent study also found that the
translational modifications may underlie some antiglial fibrillary acidic protein autoantibody
of the temporally dynamic genetic associations can enter living astroglial cells and compromise
that have been observed after TBI. For example, glial cell health, and an increase in these levels
work from our lab shows a temporally dynamic were negatively correlated with outcome (Zhang
association between the serotonin transporter etal., 2014). Also, a clinical TBI study examined
Traumatic BrainInjury 425

antipituitary antibodies and found evidence for 1993). BDNF, a neurotrophin ubiquitous in the
the role of autoimmunity in TBI-induced pitui- hippocampus, is critical in synapse maintenance,
tary dysfunction (Taniguchi etal., 1999). Because particularly with LTP, via activity-dependent
of the latent development period required for secretion of BDNF (Kovalchuk et al., 2002).
autoantibody production (Ritter et al., 2014), it Despite the possibility that BDNF is not directly
is possible that autoimmunity could interplay neuroprotective early after TBI (Failla et al.,
with other long-term degenerative (Ghirnikar 2015a; Failla etal., 2015b), it may benefit recov-
et al., 1998) and inflammatory (Schmidt et al., ery over the long term through its role in LTP,
2005) processes thought to contribute to sus- where BDNF may be an underlying substrate that
tained disruption of homeostasis and poor recov- facilitates persistent long-term memory storage
ery afterTBI. (Bekinschtein etal., 2008a, 2008b). Hippocampal
BDNF is chronically decreased in TBI (Chen
Plasticity et al., 2005), and there is evidence that thera-
Brain plasticity following TBI can take on many pies thought to increase BDNF expression in the
forms, from synaptic strength alterations to reor- brain, such as environmental enrichment (Chen
ganization of functional brain networks. Neural etal., 2005)and exercise (Griesbach etal., 2009),
networks that were present prior to injury are are promising therapies for cognitive recovery
often altered in a dynamic and heterogeneous post-TBI. Hippocampal BDNF expression is
way during recovery. In one study utilizing mag- linked to spatial memory in experimental TBI
netoencephalography, an imaging method that studies (Griesbach etal., 2009). Thus BDNFs role
utilizes electromagnetic fields that are generated in LTP may be important in the critical balance
by the net effect of the slow ionic flow of networks of synaptic maintenance in cognitive recovery.
of neurons, researchers showed that synchrony in Importantly, there are differential expression
neural signals mirrored cognitive recovery fol- patterns of BDNFs target receptors with age
lowing a brain injury (Castellanos et al., 2011). (Croll etal., 1998; Tapia-Arancibia etal., 2008)in
Functional magnetic resonance imaging studies both the prefrontal cortex (Romanczyk et al.,
suggest that considerable functional reorganiza- 2002) and the hippocampus (Webster et al.,
tion occurs following TBI. Studies show evidence, 2006). Tropomyosin-related kinase B expres-
using working memory paradigms, of increased sion peaks in young/middle adulthood and then
cerebral activation at lower workloads, even when decreases with age (Romanczyk etal., 2002). Thus
actual performance does not differ from controls an age-specific shift in BDNF receptor expression
(McAllister etal., 1999). Other studies have sug- could impact BDNF signaling, complicating the
gested altered hemispheric recruitment patterns application of therapies that target BDNF. Age
(Kasahara et al., 2011; Russell et al., 2012) in appears to be a major factor in how BDNF genet-
task-based brain activations following TBI that ics and levels early after TBI influence recovery
are not observed in control subjects. In addi- (Failla etal., 2015a; Failla etal., 2015b). The mod-
tion to reorganization, neurogenesis contributes ifying effect of age on the relationship between
to plasticity following TBI. Adult neurogenesis, chronic BDNF and outcome remains in question.
especially within the hippocampus, is a tightly
controlled process from proliferation of neural Neurodegeneration and Atrophy
stem cells to differentiation, migration, and inte- Cerebral atrophy is a widespread phenomenon
gration into long-range connections. Following in TBI (MacKenzie etal., 2002). Recent advances
experimental TBI, there is a well-documented in diffuse tensor imaging and related tools pro-
increase in neurogenesis immediately follow- vide valuable information regarding changes in
ing both focal and diffuse TBI (Bye etal., 2011). white matter integrity following DAI (Huisman
A loss of neuronal integrity in fronto-limbic et al., 2004). The cingulate gyrus, uncinate fas-
regions following TBI may facilitate neurogen- ciclus, and superior longitudinal fasciculus have
esis that is highly linked to chronic BDNF levels all been related to cognitive deficits post-TBI
(Rossi etal.,2006). (Turken et al., 2008) and are implicated in cir-
Plasticity-dependent alterations that affect cuits that facilitate depression (Hamani et al.,
recovery and homeostatic function are also man- 2011; Mayberg, 2003; Mettenburg et al., 2012).
ifested in the context of hippocampal long-term In TBI, a few studies have examined regionally
potentiation (LTP), which is critical for learning specific atrophy in fronto-limbic regions such
and memory formation (Bliss and Collingridge, as the hippocampus (Tate and Bigler, 2000)and
426 Part IV:Homeostatic Therapies

the anterior cingulate (Chen etal., 2008). Recent postmortem brain examination; therefore the
work has shown in moderate to severe TBI that prevalence of the disorder is unknown.
the degree of fronto-limbic structural atrophy in
the hippocampus and other regions was associ- Epilepsy
ated with PTD (Hudak etal., 2011). Interestingly, Posttraumatic epilepsy (PTE) is a common com-
these authors also show that regional grey mat- plication following TBI, with a strong relation-
ter atrophy is closely linked to damaged white ship between seizure risk and severity and type
matter tracts (Warner et al., 2010), suggesting of head injury (closed or penetrating; Agrawal
overall reductions in fronto-limbic connectiv- etal., 2006; Annegers etal., 1998). Importantly,
ity can occur after TBI. While BDNF may also one study found that over 80% of patients that
influence white matter tracts and functional have a seizure post-TBI will have a recurrent sei-
fronto-limbic circuits, these relationships are less zure within one year (Haltiner etal., 1997). The
clear (Egan et al., 2003; Kennedy et al., 2009). risk factors for epileptogenesis following TBI
Fronto-limbic connectivity and regional volumes likely involve multiple mechanisms; those with
have been linked to genetic variation in several a subdural hematoma or brain contusion are at
studies examining healthy populations, as well an increased risk (Annegers etal., 1998; Haltiner
as those with major depressive disorder (MDD), etal., 1997). Acommonly implicated pathology
and results also implicate gene variation within in both TBI and epilepsy is glial scarring, which
monoaminergic systems (Bartrs-Faz etal., 2002; is connected to microglial activation follow-
Bertolino etal., 2009; Honea etal., 2009; Pacheco ing initial insult. One study found that, after
etal., 2009; Pezawas etal., 2005)and neurotro- TBI, IL-1, and the granulocyto-macrophage
phins (Egan et al., 2003; Kennedy et al., 2009; colony-stimulating factor work to regulate glial
Kim et al., 2013; Montag et al., 2010; Pezawas scarring at the site of focal injury (Giulian etal.,
etal., 2008; Radua etal.,2014). 1994). In epilepsy, activated microglia, proin-
Another consideration regarding neurode- flammatory cytokines, and leukocytes from
generation in the context of TBI is chronic trau- the brain and periphery initiate inflammatory
matic encephalopathy (CTE), which has gained cascades that result in neuronal hyperexcitabil-
mainstream media attention due to football ity, which can result in seizures (Vezzani and
players in the National Football League. CTE is Friedman, 2011). The role of inflammation in
a progressive tauopathy that results from repeti- epilepsy is well established in non-TBI popula-
tive mild TBI or concussion that is traditionally tions. Evidence from our group shows that CSF/
associated with athletes and military service serum ratios of IL-1 were associated with PTE
members that are at high risk for repetitive brain risk over time, and IL-1 gene variation moder-
trauma (McKee et al., 2013). This pathology is ated serum IL-1 levels in this study (Diamond
characterized by an accumulation of hyperphos- et al., 2014). In addition to immunologically
phorylated tau abnormalities that begin focally, based mechanisms of epileptogenesis, reduction
as neurofibrillary and astrocytic tangles that in bilateral hippocampal volume, specifically
spread to adjacent cortices, and eventually leads in the dentate gyrus, is associated with TLE
to extensive neurodegeneration affecting many (Golarai etal., 2001). It is also postulated that,
brain regions, including the medial temporal lobe similar to TBI, excessive production of excita-
structures, diencephalon, and brainstem (Stein tory amino acid receptors during seizures can
et al., 2014). Importantly, the tauopathy asso- generate ROS and nitric oxide that contrib-
ciated with CTE is believed to be unique from ute to oxidative stress (Lancelot et al., 1998).
other tauopathies such as Alzheimers disease. These mechanisms can also lead to changes in
McKee and colleagues (2013) have character- excitatory/inhibitory neurotransmitter homeo-
ized progressive staging of CTE pathology from stasis, including EAAs, such as aspartic acid,
I to IV (least to most progressive) based on the that can have significant effects on epileptogen-
density of neurofibrillary tangles and coverage in esis (Agrawal et al., 2006; Janjua et al., 1990).
regions of the brain. Despite the established link Further, other studies are emerging suggesting
between repetitive blows to the head and CTE that genetic variation within neurotransmit-
incidence, the contribution of age, sex, genetics, ter signaling pathways can moderate seizure
stress, and substance abuse to disease pathophys- susceptibility in the context of TBI (Darrah
iology is currently unknown (Stein etal., 2014). et al., 2013; Diamond et al., 2014; AK Wagner
To date, CTE has been diagnosed only through etal.,2010).
Traumatic BrainInjury 427

Cognitive Dysfunction injury recovery does not coincidentally paral-


Monoaminergic Neurotransmission lel resolution of diaschisis. Further, much of the
Acute brain trauma can result in damage to the work done in clinical rehabilitation is guided by
hippocampus (Hicks etal., 1993), frontal cortex the framework of reducing neural depression and
(Dixon etal., 1987), and striatum (Dietrich etal., diaschisis (Garcia etal.,2011).
1994). These brain regions all share the common
characteristic of involvement in DA circuitry Depression
through interactions with glutamate (Centonze Neurotrophic Support
etal., 1999; Granado etal., 2008; OCarroll etal., In healthy individuals, serum BDNF has been
2006). The loss of DAergic fibers after injury implicated as a sensitive biomarker to depres-
specifically in the ventral tegmental area and sive symptoms associated with MDD (Sen etal.,
substantia nigra can influence synaptic and den- 2008). Specifically, it is known that both stress
dritic integrity in the frontal cortex and striatum and TBI are associated with attenuated hip-
(Bales etal., 2009). Also, after brain injury there pocampal BDNF expression (Marmigre et al.,
is a rapid increase in tyrosine hydroxylase (Yan 2003; Murakami et al., 2005).Hippocampal vol-
et al., 2001; Yan et al., 2007), an enzyme neces- ume is also reduced following TBI (Jorge et al.,
sary for DA synthesis. Not surprisingly, increased 2007), suggesting that BDNF expression may be
tyrosine hydroxylase expression is associated involved in PTD pathophysiology. While there is
with acute increases in DA levels in multiple evidence for the effectiveness of serum BDNF as
brain regions (Massucci et al., 2004; McIntosh a biomarker for MDD development (Hashimoto,
etal., 1994); however, over time, DA levels appear 2010; Karege et al., 2002; Sen et al., 2008), it
to be reduced in the chronic period after TBI remains unclear whether or not serum BDNF
(Massucci et al., 2004). This phenomenon may is a viable marker for depression in TBI patients
reflect more of a functional hypoDArgic state and its potential as a biomarker in the context of
(Wagner et al., 2009) rather than frank DA cell homeostatic dysfunction, plasticity, and recovery.
loss. Notably, low DA levels have been associated Further study on BDNF links to depression path-
with schizophrenia (Goldberg et al., 1991) and ways may identify potential therapeutic targets to
Parkinsons disease (Bernheimer et al., 1973), treat PTD. There is also some suggestion that the
and elevated levels have been associated with depression-risk polymorphism BDNF val66met
deficits in learning and memory (Morrow etal., could mediate the relationship between BDNF
2000). Numerous clinical studies of DAergic ago- levels and depression (Gatt etal., 2009), with one
nists (discussed specifically in greater detail later TBI study finding that the val66met SNP (specifi-
in this chapter) have shown efficacy for treating cally Val/Val homozygotes) influences treatment
cognitive deficits afterTBI. responses to antidepressants for PTD (Lanctt
etal.,2010).
Diaschisis
As previously discussed, secondary brain injury Inflammation
is characterized by significant alterations in It is well known individuals with TBI are more
brain neuroplasticity, neurotransmitter activity, likely to have elevated inflammatory cytokines
and other cellular dysfunctions. Translational and their receptors in both the peripheral blood
therapeutic neurorepair and rehabilitation and CSF (Kumar etal., 2014; Miller etal., 2009).
approaches are intended to limit destructive One of the most commonly occurring neurobe-
secondary-injury cascades and attenuate post- havioral complications after TBI is PTD, with
traumatic regional neurodepression that leads TBI survivors at a nearly 10 times increased risk
to diaschisis. Diaschisis refers to functional compared to the general population (Bombardier
depression of neurons remote from, but ana- et al., 2010). However, the pathophysiological
tomically connected to, the primary site of injury mechanisms underlying PTD development are
(Von Monakow, 1969). Mechanisms of recov- poorly understood. The literature provides a
ery from diaschisis, by nature, often result from growing body of evidence demonstrating a rela-
restoration of homeostatic functions after TBI, tionship between inflammation and depression
including normalization of cerebral metabolism outside the context of TBI. Interestingly, MDD is
and restoration of neurotransmitter systems of more prevalent in patients with conditions that
brain plasticity (Duffau, 2006)and cortical reor- lead to chronic inflammation (e.g., cardiovas-
ganization (Kleim et al., 2002). However, brain cular disease, type II diabetes, and rheumatoid
428 Part IV:Homeostatic Therapies

arthritis) compared to the general population Headache and Migraine


(Steptoe, 2007). Due to the well characterized Posttraumatic headache (PTH) is the most com-
role of inflammation in both TBI and depression monly reported symptom following TBI, with
pathology, respectively, it is reasonable to suggest prevalence estimates ranging from 30% to 90%
a potential role for inflammation in PTD devel- (Lew etal., 2006; Theeler etal., 2013). When PTH
opment. Work from our group shows that acute persists for over 1month after injury, migraine/
elevations in the CSF cytokine soluble surface probable migraine is the most likely headache
markers sVCAM-1, sICAM-1, and sFAS conferred phenotype, occurring in nearly 40% of those
a nearly four-fold increase in odds of developing who report PTH symptoms (Lucas et al., 2012).
PTD 6months post-injury (Juengst etal., 2014). Among those with a preexisting history of head-
Despite this promising initial finding, there is aches, PTH is diagnosed when symptoms are
still uncertainty regarding the temporal rela- increased or intensified soon after the primary
tionship between inflammation and PTD, such injury (Lucas, 2011). Pain following posttrau-
as the impact of chronic serum cytokine eleva- matic migraine (PTM) likely is due to neuro-
tion (Kumar et al., 2014), the role of peripheral genic inflammation (Moskowitz, 1993). Often
inflammation, and examination of inflammatory referred to as the trigeminovascular pathway,
cells such as microglia and leukocytes. Given the this pathway involves the release of neuropep-
lack of unequivocal evidence of the use of selec- tides, such as substance P, neurokinin A, and
tive serotonin reuptake inhibitors (SSRIs) in PTD calcitonin gene-related peptide, near sensory
treatment (Fann etal., 2009), further work in this fibers innervating the meninges, where they can
area could have important implications for iden- sensitize peripheral nociceptors (Longoni and
tifying therapeutic targets that restore a physi- Ferrarese, 2006; Williamson and Hargreaves,
ological inflammatory environment afterTBI. 2001). In TBI, neurogenic inflammation has been
connected to the development of edema, vaso-
Serotonergic Dysfunction dilation, and functional deficits (Nimmo et al.,
In addition to neurotrophins and inflamma- 2004). Cortical spreading depression has been
tory markers, serotonin signaling has been also observed in brain injury (Lauritzen et al., 2011;
implicated with the development of depression Strong etal., 2002)and is a phenomenon closely
(Krishnan and Nestler, 2008). Serotonergic neu- tied to migraine pathology, specifically migraine
rons have widespread connections that impact aura (Hadjikhani etal., 2001). This phenomenon
both mood and cognition. Evidence from experi- involves an intense depolarization of neuronal
mental TBI models suggests that there is an acute and glial membranes that results in acute changes
elevation of serotonin, or 5-hydroxytryptamine in [K+] and [Na+] that leads to neuronal excitabil-
(5-HT), levels followed by a chronic hypomono- ity (Kraio and Nicholson, 1978). Individuals with
aminergic state (Bales et al., 2009; Busto et al., PTM are at a greater risk for brain infarcts and
1997). Therapeutic targets of the 5-HT (1A) white matter lesions compared with the general
receptor confer neuroprotective effects, leading population (Kruit MC et al., 2004). Some have
to improvements in cognitive-related outcomes proposed that alterations in cerebral autoregula-
and a reduction in hippocampal cell loss (Kline tion can influence the ischemic lesions (Reinhard
etal., 2002). However, SSRI treatment after TBI et al., 2007). Interestingly, during migraine
does not significantly improve cognitive symp- attacks there is an increase in CBF in cingulate,
toms or depressive symptomology (Wang et al., auditory, and visual cortices and also the brain
2011). The reason for this relatively counterintui- stem (Weiller etal., 1995). Despite the possibility
tive finding is unclear. Experimental work with that it is likely a product of neurogenic inflam-
SSRIs have also failed to yield a benefit in behav- mation and autoregulatory dysfunction, the
ioral outcomes, including cognitive performance, mechanisms underlying PTM remain under-
despite some seemingly beneficial effects on neu- studied todate.
rogenesis (Wang etal., 2011; Wilson and Hamm, Migraine headaches have also been linked
2002). As previously elaborated in this chapter, it with 5-HT transmission (Raskin, 1991). In
is possible that genetic variability within the ser- fact, many drugs effective in treating migraine,
otonergic pathway influences PTD risk, and this directly or indirectly, influence 5-HT transmis-
source of innate heterogeneity in TBI response sion or receptor functioning (Raskin, 1991).
could potentially influence the response to thera- Triptans, a class of 5-HT-1B/D agonist drugs, are
peutics (Failla etal.,2013). commonly used for acute management of PTM.
Traumatic BrainInjury 429

They work by binding serotonin receptors in mediate an anti-inflammatory immune response


meningeal trigeminal afferents and mediate neu- in the CNS could be responsible for the pro-
rogenic inflammation. They also can constrict longed inflammatory response observed after
vessels dilated by calcitonin gene-related peptide TBI (Besedovsky and del Rey, 2000; Sapolsky
(Lucas, 2011). For chronic management of PTM, et al., 2000). To address this point, work from
the most common therapies are tricyclic antide- our group specifically studied the bidirectional
pressants, -blockers, calcium-channel block- neuroendocrine-immune relationships in the
ers, and antiepileptic drugs (AEDs). Therapies context of TBI. We found that cortisol medi-
that are less commonly indicated are SSRIs or ated the relationship between CSF inflammation
serotoninnorepinephrine reuptake inhibitors and global outcomes (Santarsieri et al., 2014).
(Lucas, 2011). There is no universal treatment Correlational analysis indicated that, among
protocol for individuals presenting with PTM. those in a low cortisol state, individuals with
Overall, management of headache disorders after unfavorable outcomes display a negative correla-
TBI requires a balance of treatment of physical tion between CNS inflammatory load and corti-
headache or migraine symptoms and comorbid sol. However, those with a high cortisol state and
cognitive, emotional, and sleep disorders. unfavorable outcomes have a significant positive
correlation between cortisol and CNS inflamma-
Neuroendocrine Dysfunction tion. These findings suggest that poor outcomes
A majority of patients with moderate to severe after TBI may be the result of a dysfunction in
TBI also have some kind of pituitary dysfunction neuroendocrine-immune relationships where
comorbidity that results in hypogonadism (Agha too much, and also too little inflammation can
et al., 2004; Lieberman et al., 2001; Schneider have a negative effect on outcome.
etal., 2006; J.Wagner etal., 2010). Work from our
lab shows that acute hypogonadism affects nearly PL E IO T ROPIC , E N V I RON M E N TA L ,
all cases of moderate to severe TBI. Also, among A N D I N DI V I DUA L I Z E D
TBI cases with acute hypogonadotropic hypo- I N T E RV E N T ION ST R AT EG I E S:
gonadism, approximately 40% go on to develop HOM EO STAT IC- V E R SUS
persistent hypogonadotropic hypogonadism PL A ST IC I T Y-SU PP OR T E D
(PHH), while the remainder have early-resolving M EC H A N I SM S
hypogonadotropic hypogonadism (Wagner etal.,
2012). Additional work from our group shows Common Considerations
that in a prospective cohort of men with severe forTreatment
TBI, a two-step screening of testosterone at 12 to To date, pharmacological drugs to treat TBI have
16 weeks post-injury can predict persistent hypo- failed to show efficacy due to the high degree
gonadotropic hypogonadism status over the first of heterogeneity and complicated pathophysi-
year with high sensitivity and specificity (Barton ology that accompanies the injury. Therapies
etal.,2015). that target only one physiological mechanism
Stress-related models have shown that eleva- largely have not conferred an overall benefit
tions in cortisol levels can also contribute to (Maas etal., 2010). As such, the reality is that a
hypogonadism (Kalantaridou etal., 2004), which single golden bullet treatment does not exist
could be a primary reason why acute serum hor- for TBI. Health-care providers face the difficult
mone profiles are dynamically altered following task of individualizing treatment to optimize
injury (Wagner et al., 2011a). TBI activates the recovery. Correspondingly, progress in reha-
hypothalamicpituitaryadrenal (HPA) axis that bilitation research toward that end is being pur-
increases glucocorticoid production (Grundy sued through an integrative omics approach.
et al., 2001; Lucas et al., 2006). Munck and col- Derived from our work in TBI, rehabilomics
leagues (1984) demonstrate the classic paradigm is a novel framework to address the needs of
that the anti-inflammatory effects of glucocorti- this population that incorporates a transdisci-
coids following the stress response help to medi- plinary approach to understanding the biology,
ate recovery. However, more recent work shows function, prognosis, complications, treatments,
that there is a bidirectional connection between adaptation, and recovery of patients (Wagner,
the immune and endocrine systems through 2010). The reason individuals with similar mech-
the HPA axis (Maier, 2003). Correspondingly, anisms of injury and clinical management have
it is believed that failure of glucocorticoids to vastly different recovery trajectories could be
430 Part IV:Homeostatic Therapies

multifaceted. For example, intrinsic factors, such DA-Agonist


as age and sex, which play a large role in most There is evidence to suggest that the concentra-
conditions, must be considered. Additionally, tion of DA and its receptors can lead to frontal lobe
genetic variation as well as gene by environ- deficits that can result in a wide variety of cognitive
ment interactions and epigenetic considerations issues (Jokinen etal., 2013; Narayanan etal., 2013).
may affect an individuals response to treatment Patients with Parkinsons disease have DA depletion
as well as present a risk factor for particular from neurodegeneration of the nigrostriatal path-
TBI-related complications. Biomarkers can serve way that results in prefrontal dysfunction. After
as a valuable tool to study the individuality and TBI, there is DA system disruption that is intimately
neurobiology of TBI as a complex disease, and involved with cognitive deficits (Bales etal., 2009).
information from preclinical and clinical bio- However, systemic DA-agonists could be benefi-
marker studies have moved the field forward in cial or detrimental based on the time of adminis-
better understanding the heterogeneity involved tration after injury. In addition to stimulants, two
with TBI-specific pathophysiology. For clinical DA-agonist drugs that have been studied and used
providers, biomarkers may represent a permis- clinically in TBI are bromocriptine and amanta-
sive plastic versus a dysfunctional homeostatic dine hydrochloride (AMH). Bromocriptine is a
state, either of which may have prognostic value mixed D2 agonist that has a dose-specific agonist/
with regard to outcome. Perhaps more impor- antagonist effect. At low doses (2.55 mg/kg), bro-
tant, they may also aid as dynamic markers of mocriptine has been associated with DA neuronal
treatment response. Correspondingly, both tra- firing and evidence that it acts as a partial D2 auto-
ditional and nontraditional therapies discussed receptor antagonist (Jackson etal., 1990; Lieberman
here encompass pharmacotherapies, physio- and Goldstein, 1985). At higher doses (>10 mg/kg),
therapies, and biologics commonly used to treat bromocriptine acts as more of a D2 autorecep-
TBI, and, where possible, biomarkers associated tor agonist, causing inhibition of DA release and
with treatment mechanisms and response are metabolism (Maruya etal., 2003). In clinical TBI,
discussed. there are reports that low-dose bromocriptine can
result in improvements in prefrontal and attention
Pharmacotherapies deficits (McDowell et al., 1998); however, another
Stimulants study that administered 10 mg/day of bromocrip-
Psychostimulants, including D-amphetamine tine to a population with moderate to severe TBI
(AMPH) and methylphenidate (MPH), have been did not see benefits in cognitive recovery (Whyte
prescribed recently to patients with TBI. MPH etal.,2008).
acts as a DA transporter blocker, while AMPH AMH, which has also been used as an anti-
works as a transporter substrate that promotes viral therapy for influenza type-A (Atkinson
DA release into the extracellular space and also et al., 1986) as well in Parkinsons disease
release through reverse transport (Fleckenstein (Godwin-Austen et al., 1970; Metman et al.,
et al., 2007). Despite varying mechanisms of 1998) and multiple sclerosis (Cohen and Fisher,
action, both AMPH and MPH have similar 1989), has been used as a DA enhancing agent
effects and are commonly prescribed for atten- for TBI. AMH may increase extracellular DA
tion deficit hyperactivity disorder (ADHD; concentrations through NMDA antagonism and
Scahill etal., 2004). Importantly, typical dosing reuptake inhibition through the DA transporter;
for MPH in a setting of ADHD is considerably however, chronic AMH use could increase DA
higher than for TBI (Whyte et al., 1997). MPH transporter activity (Harun and Wagner, 2014).
use is more common in TBI compared with AMH can be efficacious in increasing arousal
AMPH, with multiple clinical studies showing its and cognition, especially for agitated, restless,
efficacy in improving cognitive performance and or apathetic patients (Van Reekum et al., 1995;
function, especially in the domains of attention Sawyer et al., 2008). Also, a recent multicenter
and memory (Gualtieri and Evans, 1988; Kaelin randomized control trial showed that 4-week
etal., 1996; Kim etal., 2006; Whyte etal., 1997, administration of AMH accelerated recovery
2004). Also, experimental TBI models show that and led to benefits in patients in a minimally con-
prolonged MPH treatment after controlled corti- scious state (Giacino etal., 2012). This work pro-
cal impact normalizes many components of pre- vides promising evidence that AMH may have
synaptic striatal DA neurotransmission (Wagner significant utility as a therapeutic to improve
etal.,2009). functional recovery afterTBI.
Traumatic BrainInjury 431

Cholinergics 2012), leaving questions as to how innate het-


After a brain injury, a rapid elevation in ace- erogeneity and differences in standard care may
tylcholine (ACh) is observed (Saija etal., 1988). have influenced treatment response.
The cholinergic system is integral to cogni-
tive function, including attention, arousal, and Antidepressants
memory. Altered cholinergic states could be a Three of the most common classes of antidepres-
factor leading to cognitive deficits following TBI sants include SSRIs, tricyclic antidepressants,
(Dixon etal., 1997), even in the absence of spe- and monoamine oxidase inhibitors (MAOIs).
cific damage to the hippocampus (Lyeth et al., These three drug classes have been studied and
1990). Importantly, acute elevations in ACh are are used to treat posttraumatic behavioral and
followed by a chronically depressed cholinergic mood disorders, including PTD. The most com-
state that may contribute to prolonged mem- prehensive review of treatment for PTD was
ory deficits (Dixon et al., 1995). The reason for done by Fann and colleagues (2009), who iden-
decreased ACh in the chronic stages post-TBI tified 13 studies that examine pharmacothera-
could be altered activity of choline acetyltrans- pies, including 7 studies with SSRIs. The only
ferase, the enzyme responsible for synthesiz- study with class I evidence was a randomized
ing ACh. Specifically, choline acetyltransferase clinical trial examining the efficacy of sertraline
activity is responsible for attenuating the acute compared to placebo (Ashman etal., 2009). The
hypercholinergic state, but this enzyme may study found no statistically significant difference
also contribute to a chronic hypofunctioning between the two treatment arms. Another study
state (Levin et al., 2014). Within the context of with classII evidence compared treatment with
a hypocholinergic state, individuals with TBI MPH or sertraline versus placebo (Lee et al.,
have more sensitivity to anticholinergic phar- 2005). This study found that both experimen-
macotherapies compared with healthy individu- tal groups showed improvements in depressive
als (Griffin et al., 2003). Therefore, drugs with symptoms compared to placebo; however, MPH
anticholinergic effects should be used with cau- showed added benefit beyond sertraline for
tion in patients with TBI (Arciniegas etal., 2000; cognitive-related symptoms. Other studies have
Levin etal., 2014; Stanislav,1997). shown sertraline was well tolerated and provided
A systematic review examined the efficacy of benefits for patients with PTD (Fann etal., 2009;
various cholinergic agents following TBI (Poole Rapoport etal., 2008; Turner-Stokes etal., 2002).
and Agrawal, 2008). Studies involving acetylcho- Studies involving SSRIs have examined citalo-
linesterase inhibitors showed modest improve- pram and fluoxetine; however, evidence for their
ments in cognitive benefits. The authors cite efficacy is less established (Horsfield etal., 2002;
issues with timing of administration of the drug, Perino et al., 2001). There is limited evidence
with early administration during the hypercho- for TCA use for treating PTD. One small study
linergic state being neurotoxic and later admin- examined desipramine in a severe TBI popula-
istration being more effective. Also, studies that tion, but results are hard to interpret due to study
used a combined therapy of physostigmine and limitations (Wroblewski etal., 1996). Two other
lecithin, a choline precursor, showed no major studies examined amitriptyline for PTD and
additive effects from this combined therapy to found no treatment effects (Dinan and Mobayed,
improve memory (Levin et al., 1986; Walton, 1992; Saran, 1985). Intervention with MAOIs
1982), and these cholinesterase inhibitors are not have not been well studied (Newburn etal., 1999;
recommended for clinical administration (Poole Saran, 1985). With this evidence in mind, current
and Agrawal, 2008). However, other inhibi- knowledge about efficacy of antidepressants in
tors, including donepezil, galantamine, and PTD treatment is unclear. Due to the heterogene-
rivastigmine, have shown therapeutic potential ity of TBI, many researchers posit that PTD has
in chronic TBI (Tenovuo, 2005). Additionally, varying pathophysiology compared with MDD,
CDP-choline, an ACh precursor and key interme- thus it is possible that antidepressants efficacious
diate in the biosynthesis of phosphatidylcholine, for MDD may not provide the same benefits in
has also been shown to improve global outcome TBI (Failla etal., 2013; Saran, 1985). Due to the
and improve motor and cognitive deficits fol- high prevalence of PTD, there is a pronounced
lowing TBI (Calatayud Maldonado et al., 1991). need for more high-quality studies assessing
However, a large multisite clinical trial featuring antidepressant use and other treatments in this
CDP-choline did not show benefit (Zafonte etal., population.
432 Part IV:Homeostatic Therapies

Anticonvulsants to lessen the likelihood of a sustained chronic


As previously discussed in detail elsewhere in inflammatory state. To date, anti-inflammatory
this chapter, seizures are a common complication therapies for TBI have provided inconclusive
following TBI; however, treatment studies to date results. Some studies have examined the use of
and prophylaxis guidelines shed little light on corticosteroids; however, this did not prove to
how AEDs might influence outcome. Acompre- be efficacious in a TBI population and in fact
hensive systematic review examining the efficacy resulted in an elevated mortality risk (Edwards
of AEDs following acute TBI (Schierhout and et al., 2005). Others have examined the use of
Roberts, 1996) included six published studies, nonsteroidal anti-inflammatory therapies such as
and the prophylactics studied included: pheny- minocycline, which have been efficacious in other
toin (McQueen etal., 1983; Pechadre etal., 1991; CNS injury models (Alano et al., 2006; Maier
Temkin et al., 1990; Young et al., 1983), carba- et al., 2005; Stirling et al., 2004). Additionally,
mazepine (Gltzner et al., 1983), and pentobar- there is some evidence that the interleukin-1
bital (Manaka, 1992). The authors of the review receptor antagonist (IL-1ra) can attenuate neu-
calculated a pooled relative risk for early seizure ronal damage after injury due to its effects on
prevention of 0.34 but, due to the heterogeneity of IL-1-initiated damage (Toulmond and Rothwell,
the included studies, could not make any claims 1995)and effects on downstream neurotrophins,
regarding late seizure risk or development of such as nerve-growth factor (Dekosky et al.,
PTE following therapy. Agrawal and colleagues 1996). IL-1ra use has been well studied in other
(2006) posit that there is no current evidence that disease models, such as rheumatological diseases
one drug is better than the other. It is noted (Bresnihan etal., 1998; Cohen etal., 2002; Horai
that a recent randomized controlled trial found etal., 2000)and type II diabetes mellitus (Larsen
that levetiracetam shows modest improvement et al., 2007), but it is understudied in TBI as an
over phenytoin (Szaflarski et al., 2010) for criti- anti-inflammatory therapy.
cally ill patients with TBI. Additionally, experi- Ziebell and Morganti-Kossmann (2010)
mental work in the controlled cortical impact explain that the challenge with anti-
model shows that daily levetiracetam treatment inflammatory therapies in TBI is achieving mild
can improve neurological recovery and neuro- immunosuppression that modifies but does not
plasticity through modulation of inflammation entirely eliminate the inflammatory response.
and excitatory pathways (Zou etal., 2013). Despite With this in mind, there is a definite need for
this, there still remains a lack of consistent evi- continued research in experimental and clinical
dence that acute therapies for posttraumatic sei- models to understand the temporal dynamics
zures affect the eventual development of PTE. of both pro- and anti-inflammatory cascades in
Researchers still have significant work ahead to TBI to better differentiate the neuroprotective
determine new therapeutic targets, examine how and detrimental properties of posttraumatic
genetic heterogeneity might influence treatment inflammation. As inflammation is implicated
response, and better understand how acute pro- in several chronic complications after TBI,
phylactic therapies after TBI reduce early seizure this knowledge may be useful in driving tar-
risk andPTE. geted therapeutics for patients with multiple
TBI-related conditions.
Anti-Inflammatory Strategies
As discussed earlier, TBI is characterized by a Physiotherapies
significant inflammatory response. The rapid Exercise
activation of microglia and upregulation of pro- There is a wealth of evidence that physical activ-
inflammatory cytokines leads to a significant ity can improve and enhance mood, quality of
inflammatory load in the acute and subacute stages life, and cognitive function in both healthy and
of brain injury (Kumar et al., 2014). Among the neurologically impaired populations (Erickson
relatively new considerations for TBI therapeutic etal., 2011; Taylor etal., 1985; Wolin etal., 2007).
interventions are anti-inflammatory treatments. Exercise used alone or in conjunction with other
The goal of these therapies is not to halt the inflam- cognitive and pharmaceutical interventions
matory process in its entirety, as there is a fairly (Archer et al., 2012) can endogenously increase
well-established neuroprotective role of inflam- neurotrophins (Griesbach et al., 2004) that (at
mation in TBI (Morganti-Kossmann etal., 2002), least chronically) are important to neuronal
but to ameliorate the proinflammatory cascade plasticity and repair after TBI. One randomized
Traumatic BrainInjury 433

controlled trial in TBI showed a significant Mindfulness Meditation


improvement in depressive symptoms and an During rehabilitation of individuals with TBI
overall higher quality of life after aerobic exercise or cognitive deficits, it is imperative to imple-
(Hoffman et al., 2010). Interestingly, multiple ment learning and cognitive strategies to opti-
studies have found positive effects with Tai Chi mally engage patients during therapy. Also,
Chuan Qigong exercise on mood and self-esteem results from neuropsychological tests can be
(Blake and Batson, 2009; Gemmell and Leathem, used to guide personalized strategies for learning
2006). Animal models of TBI have identified (Cicerone etal., 2011). Aquiet and low-stimulant
voluntary exercise as a robust intervention that environment is critical to engage a patient in
leads to cell proliferation and cell survival in a single task. In addition, learning should be
the dentate gyrus of the hippocampus, which conducted in a procedural, step-by-step man-
may accelerate learning and memory deficits ner with repetition at each stage (Haslam etal.,
that are characteristic in neurological disorders 2011; McCulloch, 2007; Vakil, 2005). One highly
when introduced at an appropriate timeframe developed behavioral strategy that could be
after TBI (Griesbach etal., 2004; Hillman etal., adapted for a cognitively impaired population
2008). Exercise also leads to a corresponding up is mindfulness meditation, which is designed to
regulation of BDNF protein, which in turn can target attention and moment-to-moment aware-
have positive effects on learning, memory, and ness (Kabat-Zinn etal., 1985). This technique has
mood (Erickson et al., 2011). In fact, research- been well established as a treatment for chronic
ers posit that the positive effects on cognition pain (Kabat-Zinn, 1982, 1994). However, others
that result from exercise are mediated through have found evidence for its efficacy in a clinical
BDNF (Griesbach etal., 2009). Exercise confers acquired brain injury population for reducing
other neuroprotective effects by reducing oxi- mental fatigue, a common symptom following
dative stress and free radical production (Lima TBI (Johansson et al., 2012; Olver et al., 1996).
etal., 2009)and reducing cytokine overproduc- Another study found that mindfulness medita-
tion (Ding etal.,2006). tion can improve cognitive deficits, specifically
Despite the postulated neuroprotective stimulus overselectivity, a behavioral phenom-
effects, the timing of exercise intervention fol- enon showing characteristic hyperattentiveness
lowing TBI is an important consideration. There to certain stimuli and lack of attention to others
is considerable controversy in the mild TBI/con- (McHugh and Wood, 2013). Another study found
cussion literature regarding when athletes can preliminary evidence that mindfulness-based
return to play after injury, as early return prior therapy reduced depressive symptoms after TBI
to resolution of associated pathophysiological (Bdard et al., 2013). Mindfulness-based thera-
cascades can exacerbate symptoms and increase pies may be promising as a nonpharmaceutically
the risk for a repeat, more serious head injury based intervention during periods of stress and
(Lovell et al., 2004). Clinical and experimen- depression after TBI. However, further replica-
tal TBI studies have shown that exercise in the tion studies, strategies for optimizing interven-
acute stages following injury can negatively affect tion in populations with cognitive impairment
recovery (Kozlowski etal., 1996; Majerske etal., and behavioral problems, and examination of
2008). These findings suggest that later exercise who can benefit the most from these strategies
is better to achieve neuroprotection/restoration, should be explored.
and acute exercise can have negative effects on
outcomes. One hypothesis for these findings Cognitive Rehabilitation
may be the reduction in BDNF protein upregula- and Strategy Training
tion that occurs with acute versus more delayed Cognitive deficits are commonly observed
exercise (Griesbach etal., 2004), which may be a after TBI and can be a significant burden on
neuroprotective response in the acute phase since caregivers as well as limit potential benefit
BDNF target receptors for apoptosis are elevated from other aspects of rehabilitation therapy.
during that time (Rostami et al., 2014). In sum, Rehabilitation strategies can improve the cogni-
evidence exists that exercise has potentially tive issues that result from TBI through various
potent neurorestorative properties following strategy-training approaches; these approaches
TBI, particularly when considerations are taken largely target implicit elements of learning and
regarding timing, intensity, and individualized cognition (Cicerone etal., 2011), and applicabil-
limitations to maximize benefits. ity can be tailored to individualized needs across
434 Part IV:Homeostatic Therapies

a wide range of deficits and homeostatic dys- rehabilitation and how to normalize the homeo-
function (Sohlberg and Turkstra, 2011). These static underpinnings of cognition to optimize
issues include attention, visuospatial function- recovery.
ing, language and communication, memory, and
executive functioning. Cognitive rehabilitation Acupuncture
approaches in practice are complex and multi- The biological mechanisms underlying the effi-
factorial but can be broadly characterized into cacy of acupuncture, an alternative medicine that
two categories: (a) cognitive function retrain- dates back over 2,000years in eastern Asia (Wu,
ing and (b) functional compensation (Ponsford 1996), is controversial, with nearly one-third
et al., 2012). The former strategy focuses on of studies providing no biological rationale
attenuating specific cognitive functions that are (Moffet, 2006). Studies that have discussed the
impaired after injury through specific, organ- physiological mechanisms attributed the ben-
ized neuropsychological exercises (Malec et al., efits of acupuncture to neurochemical, segmen-
1984; Novack et al., 1996; Ben-Yishay et al., tal (gate theory), autonomic regulation, local
1987). It is important to note that cognitive effects, and effects on brain function (Moffet,
function retraining is built on the foundation 2006). The National Center for Complementary
of a bottom-up approach, which makes the and Alternative Medicine (2014) of the National
assumption that recovery from injury is based Institutes of Health cites the major benefits of
on amelioration of specific cognitive impair- acupuncture as regulating the nervous system
ments (Ponsford etal., 2012). The second broad by dynamically affecting the release of pain bio-
category of cognitive rehabilitation interven- chemicals (e.g., endorphins), as well as altering
tion is functional compensation, which empha- the release of neurotransmitters and neurohor-
sizes improvements in various domains of life, mones that affect parts of the CNS related to sen-
including mobility, self-care, domestic life, and sation, such as immunology and blood flow. Due
community. This approach could be considered to the overlap of the proposed mechanisms of
a top-down approach as it focuses on building action of acupuncture and the pathophysiology
on a persons strengths to compensate for cog- of CNS disorders, like TBI, it is reasonable to sug-
nitive impairments and personalize therapy for gest a potential therapeutic use for different neu-
individual personal goals (Ponsford etal., 2012). rological complications (Ernst etal., 2001). Wong
Improved task performance can be achieved and colleagues (2011) conducted a review of the
through internal strategies (e.g., mnemonics; literature and found four randomized controlled
Kaschel etal., 2002; Ryan and Ruff, 1988; Thoene trials that studied traditional Chinese acupunc-
and Glisky, 1995), external mechanisms, and ture in TBI populations. The authors noted that
environmental restructuring (e.g., cueing or although some studies supported the efficacy of
memory aids; Evans etal., 2003; Wilson, 1991). It acupuncture, the overall methodological qual-
is important to note that the cognitive rehabilita- ity of the studies were low, making it difficult
tion literature has certain limitations. For exam- to draw conclusions. Patients with specific TBI
ple, many studies incorporate training that uses complications, such as PTM and PTD, which do
artificial tasks that are not likely to be encoun- not have well-established pharmacological inter-
tered by patients with TBI. Also, few studies have ventions to date, may be promising candidates
been able to incorporate robust ways to quantify for acupuncture targets. However, the evidence
the ability of an individual to use a strategy to for efficacy of acupuncture for migraine (Linde
improve performance in real-world settings. et al., 1996, 2005; Loh et al., 1984) and MDD
Importantly, there is a lack of incorporation of (Mukaino etal., 2005; Wang etal., 2008)is mixed
preclinical animal work that examines cognitive and needs further study. In sum, the physiologi-
rehabilitation and strategy training. Work from cal mechanisms of acupuncture, although not
our lab (Wagner et al., 2013) discusses a clini- entirely established in itself, are likely wide rang-
cally relevant animal model using nonspatial ing and have systemic effects on the CNS, spe-
pretraining to discriminate implicit and explicit cifically in the treatment of chronic pain (Vickers
learning through the evaluation of spatial and et al., 2012). As previously emphasized, many
nonspatial learning and memory constructs TBI pharmacological drugs have failed to show
within the Morris water maze. Overall, there is a efficacy due to the characteristic high degree of
need for further study to refine our understand- heterogeneity in the secondary-injury cascade.
ing in the biological foundations of cognitive It is possible that acupuncture may be a logical
Traumatic BrainInjury 435

alternative form of therapy due to its more global, with progesterone to enhance its neuroprotective
pleiotropic effects; however, at the present time effects following TBI by targeting inflammatory,
there is not enough evidence to make a defini- apoptotic, and excitotoxicity pathways (Cekic
tive claim. Thus there is a need for well-designed and Stein, 2010). One animal study in TBI shows
experimental and clinical studies that specifically that the combined use of a pharmacological-dose
address the physiological process and homeo- of VDH (5g/kg) and progesterone was not sig-
static mechanisms that acupuncture therapy may nificantly different with respect to memory tasks
have to offer individuals withTBI. compared to progesterone alone; however, com-
bined therapy did reduce neuronal loss and reac-
Biologics tive astrocyte reduction (Tang etal., 2013). Given
Progesterone/Estradiol the complexity of VDH and progesterone, the
Numerous experimental studies have suggested exact mechanisms of actions are still unclear, but
a neuroprotective role for steroids, including some posit that the systemic anti-inflammatory
progesterone and estradiol (Roof et al., 1994; properties of both agents may complement
Schumacher et al., 1996, 2004; Stein, 2001; each other both in the brain and in the periph-
Wright et al., 2001). As noted earlier, clinical ery (Stein and Cekic, 2011). With this in mind,
TBI studies have shown significant alterations progesterone and vitamin D may be a logical
in endogenous serum and CSF hormonal pro- approach for combined therapy for TBI patients
files after injury (Garringer et al., 2013; Rogers with vitamin D deficiencies; to date there is still
and Wagner, 2006; Wagner et al., 2011a). sparse evidence for the efficacy of progesterone
Experimental studies show that progesterone and VDH therapy in humans.
has strong neuroprotective effects that target
cerebral edema, as well as reduce neuroinflam- Omega-3 FattyAcids
mation, BBB deterioration, and cellular necrosis A healthy diet rich in omega-3 fatty acids
(Stein etal., 2008). Estradiol treatment has also has powerful neuroprotective effects and can
been shown to be neuroprotective in animal increase molecules important for normal brain
models by improving synaptic transmission and function, including BDNF, which can lead to
ameliorating excitotoxicity and oxidative injury improvements in synaptic and cognitive plastic-
(Stein, 2001). There is limited research on the ity in the injured brain (Wu etal., 2004; Zuccato
use of estradiol as a therapy in clinical TBI. Two and Cattaneo, 2009). Several studies have shown
published studies have shown safety and initial that omega-3 supplementation in animal mod-
clinical efficacy of progesterone therapy for acute els of TBI lead to BDNF elevations, reduced
TBI (Wright et al., 2007; Xiao et al., 2008), but oxidative stress, and improved cell signaling
phase III trials did not support a beneficial effect (Salem et al., 2001). Interestingly, a recent ani-
(Skolnick etal., 2014; Wright etal., 2014). Given mal study found evidence that progesterone may
the complexity of endogenous hormone physi- play a large neuroprotective role in specifically
ology after TBI, future research should aim to omega-3 deficient rats through the maintenance
understand how hormone treatments influence of neuronal signaling by supporting membrane
peripheral and CNS hormone profiles and their stability and axonal growth (Tyagi etal., 2014).
downstream effects on other mechanisms of sec- Dietary and supplementation of omega-3 has also
ondary injury. been linked to improvements in MDD (Levant,
2011; Lotrich etal., 2013). Also, one experimen-
VitaminD tal study showed decreased depressive symp-
Recent evidence implicates vitamin D deficiency toms as well as increased plasma 5-HT levels
in chronic fatigue after TBI (Schnieders et al., and lower IL-6 levels following dietary intake of
2012). Vitamin D deficiency is highly prevalent omega-3 (Park etal., 2012). Omega-3 supplemen-
in the elderly (Gloth and Tobin, 1995; Visser tation reportedly also has efficacy in migraine
et al., 2003), pregnant women (Andran et al., treatment (Harel etal., 2002; Simopoulos, 2002).
2002), the obese (Buffington et al., 1993), and Due to positive links between omega-3 and
individuals with osteoporosis (Lips etal., 2006). TBI-related pathologies, there is promise for the
Furthermore, the heterogeneity of TBI pathology clinical utility of omega-3 supplementation for
makes vitamin D use in combination with other homeostatic restoration in patients with TBI.
therapies logical. Specifically, vitamin D hormone However, well-designed randomized trials are
(VDH) has begun to be evaluated as a cotherapy needed.
436 Part IV:Homeostatic Therapies

Dietary Polyphenols after TBI and aid in the design of effective per-
Polyphenols are a broad group of organic sonalized therapies. In addition to currently
substances that contain an excess of phenol used pharmacotherapies for TBI management,
groups. Two of the most common groups that emerging physiotherapies and biologics may
are believed to have CNS-protective properties have relevance in managing the wide range of
include curcumin and flavonoids. Among the conditions and complications associated with
benefits of polyphenols are powerful antioxidant TBI. Newer approaches, including comparative
and anti-inflammatory properties. Flavonoids effectiveness studies and randomized controlled
can interact with neuronal signaling, specifi- trials that incorporate effective biological indica-
cally protein kinase and lipid kinase pathways, tors for stratification, and monitoring of treat-
which can inhibit apoptosis and promote neu- ment effects may further aid in the identification
ronal survival (Spencer, 2009). Foods rich in fla- of effective homeostatic treatments that restore
vonoids include vegetables and fruits, especially function for individuals dealing with the chronic
berries, green tea, and red wine. Curcumin is a effects ofTBI.
common dietary and medicinal product used in
India that is found in turmeric and curries. One References
study found that dietary curry consumption has Agha, A., Rogers, B., Sherlock, M., OKelly, P.,
positive effects on cognition in a neurologically Tormey, W., Phillips, J., and Thompson, C.J.
intact, elderly population (Ng et al., 2006). In (2004). Anterior pituitary dysfunction in
addition to curcumin and flavonoids, resveratrol survivors of traumatic brain injury. J. Clin.
is another powerful polyphenol that may have Endocrinol. Metab. 89, 49294936.
protective effects in the heart, brain, and kidney, Agrawal, A., Timothy, J., Pandit, L., and Manju, M.
in addition to its antiaging properties (Kalantari (2006). Post-traumatic epilepsy: An overview.
and Das, 2010). Thus dietary phenols appear to Clin. Neurol. Neurosurg. 108, 433439.
have promising neuroprotective properties that Alano, C.C., Kauppinen, T.M., Valls, A.V., and
may be logical therapeutic approaches to target Swanson, R.A. (2006). Minocycline inhibits
poly(ADP-ribose) polymerase-1 at nanomolar
in reducing secondary-injury cascades that result
concentrations. Proc. Natl. Acad. Sci. U.S.A. 103,
from TBI. However, like other biologic therapies
96859690.
discussed, more treatment studies in TBI are nec-
Alderson, P., and Roberts, I. (2005). Corticosteroids
essary to move this area of research forward.
for acute traumatic brain injury. Cochrane
Database Syst. Rev. 2, CD000196.
CONCLUSIONS Andran, N., Yordam, N., and zn, A. (2002). Risk
As the literature here suggest, TBI is a complex factors for vitamin D deficiency in breast-fed
disease process whereby injury and innate het- newborns and their mothers. Nutrition
erogeneity play a prominent role in acute and 18,4750.
chronic recovery. In response to primary injury, Andriessen, T.M.J.C., Jacobs, B., and Vos, P.E. (2010).
inflammation, excitotoxicity, and BBB disruption Clinical characteristics and pathophysiological
lead to reactive astrocytosis, ischemia, and oxida- mechanisms of focal and diffuse traumatic brain
tive stress. These acute processes lead to chronic injury. J. Cell. Mol. Med. 14, 23812392.
inflammation, impaired neurotransmission, and Annegers, J.F., Hauser, W.A., Coan, S.P., and Rocca,
other types of CNS dysfunction. Further, the lit- W.A. (1998). A population-based study of sei-
erature is mixed on if, how, and when potential zures after traumatic brain injuries. N. Engl.
neuroprotectants such as neurotrophins and sex J.Med. 338,2024.
and stress steroids confer benefits in the context Ansari, M.A., Roberts, K.N., and Scheff, S.W. (2008).
of TBI. Persistent aberrations in humoral mark- Oxidative stress and modification of synaptic
ers (inflammation, neurotrophins, sex/stress proteins in hippocampus after traumatic brain
steroids) are likely contributors to chronic condi- injury. Free Radic. Biol. Med. 45, 443452.
tions and complications after TBI. Though sev- Archer, T., Svensson, K., and Alricsson, M. (2012).
eral agents have been tested, there are currently Physical exercise ameliorates deficits induced by
no known effective neuroprotectants for treat- traumatic brain injury. Acta Neurol. Scand. 125,
ing TBI. However, a better understanding of the 293302.
likely chronic disease patho-mechanisms, as well Arciniegas, D.B., Topkoff, J., and Silver, J.M. (2000).
as the mechanistic impact of innate heterogene- Neuropsychiatric aspects of traumatic brain
ity, may help minimize risk for complications injury. Curr. Treat. Options Neurol. 2, 169186.
Traumatic BrainInjury 437

Ashman, T.A., Cantor, J.B., Gordon, W.A., Spielman, L., and Medina, J.H. (2008b). BDNF is essential to
Flanagan, S., Ginsberg, A., Engmann, C., Egan, promote persistence of long-term memory stor-
M., Ambrose, F., and Greenwald, B. (2009). A age. Proc. Natl. Acad. Sci. U.S.A. 105, 27112716.
randomized controlled trial of sertraline for the Bell, M.J., Kochanek, P.M., Doughty, L.A., Carcillo, J.A.,
treatment of depression in persons with trau- Adelson, P.D., Clark, R.S., Wisniewski, S.R.,
matic brain injury. Arch. Phys. Med. Rehabil. 90, Whalen, M.J., and DeKosky, S.T. (1997).
733740. Interleukin-6 and interleukin-10 in cerebrospi-
Atkinson, W.L., Arden, N.H., Patriarca, P.A., Leslie, N., nal fluid after severe traumatic brain injury in
Lui, K., and Gohd, R. (1986). Amantadine pro- children. J. Neurotrauma 14, 451457.
phylaxis during an institutional outbreak of Ben-Yishay, Y., Piasetsky, E.B., and Rattok, J. (1987).
type a (H1N1) influenza. Arch. Intern. Med. 146, A systematic method for ameliorating disor-
17511756. ders in basic attention. In: Neuropsychological
Bales, J.W., Wagner, A.K., Kline, A.E., and Dixon, Rehabilitation. Meier, M.J., Benton, A.L., Diller, L,
C.E. (2009). Persistent cognitive dysfunction after eds. (NewYork:Guilford Press), pp. 165181.
traumatic brain injury:Adopamine hypothesis. Bernheimer, H., Birkmayer, W., Hornykiewicz, O.,
Neurosci. Biobehav. Rev. 33, 9811003. Jellinger, K., and Seitelberger, F. (1973). Brain
Baloyannis, S.J., and Gonatas, N.K. (1979). dopamine and the syndromes of Parkinson and
Distribution of anti-HRP antibodies in the Huntington:Clinical, morphological and neuro-
central nervous system of immunized rats chemical correlations. J. Neurol. Sci. 20, 415455.
after disruption of the blood brain barrier. J. Bertolino, A., Fazio, L., Di Giorgio, A., Blasi, G.,
Neuropathol. Exp. Neurol. 38, 519531. Romano, R., Taurisano, P., Caforio, G., Sinibaldi, L.,
Barton, D.J., Kumar, R.G., Arenth, P.M., Berga, S., Ursini, G., Popolizio, T., et al. (2009).
and Wagner, A.K. (2015). Persistent hypogo- Genetically determined interaction between
nadotropic hypogonadism: Temporal hormone the dopamine transporter and the D2 recep-
profiles and outcome prediction in men after tor on prefronto-striatal activity and volume in
severe traumatic brain injury. J. Head Trauma humans. J. Neurosci. 29, 12241234.
Rehabilitation. Manuscript accepted. In press. Besedovsky, H.O., and del Rey, A. (2000). The
Bartrs-Faz, D., Junqu, C., Serra-Grabulosa, J.M., cytokine-HPA axis feed-back circuit. Z. Fr
Lpez-Alomar, A., Moya, A., Bargall, N., Rheumatol. 59, II26II30.
Mercader, J.M., Moral, P., and Clemente, I.C. Blake, H., and Batson, M. (2009). Exercise interven-
(2002). Dopamine DRD2 Taq I polymorphism tion in brain injury:Apilot randomized study of
associates with caudate nucleus volume and cog- Tai Chi Qigong. Clin. Rehabil. 23, 589598.
nitive performance in memory impaired sub- Bliss, T.V., and Collingridge, G.L. (1993). A synaptic
jects. Neuroreport 13, 11211125. model of memory:Long-term potentiation in the
Bayir, H., Marion, D.W., Puccio, A.M., Wisniewski, S.R., hippocampus. Nature 361,3139.
Janesko, K.L., Clark, R.S.B., and Kochanek, P.M. Boison, D. (2008). Adenosine as a neuromodulator
(2004). Marked gender effect on lipid peroxida- in neurological diseases. Curr. Opin. Pharmacol.
tion after severe traumatic brain injury in adult 8,27.
patients. J. Neurotrauma 21,18. Bombardier, C.H., Fann, J.R., Temkin, N.R.,
Beauchamp, K., Mutlak, H., Smith, W.R., Shohami, E., Esselman, P.C., Barber, J., and Dikmen, S.S.
and Stahel, P.F. (2008). Pharmacology of trau- (2010). Rates of major depressive disorder and
matic brain injury:Where is the golden bullet? clinical outcomes following traumatic brain
Mol. Med. 14, 731740. injury. JAMA 303, 19381945.
Bdard, M., Felteau, M., Marshall, S., Cullen, N., Bonneh-Barkay, D., Zagadailov, P., Zou, H.,
Gibbons, C., Dubois, S., Maxwell, H., Niyonkuru, C., Figley, M., Starkey, A., Wang, G.,
Mazmanian, D., Weaver, B., Rees, L., etal. (2013). Bissel, S.J., Wiley, C.A., and Wagner, A.K.
Mindfulness-based cognitive therapy reduces (2010). YKL-40 Expression in traumatic brain
symptoms of depression in people with a trau- injury: An initial analysis. J. Neurotrauma 27,
matic brain injury: Results from a randomized 12151223.
controlled trial. J. Head Trauma Rehabil. Brain Trauma Foundation, American Association
Bekinschtein, P., Cammarota, M., Izquierdo, I., and of Neurological Surgeons, and Congress of
Medina, J.H. (2008a). Reviews:BDNF and mem- Neurological Surgeons. (2007). Guidelines
ory formation and storage. The Neuroscientist for the management of severe traumatic brain
14, 147156. injury. J. Neurotrauma. 24(suppl 1), S1106.
Bekinschtein, P., Cammarota, M., Katche, C., Bresnihan, B., Alvaro-Gracia, J.M., Cobby, M.,
Slipczuk, L., Rossato, J.I., Goldin, A., Izquierdo, I., Doherty, M., Domljan, Z., Emery, P., Nuki, G.,
438 Part IV:Homeostatic Therapies

Pavelka, K., Rau, R., Rozman, B., et al. (1998). dopaminergic fibres. Neurosci. Lett. 107,
Treatment of rheumatoid arthritis with recom- 256260.
binant human interleukin-1 receptor antagonist. Chen, J.-K., Johnston, K.M., Petrides, M., and Ptito, A.
Arthritis Rheum. 41, 21962204. (2008). Neural substrates of symptoms of depres-
Buffington, C., Walker, B.Jr., G.S. M.C., and Scruggs, D. sion following concussion in male athletes with
(1993). Vitamin D deficiency in the morbidly persisting postconcussion symptoms. Arch. Gen.
obese. Obes. Surg. 3, 421424. Psychiatry 65,8189.
Bullock, R., Zauner, A., and Tsuii, O. (1995). Patterns Chen, X., Li, Y., Kline, A.E., Dixon, C.E., Zafonte, R.D.,
of excitatory amino acid release and ionic flux and Wagner, A.K. (2005). Gender and environ-
after severe head trauma. In: Neurochemical mental effects on regional brain-derived neu-
Monitoring in the Intensive Care Unit, rotrophic factor expression after experimental
Tsubokawa, T., ed. (Tokyo:Springer), pp.6467. traumatic brain injury. Neuroscience 135,1117.
Bullock, R., Zauner, A., Woodward, J.J., Myseros, Chen, Y., and Swanson, R.A. (2003). Astrocytes and
J., Choi, S.C., Ward, J.D., Marmarou, A., and brain injury. J. Cereb. Blood Flow Metab. 23,
Young, H.F. (1998). Factors affecting excitatory 137149.
amino acid release following severe human head Chesnut, R.M., Marshall, L.F., Klauber, M.R., Blunt,
injury. J. Neurosurg. 89, 507518. B.A., Baldwin, N., Eisenberg, H.M., Jane, J.A.,
Busto, R., Dietrich, W.D., Globus, M.Y.-T., Alonso, O., Marmarou, A., and Foulkes, M.A. (1993). The
and Ginsberg, M.D. (1997). Extracellular release role of secondary brain injury in determining
of serotonin following fluid-percussion brain outcome from severe head injury. J. Trauma 34,
injury in rats. J. Neurotrauma 14,3542. 216222.
Buttram, S.D.W., Wisniewski, S.R., Jackson, E.K., Chiaretti, A., Antonelli, A., Riccardi, R., Genovese, O.,
Adelson, P.D., Feldman, K., Bayir, H., Berger, R.P., Pezzotti, P., Di Rocco, C., Tortorolo, L., and
Clark, R.S.B., and Kochanek, P.M. (2007). Piedimonte, G. (2008). Nerve growth factor
Multiplex assessment of cytokine and che- expression correlates with severity and out-
mokine levels in cerebrospinal fluid following come of traumatic brain injury in children. Eur.
severe pediatric traumatic brain injury: Effects J.Paediatr. Neurol. 12, 195204.
of moderate hypothermia. J. Neurotrauma 24, Cicerone, K.D., Langenbahn, D.M., Braden, C.,
17071717. Malec, J.F., Kalmar, K., Fraas, M., Felicetti, T.,
Bye, N., Carron, S., Han, X., Agyapomaa, D., Ng, S.Y., Laatsch, L., Harley, J.P., Bergquist, T., etal. (2011).
Yan, E., Rosenfeld, J.V., and Morganti-Kossmann, Evidence-based cognitive rehabilitation:Updated
M.C. (2011). Neurogenesis and glial prolifera- review of the literature from 2003 through 2008.
tion are stimulated following diffuse traumatic Arch. Phys. Med. Rehabil. 92, 519530.
brain injury in adult rats. J. Neurosci. Res. 89, Cohen, R.A., and Fisher, M. (1989). Amantadine
9861000. treatment of fatigue associated with multiple
Calatayud Maldonado, V., Calatayud Prez, J.B., and sclerosis. Arch. Neurol. 46, 676680.
Aso Escario, J. (1991). Effects of CDP-choline Cohen, S., Hurd, E., Cush, J., Schiff, M., Weinblatt, M.E.,
on the recovery of patients with head injury. J. Moreland, L.W., Kremer, J., Bear, M.B., Rich, W.J.,
Neurol. Sci. 103, Suppl,1518. and McCabe, D. (2002). Treatment of rheuma-
Castellanos, N.P., Leyva, I., Buld, J.M., Bajo, R., toid arthritis with anakinra, a recombinant
Pal, N., Cuesta, P., Ordez, V.E., Pascua, C.L., human interleukin-1 receptor antagonist, in
Boccaletti, S., Maest, F., et al. (2011). combination with methotrexate: Results of a
Principles of recovery from traumatic brain twenty-fourweek, multicenter, randomized,
injury: Reorganization of functional networks. double-blind, placebo-controlled trial. Arthritis
Neuroimage 55, 11891199. Rheum. 46, 614624.
Cekic, M., and Stein, D.G. (2010). Traumatic brain Coles, J.P., Fryer, T.D., Smielewski, P., Chatfield, D.A.,
injury and aging:Is a combination of progester- Steiner, L.A., Johnston, A.J., Downey, S.P.M.J.,
one and vitamin D hormone a simple solution to Williams, G.B., Aigbirhio, F., Hutchinson, P.J.,
a complex problem? Neurotherapeutics 7,8190. etal. (2004). Incidence and mechanisms of cere-
Centonze, D., Gubellini, P., Picconi, B., Calabresi, P., bral ischemia in early clinical head injury. J.
Giacomini, P., and Bernardi, G. (1999). Unilateral Cereb. Blood Flow Metab. 24, 202211.
dopamine denervation blocks corticostriatal Conley, Y.P., Okonkwo, D., Deslouches, S., Alexander,
LTP. J. Neurophysiol. 82, 35753579. S., Puccio, A.M., Beers, S.R., and Ren, D.
Chapman, A.G., Drmuller, N., Lees, G.J., and (2014). Mitochondrial polymorphisms impact
Meldrum, B.S. (1989). Excitotoxicity of NMDA outcomes after severe traumatic brain injury. J.
and kainic acid is modulated by nigrostriatal Neurotrauma. 31,3441.
Traumatic BrainInjury 439

Croll, S.D., Ip, N.Y., Lindsay, R.M., and Wiegand, S.J. Gogichaisvili, T., Golden, N., Hartzenberg, B.,
(1998). Expression of BDNF and trkB as a func- et al. (2005). Final results of MRC CRASH, a
tion of age and cognitive performance. Brain randomised placebo-controlled trial of intra-
Res. 812, 200208. venous corticosteroid in adults with head
Darrah, S.D., Miller, M.A., Ren, D., Hoh, N.Z., injury-outcomes at 6 months. Lancet 365,
Scanlon, J.M., Conley, Y.P., and Wagner, A.K. 19571959.
(2013). Genetic variability in glutamic acid decar- Egan, M.F., Kojima, M., Callicott, J.H., Goldberg,
boxylase genes:Associations with post-traumatic T.E., Kolachana, B.S., Bertolino, A., Zaitsev, E.,
seizures after severe TBI. Epilepsy Res. 103, Gold, B., Goldman, D., Dean, M., et al. (2003).
180194. The BDNF val66met polymorphism affects
Dekosky, S.T., Styren, S.D., OMalley, M.E., Goss, J.R., activity-dependent secretion of BDNF and
Kochanek, P., Marion, D., Evans, C.H., and human memory and hippocampal function. Cell
Robbins, P.D. (1996). Interleukm-1 receptor 112, 257269.
antagonist suppresses neurotrophin response in Erickson, K.I., Voss, M.W., Prakash, R.S., Basak, C.,
injured rat brain. Ann. Neurol. 39, 123127. Szabo, A., Chaddock, L., Kim, J.S., Heo, S., Alves, H.,
Diamond, M., Ritter, A., Failla, M., Boles, J., White, S.M., et al. (2011). Exercise training
Conley, Y., Kochanek, P., and Wagner, A. (2014). increases size of hippocampus and improves mem-
IL-1 influences on post-traumatic epilepsy ory. Proc. Natl. Acad. Sci. U.S.A. 108, 30173022.
development:A genetics and proteomics cohort Ernst, E., Pittler, M.H., Stevinson, C., and White, A.
study. Epilepsia 55, 11091119. (2001). The Desktop Guide to Complementary
Dietrich, W.D., Alonso, O., and Halley, M. (1994). and Alternative Medicine: An Evidence-Based
Early microvascular and neuronal consequences Approach. (St. Louis, MO:Mosby).
of traumatic brain injury: A light and electron Evans, J.J., Wilson, B.A., Needham, P., and Brentnall,
microscopic study in rats. J. Neurotrauma 11, S. (2003). Who makes good use of memory aids?
289301. Results of a survey of people with acquired brain
Dinan, T.G., and Mobayed, M. (1992). Treatment injury. J. Int. Neuropsychol. Soc. 9, 925935.
resistance of depression after head injury:Apre- Failla, M.D., Burkhardt, J.N., Miller, M.A., Scanlon,
liminary study of amitriptyline response. Acta J.M., Conley, Y.P., Ferrell, R.E., and Wagner, A.K.
Psychiatr. Scand. 85, 292294. (2013). Variants of SLC6A4 in depression risk
Ding, Y.-H., Mrizek, M., Lai, Q., Wu, Y., Reyes, following severe TBI. Brain Inj. 27, 696706.
R., Jr, Li, J., Davis, W.W., and Ding, Y. (2006). Failla, M.D., Conley, Y.P., & Wagner, A.K. (2015a).
Exercise preconditioning reduces brain damage Brain-Derived Neurotrophic Factor (BDNF)
and inhibits TNF-alpha receptor expression after in Traumatic Brain InjuryRelated Mortality
hypoxia/reoxygenation:An in vivo and in vitro Interrelationships Between Genetics and Acute
study. Curr. Neurovasc. Res. 3, 263271. Systemic and Central Nervous System BDNF
Dixon, C.E., Bao, J., Johnson, K.M., Yang, K., Profiles. Neurorehabilitation and neural repair.
Whitson, J., Clifton, G.L., and Hayes, R.L. (1995). 2015 May 15. Advance online publication.
Basal and scopolamine-evoked release of hippo- Failla, M.D., Kumar, R.G., Peitzman, A.B., Conley,
campal acetylcholine following traumatic brain Y.P., Ferrell, R.E., and Wagner, A.K. (2015b).
injury in rats. Neurosci. Lett. 198, 111114. Variation in the BDNF gene interacts with age to
Dixon, C.E., Ma, X., and Marion, D.W. (1997). Effects predict mortality in a prospective, longitudinal
of CDP-choline treatment on neurobehavioral cohort with severe TBI. Neurorehabil. Neural
deficits after TBI and on hippocampal and neo- Repair 29, 234246.
cortical acetlycholine release. J. Neurotrauma 14, Fann, J.R., Hart, T., and Schomer, K.G. (2009).
161169. Treatment for depression after traumatic brain
Dixon, C.E., Lyeth, B.G., Povlishock, J.T., Findling, R.L., injury:Asystematic review. J. Neurotrauma 26,
Hamm, R.J., Marmarou, A., Young, H.F., and 23832402.
Hayes, R.L. (1987). A fluid percussion model of Fawcett, J.W., and Asher, R.A. (1999). The glial scar
experimental brain injury in the rat. J Neurosurg. and central nervous system repair. Brain Res.
67, 110119. Bull. 49, 377391.
Duffau, H. (2006). Brain plasticity:from pathophysi- Fedele, D.E., Li, T., Lan, J.Q., Fredholm, B.B., and
ological mechanisms to therapeutic applica- Boison, D. (2006). Adenosine A1 receptors are
tions. J. Clin. Neurosci. Off. J. Neurosurg. Soc. crucial in keeping an epileptic focus localized.
Australas. 13, 885897. Exp. Neurol. 200, 184190.
Edwards, P., Arango, M., Balica, L., Cottingham, R., Feuerstein, G.Z., Wang, X., and Barone, F.C.
El-Sayed, H., Farrell, B., Fernandes, J., (1998). The role of cytokines in the
440 Part IV:Homeostatic Therapies

neuropathology of stroke and neurotrauma. cytokines and chemokines. Neurochem. Res. 23,
Neuroimmunomodulation 5, 143159. 329340.
Filloux, F., and Wamsley, J.K. (1991). Dopaminergic Giacino, J.T., Whyte, J., Bagiella, E., Kalmar, K., Childs,
modulation of excitotoxicity in rat striatum: N., Khademi, A., Eifert, B., Long, D., Katz, D.I.,
Evidence from nigrostriatal lesions. Synapse 8, Cho, S., etal. (2012). Placebo-controlled trial of
281288. amantadine for severe traumatic brain injury. N.
Fischer, S., Wobben, M., Marti, H.H., Renz, D., and Engl. J.Med. 366, 819826.
Schaper, W. (2002). Hypoxia-induced hyperper- Giulian, D., Li, J., Li, X., George, J., and Rutecki, P.A.
meability in brain microvessel endothelial cells (1994). The impact of microglia-derived cyto-
involves VEGF-mediated changes in the expres- kines upon gliosis in the CNS. Dev. Neurosci. 16,
sion of zonula occludens-1. Microvasc. Res. 128136.
63,7080. Globus, M.Y., Ginsberg, M.D., Dietrich, W.D., Busto,
Fleckenstein, A.E., Volz, T.J., Riddle, E.L., Gibb, J.W., R., and Scheinberg, P. (1987). Substantia nigra
and Hanson, G.R. (2007). New insights into the lesion protects against ischemic damage in the
mechanism of action of amphetamines. Annu. striatum. Neurosci. Lett. 80, 251256.
Rev. Pharmacol. Toxicol. 47, 681698. Gloth, F.M. III, and Tobin, J.D. (1995). Vitamin D
Fredholm, B.B., Chen, J.-F., Cunha, R.A., deficiency in older people. J. Am. Geriatr. Soc.
Svenningsson, P., and Vaugeois, J.-M. (2005). 43,822.
Adenosine and brain function. Int. Rev. Gltzner, F.L., Haubitz, I., Miltner, F., Kapp, G.,
Neurobiol. 63, 191270. and Pflughaupt, K.W. (1983). [Seizure preven-
Gao, W.M., Chadha, M.S., Kline, A.E., Clark, R.S., tion using carbamazepine following severe brain
Kochanek, P.M., Dixon, C.E., & Jenkins, L.W. injuries]. Neurochirurgia (Stuttg.) 26,6679.
(2006). Immunohistochemical analysis of his- Godwin-Austen, R.B., Frears, C.C., Bergmann,
tone H3 acetylation and methylationevidence S., Parkes, J.D., and Knill-Jones, R.P. (1970).
for altered epigenetic signaling following trau- Combined treatment of Parkinsonism with
matic brain injury in immature rats. Brain L-dopa and amantadine. Lancet 296, 383385.
research, 1070,3134. Golarai, G., Greenwood, A.C., Feeney, D.M., and
Garcia, A.N., Shah, M.A., Dixon, C.E., Wagner, A.K., Connor, J.A. (2001). Physiological and structural
and Kline, A.E. (2011). Biologic and plastic effects evidence for hippocampal involvement in persis-
of experimental traumatic brain injury treat- tent seizure susceptibility after traumatic brain
ment paradigms and their relevance to clinical injury. J. Neurosci. 21, 85238537.
rehabilitation. PM R 3, S18S27. Goldberg, T.E., Bigelow, L.B., Weinberger, D.R.,
Garringer, J.A., Niyonkuru, C., McCullough, E.H., Daniel, D.G., and Kleinman, J.E. (1991).
Loucks, T., Dixon, C.E., Conley, Y.P., Berga, S., and Cognitive and behavioral effects of the coadmin-
Wagner, A.K. (2013). Impact of aromatase genetic istration of dextroamphetamine and haloperidol
variation on hormone levels and global outcome in schizophrenia. Am. J.Psychiatry 148,7884.
after severe TBI. J. Neurotrauma 30, 14151425. Granado, N., Ortiz, O., Surez, L.M., Martn, E.D.,
Gatt, J.M., Nemeroff, C.B., Dobson-Stone, C., Paul, Cea, V., Sols, J.M., and Moratalla, R. (2008). D1
R.H., Bryant, R.A., Schofield, P.R., Gordon, E., but not D5 dopamine receptors are critical for
Kemp, A.H., and Williams, L.M. (2009). LTP, spatial learning, and LTP-induced arc and
Interactions between BDNF Val66Met poly- zif268 expression in the hippocampus. Cereb.
morphism and early life stress predict brain and Cortex 18,112.
arousal pathways to syndromal depression and Greengard, P., Allen, P.B., and Nairn, A.C. (1999).
anxiety. Mol. Psychiatry 14, 681695. Beyond the dopamine receptor:The DARPP-32/
Gemmell, C., and Leathem, J.M. (2006). A protein phosphatase-1 cascade. Neuron 23,
study investigating the effects of Tai Chi 435447.
Chuan:Individuals with traumatic brain injury Greve, M.W., and Zink, B.J. (2009). Pathophysiology
compared to controls. Brain Inj. 20, 151156. of traumatic brain injury. Mt. Sinai J. Med. 76,
Gennarelli, T.A. (1993). Mechanisms of brain injury. 97104.
J. Emerg. Med. 11 Suppl 1,511. Griesbach, G.S., Hovda, D.A., and Gomez-Pinilla, F.
Gentry, L., Godersky, J., and Thompson, B. (1988). (2009). Exercise-induced improvement in cogni-
MR imaging of head trauma:Review of the dis- tive performance after traumatic brain injury in
tribution and radiopathologic features of trau- rats is dependent on BDNF activation. Brain Res.
matic lesions. Am. J.Roentgenol. 150, 663672. 1288, 105115.
Ghirnikar, R.S., Lee, Y.L., and Eng, L.F. (1998). Griesbach, G.S., Hovda, D.A., Molteni, R., Wu, A.,
Inflammation in traumatic brain injury:Role of and Gomez-Pinilla, F. (2004). Voluntary exercise
Traumatic BrainInjury 441

following traumatic brain injury: brain-derived Haslam, C., Hodder, K.I., and Yates, P.J. (2011).
neurotrophic factor upregulation and recovery Errorless learning and spaced retrieval:How do
of function. Neuroscience 125, 129139. these methods fare in healthy and clinical popu-
Griffin, S.L., van Reekum, R., and Masanic, C. (2003). lations? J. Clin. Exp. Neuropsychol. 33, 432447.
A review of cholinergic agents in the treatment Hastings, T.G. (1995). Enzymatic oxidation of dopa-
of neurobehavioral deficits following traumatic mine: The role of prostaglandin H synthase. J.
brain injury. J. Neuropsychiatry Clin. Neurosci. Neurochem. 64, 919924.
15,1726. Headrick, J.P., Bendall, M.R., Faden, A.I., and Vink, R.
Grundy, P.L., Harbuz, M.S., Jessop, D.S., (1994). Dissociation of Adenosine levels from
Lightman, S.L., and Sharples, P.M. (2001). The bioenergetic state in experimental brain
hypothalamo-pituitary-adrenal axis response trauma: Potential role in secondary injury. J.
to experimental traumatic brain injury. J. Cereb. Blood Flow Metab. 14, 853861.
Neurotrauma 18, 13731381. Hekmatpanah, J., and Hekmatpanah, C.R. (1985).
Gualtieri, C.T., and Evans, R.W. (1988). Stimulant Micro vascular alterations following cerebral
treatment for the neurobehavioural sequelae of contusion in rats:Light, scanning, and electron
traumatic brain injury. Brain Inj. 2, 273290. microscope study. J. Neurosurg. 62, 888897.
Gunter, T.E., Gunter, K.K., Sheu, S.S., and Gavin, Hensler, T., Sauerland, S., Bouillon, B., Raum, M.,
C.E. (1994). Mitochondrial calcium trans- Rixen, D., Helling, H.-J., Andermahr, J., and
port: Physiological and pathological relevance. Neugebauer, E.A.M. (2002). Association between
Am. J.Physiol. 267, C313C339. injury pattern of patients with multiple inju-
Hadjikhani, N., del Rio, M.S., Wu, O., Schwartz, D., ries and circulating levels of soluble tumor
Bakker, D., Fischl, B., Kwong, K.K., Cutrer, necrosis factor receptors, interleukin-6 and
F.M., Rosen, B.R., and Tootell, R.B. (2001). interleukin-10, and polymorphonuclear neutro-
Mechanisms of migraine aura revealed by func- phil elastase. J. Trauma 52, 962970.
tional MRI in human visual cortex. Proc. Natl. Hetman, M., Kanning, K., Cavanaugh, J.E., and Xia, Z.
Acad. Sci. U.S.A. 98, 46874692. (1999). Neuroprotection by brain-derived neu-
Halliday, A.L. (1999). Pathophysiology. In: Traumatic rotrophic factor is mediated by extracellular
Brain Injury. Marion, DW, ed. (New York: signal-regulated kinase and phosphatidylinositol
Thieme Medical), pp.2938. 3-kinase. J. Biol. Chem. 274, 2256922580.
Halliwell, B. (1991). Reactive oxygen species in liv- Hicks, R.R., Smith, D.H., Lowenstein, D.H., Saint
ing systems: Source, biochemistry, and role in Marie, R., and McIntosh, T.K. (1993). Mild exper-
human disease. Am. J.Med. 91, S14S22. imental brain injury in the rat induces cognitive
Haltiner, A.M., Temkin, N.R., and Dikmen, S.S. deficits associated with regional neuronal loss in
(1997). Risk of seizure recurrence after the first the hippocampus. J. Neurotrauma 10, 405414.
late posttraumatic seizure. Arch. Phys. Med. Hillman, C.H., Erickson, K.I., and Kramer, A.F.
Rehabil. 78, 835840. (2008). Be smart, exercise your heart: Exercise
Hamani, C., Mayberg, H., Stone, S., Laxton, A., effects on brain and cognition. Nat. Rev.
Haber, S., and Lozano, A.M. (2011). The sub- Neurosci. 9,5865.
callosal cingulate gyrus in the context of major Hoffman, J.M., Bell, K.R., Powell, J.M., Behr, J.,
depression. Biol. Psychiatry 69, 301308. Dunn, E.C., Dikmen, S., and Bombardier, C.H.
Harel, Z., Gascon, G., Riggs, S., Vaz, R., Brown, W., (2010). A randomized controlled trial of exercise
and Exil, G. (2002). Supplementation with to improve mood after traumatic brain injury.
omega-3 polyunsaturated fatty acids in the man- PM R 2, 911919.
agement of recurrent migraines in adolescents. J. Hoh, N.Z., Wagner, A.K., Alexander, S.A., Clark,
Adolesc. Health 31, 154161. R.B., Beers, S.R., Okonkwo, D.O., Ren, D., and
Harun, R., and Wagner, A.K. (2014). The neu- Conley, Y.P. (2010). BCL2 Genotypes:Functional
robiological basis of pharmacological and neurobehavioral outcomes after severe trau-
approaches for patients with traumatic brain matic brain injury. J. Neurotrauma 27, 14131427.
injury. In: Understanding Traumatic Brain Honea, R., Verchinski, B.A., Pezawas, L., Kolachana,
Injury:Current Research and Future Directions. B.S., Callicott, J.H., Mattay, V.S., Weinberger, D.R.,
Levin, H.S., Shum, D.H.K., and Chan, R.C.K, eds. and Meyer-Lindenberg, A. (2009). Impact of
(Oxford:Oxford University Press), pp. 255300. interacting functional variants in COMT on
Hashimoto, K. (2010). Brain-derived neurotrophic regional gray matter volume in human brain.
factor as a biomarker for mood disorders: An Neuroimage 45,4451.
historical overview and future directions. Horai, R., Saijo, S., Tanioka, H., Nakae, S., Sudo, K.,
Psychiatry Clin. Neurosci. 64, 341357. Okahara, A., Ikuse, T., Asano, M., and
442 Part IV:Homeostatic Therapies

Iwakura, Y. (2000). Development of chronic Juengst, S.B., Kumar, R.G., Failla, M.D., Goyal, A.,
inflammatory arthropathy resembling rheu- and Wagner, A.K. (2014). Acute inflamma-
matoid arthritis in interleukin 1 receptor tory biomarker profiles predict depression risk
antagonistdeficient mice. J. Exp. Med. 191, following moderate to severe traumatic brain
313320. injury. J. Head Trauma Rehabil. 30, 207218.
Horsfield, S.A., Rosse, R.B., Tomasino, V., Schwartz, Kabat-Zinn, J. (1982). An outpatient program in
B.L., Mastropaolo, J., and Deutsch, S.I. (2002). behavioral medicine for chronic pain patients
Fluoxetines effects on cognitive performance based on the practice of mindfulness medita-
in patients with traumatic brain injury. Int. tion:Theoretical considerations and preliminary
J.Psychiatry Med. 32, 337344. results. Gen. Hosp. Psychiatry 4,3347.
Hudak, A., Warner, M., Marquez de la Plata, C., Kabat-Zinn, J. (1994). Wherever You Go, There You
Moore, C., Harper, C., and Diaz-Arrastia, R. Are: Mindfulness Meditation in Everyday Life
(2011). Brain morphometry changes and depres- (NewYork:Hyperion).
sive symptoms after traumatic brain injury. Kabat-Zinn, J., Lipworth, L., and Burney, R. (1985).
Psychiatry Res. 191, 160165. The clinical use of mindfulness meditation for
Huisman, T.A.G.M., Schwamm, L.H., Schaefer, P.W., the self-regulation of chronic pain. J. Behav.
Koroshetz, W.J., Shetty-Alva, N., Ozsunar, Y., Med. 8, 163190.
Wu, O., and Sorensen, A.G. (2004). Diffusion Kaelin, D.L., Cifu, D.X., and Matthies, B. (1996).
tensor imaging as potential biomarker of white Methylphenidate effect on attention deficit in
matter injury in diffuse axonal injury. Am. the acutely brain-injured adult. Arch. Phys. Med.
J.Neuroradiol. 25, 370376. Rehabil. 77,69.
Jackson, D.M., Martin, L.P., Larsson, L.G., Cox, R.F., Kalantari, H., and Das, D.K. (2010). Physiological
Waszczak, B.L., and Ross, S.B. (1990). effects of resveratrol. BioFactors 36, 401406.
Behavioural, biochemical and electrophysi- Kalantaridou, S.N., Makrigiannakis, A., Zoumakis,
ological studies on the motor depressant and E., and Chrousos, G.P. (2004). Stress and the
stimulant effects of bromocriptine. Naunyn. female reproductive system. J. Reprod. Immunol.
Schmiedebergs Arch. Pharmacol. 342, 290299. 62,6168.
Jaenisch, R., and Bird, A. (2003). Epigenetic regula- Kalish, H., and Phillips, T.M. (2010). Analysis of
tion of gene expression: How the genome inte- neurotrophins in human serum by immunoaf-
grates intrinsic and environmental signals. Nat. finity capillary electrophoresis (ICE) following
Genet. 33, 245254. traumatic head injury. J. Chromatogr. B Analyt.
Janjua, N.A., Mori, A., and Hiramatsu, M. (1990). Technol. Biomed. Life Sci. 878,194.
Increased aspartic acid release from the Karege, F., Perret, G., Bondolfi, G., Schwald, M.,
iron-induced epileptogenic focus. Epilepsy Res. Bertschy, G., and Aubry, J.-M. (2002). Decreased
6, 215220. serum brain-derived neurotrophic factor levels
Jeong, H.-K., Ji, K., Min, K., and Joe, E.-H. (2013). in major depressed patients. Psychiatry Res. 109,
Brain inflammation and microglia: Facts and 143148.
misconceptions. Exp. Neurobiol. 22,5967. Kasahara, M., Menon, D.K., Salmond, C.H.,
Johansson, B., Bjuhr, H., and Rnnbck, L. (2012). Outtrim, J.G., Tavares, J.V.T., Carpenter, T.A.,
Mindfulness-based stress reduction (MBSR) Pickard, J.D., Sahakian, B.J., and Stamatakis,
improves long-term mental fatigue after stroke or E.A. (2011). Traumatic brain injury alters the
traumatic brain injury. Brain Inj. 26, 16211628. functional brain network mediating working
Johnson, V.E., Stewart, J.E., Begbie, F.D., memory. Brain Inj. 25, 11701187.
Trojanowski, J.Q., Smith, D.H., and Stewart, W. Kaschel, R., Sala, S.D., Cantagallo, A., Fahlbck, A.,
(2013). Inflammation and white matter degen- Laaksonen, R., and Kazen, M. (2002). Imagery
eration persist for years after a single traumatic mnemonics for the rehabilitation of mem-
brain injury. Brain J.Neurol. 136,2842. ory: A randomised group controlled trial.
Jokinen, P., Karrasch, M., Brck, A., Johansson, J., Neuropsychol. Rehabil. 12, 127153.
Bergman, J., and Rinne, J.O. (2013). Cognitive Kennedy, K.M., Rodrigue, K.M., Land, S.J., and
slowing in Parkinsons disease is related to fron- Raz, N. (2009). BDNF Val66Met polymor-
tostriatal dopaminergic dysfunction. J. Neurol. phism influences age differences in micro-
Sci. 329,2328. structure of the corpus callosum. Front. Hum.
Jorge, R.E., Acion, L., Starkstein, S.E., and Magnotta, V. Neurosci.3,19.
(2007). Hippocampal volume and mood disor- Kim, H.-J., Lee, J.-H., and Kim, S.-H. (2010).
ders after traumatic brain injury. Biol. Psychiatry Therapeutic effects of human mesenchymal stem
62, 332338. cells on traumatic brain injury in rats:Secretion
Traumatic BrainInjury 443

of neurotrophic factors and inhibition of apopto- severe TBI and associations with 6 and 12month
sis. J. Neurotrauma 27, 131138. outcome. J. Head Trauma Rehabil. Advance
Kim, S.N., Kang, D.-H., Yun, J.-Y., Lee, T.Y., Jung, W.H., online publication.
Jang, J.H., and Kwon, J.S. (2013). Impact of the Lancelot, E., Lecanu, L., Revaud, M.L., Boulu,
BDNF Val66Met polymorphism on regional R.G., Plotkine, M., and Callebert, J. (1998).
brain gray matter volumes: Relevance to the Glutamate induces hydroxyl radical formation
stress response. Psychiatry Investig. 10, 173179. in vivo via activation of nitric oxide synthase
Kim, Y.-H., Ko, M.-H., Na, S.-Y., Park, S.-H., and in Sprague-Dawley rats. Neurosci. Lett. 242,
Kim, K.-W. (2006). Effects of single-dose methyl- 131134.
phenidate on cognitive performance in patients Lanctt, K.L., Rapoport, M.J., Chan, F., Rajaram,
with traumatic brain injury: A double-blind R.D., Strauss, J., Sicard, T., McCullagh, S.,
placebo-controlled study. Clin. Rehabil. Feinstein, A., Kiss, A., Kennedy, J.L., etal. (2010).
20,2430. Genetic predictors of response to treatment with
Kirchhoff, C., Stegmaier, J., Bogner, V., Buhmann, S., citalopram in depression secondary to traumatic
Mussack, T., Kreimeier, U., Mutschler, W., and brain injury. Brain Inj. 24, 959969.
Biberthaler, P. (2006). Intrathecal and systemic Larsen, C.M., Faulenbach, M., Vaag, A., Vlund, A.,
concentration of NT-proBNP in patients with Ehses, J.A., Seifert, B., Mandrup-Poulsen, T.,
severe traumatic brain injury. J. Neurotrauma and Donath, M.Y. (2007). Interleukin-1receptor
23, 943949. antagonist in type 2 diabetes mellitus. N. Engl.
Kleim, J.A., Barbay, S., Cooper, N.R., Hogg, T.M., J.Med. 356, 15171526.
Reidel, C.N., Remple, M.S., and Nudo, R.J. Lauritzen, M., Dreier, J.P., Fabricius, M., Hartings,
(2002). Motor learning-dependent synaptogene- J.A., Graf, R., and Strong, A.J. (2011). Clinical rel-
sis is localized to functionally reorganized motor evance of cortical spreading depression in neu-
cortex. Neurobiol. Learn. Mem. 77,6377. rological disorders:migraine, malignant stroke,
Kline, A.E., Yu, J., Massucci, J.L., Zafonte, R.D., subarachnoid and intracranial hemorrhage, and
and Dixon, C.E. (2002). Protective effects of the traumatic brain injury. J. Cereb. Blood Flow
5-HT1A receptor agonist 8-hydroxy-2-(di-n- Metab. 31,1735.
propylamino)tetralin against traumatic brain Lee, H., Kim, S.-W., Kim, J.-M., Shin, I.-S., Yang,
injury-induced cognitive deficits and neuropa- S.-J., and Yoon, J.-S. (2005). Comparing effects of
thology in adult male rats. Neurosci. Lett. 333, methylphenidate, sertraline and placebo on neu-
179182. ropsychiatric sequelae in patients with traumatic
Kobori, N., Clifton, G.L., and Dash, P.K. (2006). brain injury. Hum. Psychopharmacol. Clin. Exp.
Enhanced catecholamine synthesis in the 20, 97104.
prefrontal cortex after traumatic brain Lee, J.H., Martin, N.A., Alsina, G., McArthur, D.L.,
injury: Implications for prefrontal dysfunction. Zaucha, K., Hovda, D.A., and Becker, D.P. (1997).
J. Neurotrauma 23, 10941102. Hemodynamically significant cerebral vaso-
Kovalchuk, Y., Hanse, E., Kafitz, K.W., and spasm and outcome after head injury: A pro-
Konnerth, A. (2002). Postsynaptic induction of spective study. J. Neurosurg. 87, 221233.
BDNF-mediated long-term potentiation. Science Levant, B. (2011). N-3 (omega-3) Fatty acids in post-
295, 17291734. partum depression: Implications for prevention
Kozlowski, D.A., James, D.C., and Schallert, T. and treatment. Depress. Res. Treat. 2011, 467349.
(1996). Use-dependent exaggeration of neuro- Levin, H., Shum, D., and Chan, R. (2014).
nal injury after unilateral sensorimotor cortex Understanding Traumatic Brain Injury:Current
lesions. J. Neurosci. 16, 47764786. Research and Future Directions (Oxford:Oxford
Kraio, R.P., and Nicholson, C. (1978). Extracellular University Press).
ionic variations during spreading depression. Levin, H.S., Peters, B.H., Kalisky, Z., High, W.M. Jr,
Neuroscience 3, 10451059. Von Laufen, A., Eisenberg, H.M., Morrison, D.P.,
Krishnan, V., and Nestler, E.J. (2008). The molecu- and Gary, H.E. Jr.(1986). Effects of oral physo-
lar neurobiology of depression. Nature 455, stigmine and lecithin on memory and attention
894902. in closed head-injured patients. Cent. Nerv. Syst.
Kruit, M.C., van Buchem, M.A., Hofman, P.M., Trauma 3, 333342.
Bakkers, J.T., Terwindt, G.M., Ferrari, M.D., and Lew, H.L., Lin, P.-H., Fuh, J.-L., Wang, S.-J., Clark,
Launer, L.J. (2004). Migraine as a risk factor for D.J., and Walker, W.C. (2006). Characteristics
subclinical brain lesions. JAMA 291, 427434. and treatment of headache after traumatic brain
Kumar, RG., Boles, JA., and Wagner, AK. (2014). injury: A focused review. Am. J. Phys. Med.
Chronic inflammation characterization after Rehabil. 85, 619627.
444 Part IV:Homeostatic Therapies

Liao, Z.B., Zhi, X.G., Shi, Q.H., and He, Z.H. (2008). after traumatic brain injury. Cephalalgia 32,
Recombinant human erythropoietin adminis- 600606.
tration protects cortical neurons from traumatic Lucas, S.-M., Rothwell, N.J., and Gibson, R.M. (2006).
brain injury in rats. Eur. J.Neurol. 15, 140149. The role of inflammation in CNS injury and dis-
Lieberman, A.N., and Goldstein, M. (1985). ease. Br. J.Pharmacol. 147 Suppl 1, S232S240.
Bromocriptine in Parkinson disease. Pharmacol. Lundberg, J., Karimi, M., von Gertten, C., Holmin, S.,
Rev. 37, 217227. Ekstrm, T. J., & Sandberg-Nordqvist, A. C.
Lieberman, S.A., Oberoi, A.L., Gilkison, C.R., Masel, (2009). Traumatic brain injury induces relo-
B.E., and Urban, R.J. (2001). Prevalence of neu- calization of DNA-methyltransferase 1.
roendocrine dysfunction in patients recovering Neuroscience letters, 457,811.
from traumatic brain injury. J. Clin. Endocrinol. Lyeth, B.G., Jenkins, L.W., Hamm, R.J., Dixon, C.E.,
Metab. 86, 27522756. Phillips, L.L., Clifton, G.L., Young, H.F., and
Lima, F.D., Oliveira, M.S., Furian, A.F., Souza, Hayes, R.L. (1990). Prolonged memory impair-
M.A., Rambo, L.M., Ribeiro, L.R., Silva, L.F.A., ment in the absence of hippocampal cell death
Retamoso, L.T., Hoffmann, M.S., Magni, D.V., following traumatic brain injury in the rat. Brain
et al. (2009). Adaptation to oxidative challenge Res. 526, 249258.
induced by chronic physical exercise prevents Maas, A.I., Stocchetti, N., and Bullock, R. (2008).
Na+,K+-ATPase activity inhibition after trau- Moderate and severe traumatic brain injury in
matic brain injury. Brain Res. 1279, 147155. adults. Lancet Neurol. 7, 728741.
Linde, K., Allais, G., Brinkhaus, B., Manheimer, E., Maas, A.I.R., Roozenbeek, B., and Manley, G.T.
Vickers, A., and White, A.R. (1996). Acupuncture (2010). Clinical trials in traumatic brain
for migraine prophylaxis. Cochrane Database injury: Past experience and current develop-
Syst Rev. 21, CD001218. ments. Neurotherapeutics 7, 115126.
Linde, K., Streng, A., Jrgens, S., Hoppe, A., Brinkhaus, MacKenzie, J.D., Siddiqi, F., Babb, J.S., Bagley, L.J.,
B., Witt, C., Wagenpfeil, S., Pfaffenrath, V., Mannon, L.J., Sinson, G.P., and Grossman, R.I.
Hammes, M.G., and Weidenhammer, W. (2005). (2002). Brain atrophy in mild or moderate trau-
Acupuncture for patients with migraine: A ran- matic brain injury: A longitudinal quantitative
domized controlled trial. JAMA 293, 21182125. analysis. Am. J.Neuroradiol. 23, 15091515.
Lips, P., Hosking, D., Lippuner, K., Norquist, J.M., Maier, B., Schwerdtfeger, K., Mautes, A., Holanda, M.,
Wehren, L., Maalouf, G., Ragi-Eis, S., and Mller, M., Steudel, W.I., and Marzi, I. (2001).
Chandler, J. (2006). The prevalence of vitamin D Differential release of interleukines 6, 8, and 10
inadequacy amongst women with osteoporosis: in cerebrospinal fluid and plasma after traumatic
An international epidemiological investigation. brain injury. Shock 15, 421426.
J. Intern. Med. 260, 245254. Maier, B., Laurer, H.-L., Rose, S., Buurman, W.A.,
Loh, L., Nathan, P.W., Schott, G.D., and Zilkha, K.J. and Marzi, I. (2005). Physiological levels of pro-
(1984). Acupuncture versus medical treatment and anti-inflammatory mediators in cerebro-
for migraine and muscle tension headaches. J. spinal fluid and plasma: A normative study. J.
Neurol. Neurosurg. Psychiatry 47, 333337. Neurotrauma 22, 822835.
Longoni, M., and Ferrarese, C. (2006). Inflammation Maier, S.F. (2003). Bi-directional immunebrain
and excitotoxicity: role in migraine pathogen- communication: Implications for understand-
esis. Neurol. Sci. 27, s107s110. ing stress, pain, and cognition. Brain. Behav.
Lotrich, F.E., Sears, B., and McNamara, R.K. (2013). Immun. 17,6985.
Elevated ratio of arachidonic acid to long-chain Majerske, C.W., Mihalik, J.P., Ren, D., Collins, M.W.,
omega-3 fatty acids predicts depression devel- Reddy, C.C., Lovell, M.R., and Wagner, A.K.
opment following interferon-alpha treat- (2008). Concussion in sports: Postconcussive
ment: Relationship with interleukin-6. Brain. activity levels, symptoms, and neurocognitive
Behav. Immun. 31,4853. performance. J. Athl. Train. 43,265.
Lovell, M., Collins, M., and Bradley, J. (2004). Return Malec, J., Jones, R., Rao, N., and Stubbs, K. (1984).
to play following sports-related concussion. Clin. Video game practice effects on sustained atten-
Sports Med. 23, 421441. tion in patients with craniocerebral trauma.
Lucas, S. (2011). Headache management in concus- Cogn. Rehabil. 2,1823.
sion and mild traumatic brain injury. PM R 3, Manaka, S. (1992). Cooperative prospective study
S406S412. on posttraumatic epilepsy: Risk factors and
Lucas, S., Hoffman, J.M., Bell, K.R., Walker, W., and the effect of prophylactic anticonvulsant. Jpn.
Dikmen, S. (2012). Characterization of headache J.Psychiatry Neurol. 46, 311315.
Traumatic BrainInjury 445

Marchi, N., Bazarian, J.J., Puvenna, V., Janigro, M., concentrations after experimental brain injury
Ghosh, C., Zhong, J., Zhu, T., Blackman, E., in the rat. J. Neurochem. 63, 14261433.
Stewart, D., Ellis, J., etal. (2013). Consequences McKee, A.C., Stein, T.D., Nowinski, C.J., Stern, R.A.,
of repeated blood-brain barrier disruption in Daneshvar, D.H., Alvarez, V.E., Lee, H.-S., Hall, G.,
football players. PLoS One 8, e56805. Wojtowicz, S.M., and Baugh, C.M. (2013).
Marmigre, F., Givalois, L., Rage, F., Arancibia, S., The spectrum of disease in chronic traumatic
and Tapia-Arancibia, L. (2003). Rapid induction encephalopathy. Brain 136,4364.
of BDNF expression in the hippocampus during McQueen,J.K.,Blackwood,D.H.,Harris,P.,Kalbag,R.M.,
immobilization stress challenge in adult rats. and Johnson, A.L. (1983). Low risk of late
Hippocampus 13, 646655. post-traumatic seizures following severe head
Maruya, H., Watanabe, Y., Okita, M., Lawlor, G.F., injury: Implications for clinical trials of prophy-
Utsumi, H., and Niitsuma, T. (2003). Inhibitory laxis. J. Neurol. Neurosurg. Psychiatry 46, 899904.
effects of D2 agonists by striatal injection on Metman, L.V., Del Dotto, P., Van Den Munckhof, P.,
excessive release of dopamine and hyperactivity Fang, J., Mouradian, M.M., and Chase, T.N.
induced by Bay K 8644 in rats. Neuroscience 118, (1998). Amantadine as treatment for dyskinesias
10911098. and motor fluctuations in Parkinsons disease.
Massucci, J.L., Kline, A.E., Ma, X., Zafonte, R.D., Neurology 50, 13231326.
and Dixon, C.E. (2004). Time dependent altera- Mettenburg, J.M., Benzinger, T.L., Shimony, J.S.,
tions in dopamine tissue levels and metabolism Snyder, A.Z., and Sheline, Y.I. (2012). Diminished
after experimental traumatic brain injury in rats. performance on neuropsychological testing in
Neurosci. Lett. 372, 127131. late life depression is correlated with microstruc-
May, A.K., Dossett, L.A., Norris, P.R., Hansen, E.N., tural white matter abnormalities. Neuroimage
Dorsett, R.C., Popovsky, K.A., and Sawyer, R.G. 60, 21822190.
(2008). Estradiol is associated with mortality in Miller, A.H., Maletic, V., and Raison, C.L. (2009).
critically ill trauma and surgical patients. Crit. Inflammation and its discontents: The role
Care Med. 36,6268. of cytokines in the pathophysiology of major
Mayberg, H.S. (2003). Modulating dysfunctional depression. Biol. Psychiatry 65, 732741.
limbic-cortical circuits in depression: Towards Miller, M.A., Conley, Y., Scanlon, J.M., Ren, D., Ilyas
development of brain-based algorithms for diag- Kamboh, M., Niyonkuru, C., and Wagner, A.K.
nosis and optimised treatment. Br. Med. Bull. 65, (2010). APOE genetic associations with seizure
193207. development after severe traumatic brain injury.
Mazzeo, A.T., Beat, A., Singh, A., and Bullock, M.R. Brain Inj. 24, 14681477.
(2009). The role of mitochondrial transition pore, Moffet, H.H. (2006). How might acupuncture work?
and its modulation, in traumatic brain injury A systematic review of physiologic rationales
and delayed neurodegeneration after TBI. Exp. from clinical trials. BMC Complement. Altern.
Neurol. 218, 363370. Med.6,25.
McAllister, T.W., Saykin, A.J., Flashman, L.A., Montag, C., Markett, S., Basten, U., Stelzel, C.,
Sparling, M.B., Johnson, S.C., Guerin, S.J., Fiebach, C., Canli, T., and Reuter, M. (2010).
Mamourian, A.C., Weaver, J.B., and Yanofsky, N. Epistasis of the DRD2/ANKK1 Taq Ia and
(1999). Brain activation during working memory the BDNF Val66Met polymorphism impacts
1 month after mild traumatic brain injury: A novelty seeking and harm avoidance.
functional MRI study. Neurology 53, 13001308. Neuropsychopharmacology 35, 18601867.
McCulloch, K. (2007). Attention and dual-task con- Morganti-Kossmann, M.C., Rancan, M., Stahel, P.F.,
ditions: Physical therapy implications for indi- and Kossmann, T. (2002). Inflammatory response
viduals with acquired brain injury: J. Neurol. in acute traumatic brain injury:Adouble-edged
Phys. Ther. 31, 104118. sword. Curr. Opin. Crit. Care 8, 101105.
McDowell, S., Whyte, J., and DEsposito, M. (1998). Morrow, B.A., Roth, R.H., and Elsworth, J.D. (2000).
Differential effect of a dopaminergic agonist on TMT, a predator odor, elevates mesoprefron-
prefrontal function in traumatic brain injury tal dopamine metabolic activity and disrupts
patients. Brain 121, 11551164. short-term working memory in the rat. Brain
McHugh, L., and Wood, R. (2013). Stimulus Res. Bull. 52, 519523.
over-selectivity in temporal brain injury: Moskowitz, M.A. (1993). Neurogenic inflamma-
Mindfulness as a potential intervention. Brain tion in the pathophysiology and treatment of
Inj. 27, 15951599. migraine. Neurology 43,S16.
McIntosh, T.K., Yu, T., and Gennarelli, T.A. (1994). Mukaino, Y., Park, J., White, A., and Ernst, E.
Alterations in regional brain catecholamine (2005). The effectiveness of acupuncture for
446 Part IV:Homeostatic Therapies

depressiona systematic review of randomised metabolism in comatose patients with acute head
controlled trials. Acupunct. Med. 23,7076. injury. J. Neurosurg. 61, 241253.
Munck, A., Guyre, P.M., and Holbrook, N.J. (1984). OCarroll, C.M., Martin, S.J., Sandin, J., Frenguelli,
Physiological functions of glucocorticoids in B., and Morris, R.G.M. (2006). Dopaminergic
stress and their relation to pharmacological modulation of the persistence of one-trial
actions. Endocr. Rev. 5,2544. hippocampus-dependent memory. Learn. Mem.
Murakami, S., Imbe, H., Morikawa, Y., Kubo, C., and 13, 760769.
Senba, E. (2005). Chronic stress, as well as acute Olver, J.H., Ponsford, J.L., and Curran, C.A. (1996).
stress, reduces BDNF mRNA expression in the Outcome following traumatic brain injury: a
rat hippocampus but less robustly. Neurosci. Res. comparison between 2 and 5 years after injury.
53, 129139. Brain Inj. 10, 841848.
Myer, D.J., Gurkoff, G.G., Lee, S.M., Hovda, D.A., Ozturk, E., Demirbilek, S., Kadir But, A., Saricicek,
and Sofroniew, M.V. (2006). Essential protective V., Gulec, M., Akyol, O., and Ozcan Ersoy, M.
roles of reactive astrocytes in traumatic brain (2005). Antioxidant properties of propofol and
injury. Brain 129, 27612772. erythropoietin after closed head injury in rats.
Narayanan, N.S., Rodnitzky, R.L., and Uc, E.Y. Prog. Neuropsychopharmacol. Biol. Psychiatry
(2013). Prefrontal dopamine signaling and cog- 29, 922927.
nitive symptoms of Parkinsons disease. Rev. Ozturk, E., Demirbilek, S., Krolu, A., But, A.,
Neurosci. 24, 267278. Bege, Z.., Glec, M., Akyol, ., and Ersoy,
National Center for Complementary and Alternative M.. (2008). Propofol and erythropoietin anti-
Medicine. (2014). Acupuncture. https://nccih. oxidant properties in rat brain injured tissue.
nih.gov/health/acupuncture#how. Prog. Neuropsychopharmacol. Biol. Psychiatry
Nekludov, M., Antovic, J., Bredbacka, S., and 32,8186.
Blombck, M. (2007). Coagulation abnormali- Pacheco, J., Beevers, C.G., Benavides, C., McGeary, J.,
ties associated with severe isolated traumatic Stice, E., and Schnyer, D.M. (2009). Frontal-limbic
brain injury:Cerebral arterio-venous differences white matter pathway associations with the
in coagulation and inflammatory markers. J. serotonin transporter gene promoter region
Neurotrauma 24, 174180. (5-HTTLPR) polymorphism. J. Neurosci. 29,
Newburn, G., Edwards, R., Thomas, H., Collier, J., 62296233.
Fox, K., and Collins, C. (1999). Moclobemide Park, Y., Moon, H.-J., and Kim, S.-H. (2012). N-3
in the treatment of major depressive disorder polyunsaturated fatty acid consumption pro-
(DSM-3) following traumatic brain injury. Brain duces neurobiological effects associated with
Inj. 13, 637642. prevention of depression in rats after the forced
Ng, T.-P., Chiam, P.-C., Lee, T., Chua, H.-C., Lim, L., swimming test. J. Nutr. Biochem. 23, 924928.
and Kua, E.-H. (2006). Curry consumption Pechadre, J.C., Lauxerois, M., Colnet, G., Commun, C.,
and cognitive function in the elderly. Am. Dimicoli, C., Bonnard, M., Gibert, J., and
J.Epidemiol. 164, 898906. Chabannes, J. (1991). [Prevention of late
Nimmo, A.J., Cernak, I., Heath, D.L., Hu, X., Bennett, post-traumatic epilepsy by phenytoin in severe
C.J., and Vink, R. (2004). Neurogenic inflamma- brain injuries:2years follow-up]. Presse Med 20,
tion is associated with development of edema 841845.
and functional deficits following traumatic brain Perino, C., Rago, R., Cicolini, A., Torta, R., and
injury in rats. Neuropeptides 38,4047. Monaco, F. (2001). Mood and behavioural dis-
Nishi, A., Bibb, J.A., Matsuyama, S., Hamada, M., orders following traumatic brain injury:Clinical
Higashi, H., Nairn, A.C., and Greengard, P. evaluation and pharmacological management.
(2002). Regulation of DARPP-32 dephosphory- Brain Inj. 15, 139148.
lation at PKA- and Cdk5-sites by NMDA and Pezawas, L., Meyer-Lindenberg, A., Drabant, E.M.,
AMPA receptors: Distinct roles of calcineurin Verchinski, B.A., Munoz, K.E., Kolachana,
and protein phosphatase-2A. J. Neurochem. 81, B.S., Egan, M.F., Mattay, V.S., Hariri, A.R., and
832841. Weinberger, D.R. (2005). 5-HTTLPR polymor-
Novack, T.A., Caldwell, S.G., Duke, L.W., Bergquist, phism impacts human cingulate-amygdala
T.F., and Gage, R.J. (1996). Focused versus interactions:Agenetic susceptibility mechanism
unstructured intervention for attention deficits for depression. Nat Neurosci 8, 828834.
after traumatic brain injury. J. Head Trauma Pezawas, L., Meyer-Lindenberg, A., Goldman, A.L.,
Rehabil. 11,5260. Verchinski, B.A., Chen, G., Kolachana, B.S., Egan,
Obrist, W.D., Langfitt, T.W., Jaggi, J.L., Cruz, J., and M.F., Mattay, V.S., Hariri, A.R., and Weinberger,
Gennarelli, T.A. (1984). Cerebral blood flow and D.R. (2008). MET BDNF protects against
Traumatic BrainInjury 447

morphological S allele effects of 5-HTTLPR. Rogers, E., and Wagner, A.K. (2006). Gender, sex ste-
Mol. Psychiatry 13,654. roids, and neuroprotection following traumatic
Pfenninger, E.G., Reith, A., Breitig, D., Grnert, A., brain injury. J. Head Trauma Rehabil. 21, 279281.
and Ahnefeld, F.W. (1989). Early changes of intra- Romanczyk, T.B., Weickert, C.S., Webster, M.J.,
cranial pressure, perfusion pressure, and blood Herman, M.M., Akil, M., and Kleinman, J.E.
flow after acute head injury. Part 1: An experi- (2002). Alterations in trkB mRNA in the human
mental study of the underlying pathophysiology. prefrontal cortex throughout the lifespan. Eur.
J. Neurosurg. 70, 774779. J.Neurosci. 15, 269280.
Ponsford, J., Sloan, S., and Snow, P. (2012). Traumatic Roof, R.L., Duvdevani, R., Braswell, L., and Stein,
Brain Injury: Rehabilitation for Everyday D.G. (1994). Progesterone facilitates cognitive
Adaptive Living (NewYork:Psychology Press). recovery and reduces secondary neuronal loss
Poole, N.A., and Agrawal, N. (2008). Cholinomimetic caused by cortical contusion injury in male rats.
agents and neurocognitive impairment following Exp. Neurol. 129,6469.
head injury: A systematic review. Brain Inj. 22, Rossi, C., Angelucci, A., Costantin, L., Braschi,
519534. C., Mazzantini, M., Babbini, F., Fabbri, M.E.,
Radua, J., El-Hage, W., Mont, G.C., Gohier, B., Tessarollo, L., Maffei, L., Berardi, N., etal. (2006).
Tropeano, M., Phillips, M.L., and Surguladze, S.A. Brain-derived neurotrophic factor (BDNF) is
(2014). COMT Val158Met x SLC6A4 5-HTTLPR required for the enhancement of hippocampal
interaction impacts on gray matter volume of neurogenesis following environmental enrich-
regions supporting emotion processing. Soc. ment. Eur. J.Neurosci. 24, 18501856.
Cogn. Affect. Neurosci. 9, 12321238. Rostami, E., Krueger, F., Plantman, S., Davidsson, J.,
Rangel-Castilla, L., Gasco, J., Nauta, H.J.W., Agoston, D., Grafman, J., and Risling, M. (2014).
Okonkwo, D.O., and Robertson, C.S. (2008). Alteration in BDNF and its receptors, full-length
Cerebral pressure autoregulation in traumatic and truncated TrkB and p75(NTR) following
brain injury. Neurosurg. Focus 25,E7. penetrating traumatic brain injury. Brain Res.
Rapoport, M.J., Chan, F., Lanctot, K., Herrmann, N., 1542, 195205.
McCullagh, S., and Feinstein, A. (2008). An Russell, K.C., Arenth, P.M., Scanlon, J.M., Kessler, L.,
open-label study of citalopram for major and Ricker, J.H. (2012). Hemispheric and execu-
depression following traumatic brain injury. J. tive influences on low-level language process-
Psychopharmacol. (Oxf.) 22, 860864. ing after traumatic brain injury. Brain Inj. 26,
Raskin, N.H. (1991). Serotonin receptors and head- 984995.
ache. N. Engl. J.Med. 325, 353354. Ryan, T.V., and Ruff, R.M. (1988). The efficacy of
Reinhard, M., Wehrle-Wieland, E., Roth, M., Niesen, structured memory retraining in a group com-
W.D., Timmer, J., Weiller, C., and Hetzel, A. parison of head trauma patients. Arch. Clin.
(2007). Preserved dynamic cerebral autoregula- Neuropsychol. 3, 165179.
tion in the middle cerebral artery among persons Saija, A., Hayes, R.L., Lyeth, B.G., Edward Dixon, C.,
with migraine. Exp. Brain Res. 180, 517523. Yamamoto, T., and Robinson, S.E. (1988). The
Ritter, A., Zhang, Z., Wang, K., and Wagner, A. effect of concussive head injury on central cho-
(2014). GFAP autoantibody developmental trajec- linergic neurons. Brain Res. 452, 303311.
tory after TBI:Characterization and associations Salem, N.Jr., Litman, B., Kim, H.-Y., and Gawrisch, K.
with interleukin-7. J. Neurotrauma 31, A40A40. (2001). Mechanisms of action of docosahexae-
Roberts, I., Yates, D., Sandercock, P., Farrell, B., noic acid in the nervous system. Lipids 36,
Wasserberg, J., Lomas, G., Cottingham, R., 945959.
Svoboda, P., Brayley, N., and Mazairac, G. Santarsieri, M., Niyonkuru, C., McCullough,
(2004). Effect of intravenous corticosteroids E.H., Dobos, J.A., Dixon, C.E., Berga, S.L., and
on death within 14 days in 10008 adults with Wagner, A.K. (2014). Cerebrospinal fluid cortisol
clinically significant head injury (MRC CRASH and progesterone profiles and outcomes prog-
trial): Randomised placebo-controlled trial. nostication after severe traumatic brain injury. J.
Lancet 364, 13211328. Neurotrauma 31, 699712.
Rodrguez-Baeza, A., Reina-de la Torre, F., Poca, A., Sapolsky, R.M., Romero, L.M., and Munck, A.U.
Mart, M., and Garnacho, A. (2003). (2000). How do glucocorticoids influence stress
Morphological features in human cortical responses? Integrating permissive, suppressive,
brain microvessels after head injury: A three- stimulatory, and preparative actions. Endocr.
dimensional and immunocytochemical study. Rev. 21,5589.
Anat. Rec. A. Discov. Mol. Cell. Evol. Biol. 273, Sarabia, R., Lobato, R.D., Rivas, J.J., Cordobs, F.,
583593. Rubio, J., Cabrera, A., Gomez, P., Muoz, M.J.,
448 Part IV:Homeostatic Therapies

and Madera, A. (1988). Cerebral hemisphere Shapira, Y., Setton, D., Artru, A.A., and Shohami, E.
swelling in severe head injury patients. Acta (1993). Blood-brain barrier permeability, cere-
Neurochir. Suppl. (Wien) 42,4046. bral edema, and neurologic function after closed
Saran, A.S. (1985). Depression after minor closed head injury in rats. Anesth. Analg. 77, 141148.
head injury:Role of dexamethasone suppression Shiozaki, T., Hayakata, T., Tasaki, O., Hosotubo,
test and antidepressants. J. Clin. Psychiatry 46, H., Fuijita, K., Mouri, T., Tajima, G., Kajino, K.,
335338. Nakae, H., Tanaka, H., etal. (2005). Cerebrospinal
Sawyer, E., Mauro, L.S., and Ohlinger, M.J. (2008). fluid concentrations of anti-inflammatory
Amantadine enhancement of arousal and mediators in early-phase severe traumatic brain
cognition after traumatic brain injury. Ann. injury. Shock 23, 406410.
Pharmacother. 42, 247252. Shlosberg, D., Benifla, M., Kaufer, D., and Friedman,
Scahill, L., Carroll, D., and Burke, K. (2004). A. (2010). Blood-brain barrier breakdown as a
Methylphenidate:Mechanism of action and clin- therapeutic target in traumatic brain injury. Nat.
ical update. J. Child Adolesc. Psychiatr. Nurs. Rev. Neurol. 6, 393403.
17,8586. Silva, C.G., Porcincula, L.O., Canas, P.M.,
Schierhout, G., and Roberts, I. (1996). Antiepileptic Oliveira, C.R., and Cunha, R.A. (2007).
drugs for preventing seizures following acute Blockade of adenosine A(2A) receptors prevents
traumatic brain injury. In Cochrane Database staurosporine-induced apoptosis of rat hippo-
Syst. Rev. 4, CD000173. campal neurons. Neurobiol. Dis. 27, 182189.
Schmidt, O.I., Heyde, C.E., Ertel, W., and Stahel, P.F. Simopoulos, A.P. (2002). Omega-3 fatty acids in
(2005). Closed head injuryan inflammatory inflammation and autoimmune diseases. J. Am.
disease? Brain Res. Brain Res. Rev. 48, 388399. Coll. Nutr. 21, 495505.
Schneider, H.J., Schneider, M., Saller, B., Petersenn, S., Singh, I.N., Sullivan, P.G., and Hall, E.D. (2007).
Uhr, M., Husemann, B., Von Rosen, F., and Stalla, Peroxynitrite-mediated oxidative damage to
G.K. (2006). Prevalence of anterior pituitary brain mitochondria: Protective effects of per-
insufficiency 3 and 12 months after traumatic oxynitrite scavengers. J. Neurosci. Res. 85,
brain injury. Eur. J.Endocrinol. 154, 259265. 22162223.
Schnieders, J., Willemsen, D., and de Boer, H. (2012). Singhal, A., Baker, A.J., Hare, G.M.T., Reinders, F.X.,
Factors contributing to chronic fatigue after Schlichter, L.C., and Moulton, R.J. (2002).
traumatic brain injury:J. Head Trauma Rehabil. Association between cerebrospinal fluid
27, 404412. interleukin-6 concentrations and outcome
Schober, M.E., Ke, X., Xing, B., Block, B.P., Requena, after severe human traumatic brain injury. J.
D.F., McKnight, R., Lane, R.H. (2012). Traumatic Neurotrauma 19, 929937.
brain injury increased IGF-1B mRNA and Skolnick, B.E., Maas, A.I., Narayan, R.K., van der
altered IGF-1 exon 5 and promoter region epi- Hoop, R.G., MacAllister, T., Ward, J.D., Nelson,
genetic characteristics in the rat pup hippocam- N.R., and Stocchetti, N. (2014). A clinical trial of
pus. 29, 20752085. progesterone for severe traumatic brain injury.
Schumacher, M., Guennoun, R., Robert, F., N. Engl. J.Med. 371, 24672476.
Carelli, C., Gago, N., Ghoumari, A., Gonzalez Soares, H.D., Hicks, R.R., Smith, D., and McIntosh,
Deniselle, M.C., Gonzalez, S.L., Ibanez, C., and T.K. (1995). Inflammatory leukocytic recruit-
Labombarda, F. (2004). Local synthesis and ment and diffuse neuronal degeneration are
dual actions of progesterone in the nervous sys- separate pathological processes resulting
tem:Neuroprotection and myelination. Growth from traumatic brain injury. J. Neurosci. 15,
Horm. IGF Res. 14,1833. 82238233.
Schumacher, M., Robel, P., and Baulieu, E. (1996). Sofroniew, M.V., Howe, C.L., and Mobley, W.C.
Development and regeneration of the nervous (2001). Nerve growth factor signaling, neuropro-
system: A role for neurosteroids (Part 1 of 2). tection, and neural repair. Annu. Rev. Neurosci.
Dev. Neurosci. 18,613. 24, 12171281.
Sen, S., Duman, R., and Sanacora, G. (2008). Serum Sohlberg, M.M., and Turkstra, L.S. (2011). Optimizing
brain-derived neurotrophic factor, depression, Cognitive Rehabilitation: Effective Instructional
and antidepressant medications: Meta-analyses Methods (NewYork:Guilford Press).
and implications. Biol. Psychiatry 64, 527532. Spencer, J.P.E. (2009). Flavonoids and brain
Shao, C., Roberts, K.N., Markesbery, W.R., Scheff, health:Multiple effects underpinned by common
S.W., and Lovell, M.A. (2006). Oxidative stress mechanisms. Genes Nutr. 4, 243250.
in head trauma in aging. Free Radic. Biol. Med. Spratt, D.I., Kramer, R.S., Morton, J.R., Lucas,
41,7785. F.L., Becker, K., and Longcope, C. (2008).
Traumatic BrainInjury 449

Characterization of a prospective human model Sullivan, P.G., Thompson, M.B., and Scheff, S.W.
for study of the reproductive hormone responses (1999). Cyclosporin A attenuates acute mito-
to major illness. Am. J. Physiol. Endocrinol. chondrial dysfunction following traumatic brain
Metab. 295, E63E69. injury. Exp. Neurol. 160, 226234.
Spratt, D.I., Morton, J.R., Kramer, R.S., Mayo, S.W., Suzuki, T., Bramlett, H.M., and Dietrich, W.D.
Longcope, C., and Vary, C.P.H. (2006). Increases (2003). The importance of gender on the benefi-
in serum estrogen levels during major illness cial effects of posttraumatic hypothermia. Exp.
are caused by increased peripheral aromatiza- Neurol. 184, 10171026.
tion. Am. J. Physiol. Endocrinol. Metab. 291, Szaflarski, J.P., Sangha, K.S., Lindsell, C.J., and
E631E638. Shutter, L.A. (2010). Prospective, randomized,
Stahel, P.F., Morganti-Kossmann, M.C., Perez, D., single-blinded comparative trial of intravenous
Redaelli, C., Gloor, B., Trentz, O., and levetiracetam versus phenytoin for seizure pro-
Kossmann, T. (2001). Intrathecal levels of phylaxis. Neurocrit. Care 12, 165172.
complement-derived soluble membrane attack Tang, H., Hua, F., Wang, J., Sayeed, I., Wang, X., Chen, Z.,
complex (sC5b-9) correlate with blood-brain Yousuf, S., Atif, F., and Stein, D.G. (2013).
barrier dysfunction in patients with traumatic Progesterone and vitamin D: Improvement
brain injury. J. Neurotrauma 18, 773781. after traumatic brain injury in middle-aged rats.
Stanislav, S.W. (1997). Cognitive effects of antipsy- Horm. Behav. 64, 527538.
chotic agents in persons with traumatic brain Taniguchi, T., Koido, Y., Aiboshi, J., Yamashita, T.,
injury. Brain Inj. 11, 335342. Suzaki, S., and Kurokawa, A. (1999). Change in
Stein, D.G. (2001). Brain damage, sex hormones and the ratio of interleukin-6 to interleukin-10 pre-
recovery:Anew role for progesterone and estro- dicts a poor outcome in patients with systemic
gen? Trends Neurosci. 24, 386391. inflammatory response syndrome. Crit. Care
Stein, D.G. (2011). Progesterone in the treatment of Med. 27, 12621264.
acute traumatic brain injury:Aclinical perspec- Tapia-Arancibia, L., Aliaga, E., Silhol, M., and
tive and update. Neuroscience 191, 101106. Arancibia, S. (2008). New insights into brain
Stein, D.G., and Cekic, M.M. (2011). Progesterone BDNF function in normal aging and Alzheimer
and vitamin D hormone as a biologic treatment disease. Brain Res. Rev. 59, 201220.
of traumatic brain injury in the aged. PM R 3, Tate, D.F., and Bigler, E.D. (2000). Fornix and
S100S110. Hippocampal atrophy in traumatic brain injury.
Stein, D.G., Wright, D.W., and Kellermann, A.L. Learn. Mem. 7, 442446.
(2008). Does progesterone have neuroprotective Taylor, C.B., Sallis, J.F., and Needle, R. (1985). The
properties? Ann. Emerg. Med. 51, 164172. relation of physical activity and exercise to men-
Stein, T.D., Alvarez, V.E., and McKee, A.C. (2014). tal health. Public Health Rep. 100, 195202.
Chronic traumatic encephalopathy:Aspectrum Temkin, N.R., Dikmen, S.S., Wilensky, A.J., Keihm, J.,
of neuropathological changes following repeti- Chabal, S., and Winn, H.R. (1990). A random-
tive brain trauma in athletes and military per- ized, double-blind study of phenytoin for the
sonnel. Alzheimers Res. Ther.6,4. prevention of post-traumatic seizures. N. Engl.
Stein, T.D., Fedynyshyn, J.P., and Kalil, R.E. (2002). J.Med. 323, 497502.
Circulating autoantibodies recognize and bind Tenovuo, O. (2005). Central acetylcholinesterase
dying neurons following injury to the brain. J. inhibitors in the treatment of chronic traumatic
Neuropathol. Exp. Neurol. 61, 11001108. brain injuryclinical experience in 111 patients.
Steptoe, A. (2007). Depression and Physical Illness Prog. Neuropsychopharmacol. Biol. Psychiatry
(Cambridge, UK:Cambridge University Press). 29,6167.
Stirling, D.P., Khodarahmi, K., Liu, J., McPhail, Theeler, B., Lucas, S., Riechers, R.G., and Ruff, R.L.
L.T., McBride, C.B., Steeves, J.D., Ramer, M.S., (2013). Post-traumatic headaches in civilians
and Tetzlaff, W. (2004). Minocycline treatment and military personnel:Acomparative, clinical
reduces delayed oligodendrocyte death, attenu- review. Headache J.Head Face Pain 53, 881900.
ates axonal dieback, and improves functional Thoene, A.I., and Glisky, E.L. (1995). Learning of
outcome after spinal cord injury. J. Neurosci. 24, nameface associations in memory impaired
21822190. patients:Acomparison of different training pro-
Strong, A.J., Fabricius, M., Boutelle, M.G., Hibbins, S.J., cedures. J. Int. Neuropsychol. Soc. 1,2938.
Hopwood, S.E., Jones, R., Parkin, M.C., and Toulmond, S., and Rothwell, N.J. (1995). Interleukin-1
Lauritzen, M. (2002). Spreading and synchro- receptor antagonist inhibits neuronal damage
nous depressions of cortical activity in acutely caused by fluid percussion injury in the rat. Brain
injured human brain. Stroke 33, 27382743. Res. 671, 261266.
450 Part IV:Homeostatic Therapies

Turken, A.U., Whitfield-Gabrieli, S., Bammer, R., (2011b). CSF Bcl-2 and cytochrome C temporal
Baldo, J., Dronkers, N.F., and Gabrieli, J.D.E. profiles in outcome prediction for adults with
(2008). Cognitive processing speed and the severe TBI. J. Cereb. Blood Flow Metab. 31,
structure of white matter pathways:Convergent 18861896.
evidence from normal variation and lesion stud- Wagner, A.K., Brayer, S.W., Hurwitz, M., Niyonkuru,
ies. NeuroImage 42, 10321044. C., Zou, H., Failla, M., Arenth, P., Manole, M.D.,
Turner-Stokes, L., Hassan, N., Pierce, K., and Clegg, F. Skidmore, E., and Thiels, E. (2013). Non-spatial
(2002). Managing depression in brain injury pre-training in the water maze as a clinically rel-
rehabilitation: The use of an integrated care evant model for evaluating learning and memory
pathway and preliminary report of response to in experimental TBI. Neurobiol. Learn. Mem.
sertraline. Clin. Rehabil. 16, 261268. 106,7186.
Tyagi, E., Agrawal, R., Ying, Z., and Gomez-Pinilla, F. Wagner, A.K., Brett, C.A., McCullough, E.H.,
(2014). TBI and sex:Crucial role of progesterone Niyonkuru, C., Loucks, T.L., Dixon, C.E., Ricker,
protecting the brain in an omega-3 deficient con- J., Arenth, P., and Berga, S.L. (2012). Persistent
dition. Exp. Neurol. 253,4151. hypogonadism influences estradiol synthesis,
Vakil, E. (2005). The effect of moderate to severe cognition and outcome in males after severe TBI.
traumatic brain injury (TBI) on different aspects Brain Inj. 26, 12261242.
of memory: A selective review. J. Clin. Exp. Wagner, A.K., Drewencki, L.L., Chen, X., Santos,
Neuropsychol. 27, 9771021. F.R., Khan, A.S., Harun, R., Torres, G.E.,
Van Reekum, R., Bayley, M., Garner, S., Burke, I.M., Michael, A.C., and Dixon, C.E. (2009). Chronic
Fawcett, S., Hart, A., and Thompson, W. (1995). methylphenidate treatment enhances striatal
N of 1 study:Amantadine for the amotivational dopamine neurotransmission after experimen-
syndrome in a patient with traumatic brain tal traumatic brain injury. J. Neurochem. 108,
injury. Brain Inj. 9,4954. 986997.
Vezzani, A., and Friedman, A. (2011). Brain inflam- Wagner, A.K., Fabio, A., Puccio, A.M., Hirschberg, R.,
mation as a biomarker in epilepsy. Biomark. Li, W., Zafonte, R.D., and Marion, D.W. (2005).
Med. 5, 607614. Gender associations with cerebrospinal fluid
Vickers, A.J., Cronin, A.M., Maschino, A.C., Lewith, glutamate and lactate/pyruvate levels after severe
G., MacPherson, H., Foster, N.E., Sherman, K.J., traumatic brain injury. Crit. Care Med. 33,
Witt, C.M., Linde, K., and Acupuncture Trialists 407413.
Collaboration (2012). Acupuncture for chronic Wagner, A.K., Miller, M.A., Scanlon, J., Ren, D.,
pain: Individual patient data meta-analysis. Kochanek, P.M., and Conley, Y.P. (2010).
Arch. Intern. Med. 172, 14441453. Adenosine A1 receptor gene variants associated
Visser, M., Deeg, D.J., and Lips, P. (2003). Low vita- with post-traumatic seizures after severe TBI.
min D and high parathyroid hormone levels Epilepsy Res. 90, 259272.
as determinants of loss of muscle strength and Wagner, A.K., Ren, D., Conley, Y.P., Ma, X., Kerr,
muscle mass (sarcopenia): The Longitudinal M.E., Zafonte, R.D., Puccio, A.M., Marion, D.W.,
Aging Study Amsterdam. J. Clin. Endocrinol. and Dixon, C.E. (2007). Sex and genetic asso-
Metab. 88, 57665772. ciations with cerebrospinal fluid dopamine and
Von Monakow, C. (1969). Diaschisis In: Brain and metabolite production after severe traumatic
Behaviour 1:Mood, States, and Mind. Pribram, brain injury. J. Neurosurg. 106, 538547.
K.H., ed. (Baltimore: Penguin).1, 2762. (origi- Wagner, J., Dusick, J.R., McArthur, D.L., Cohan, P.,
nally published in1914). Wang, C., Swerdloff, R., Boscardin, W.J., and
Wagner, A.K. (2010). TBI translational rehabilita- Kelly, D.F. (2010). Acute gonadotroph and
tion research in the 21st Century: exploring somatotroph hormonal suppression after trau-
a Rehabilomics research model. Eur. J. Phys. matic brain injury. J. Neurotrauma 27, 10071019.
Rehabil. Med. 46, 549556. Walton, R.G. (1982). Lecithin and physostigmine for
Wagner, A., McCullough, E.H., Niyonkuru, C., posttraumatic memory and cognitive deficits.
Ozawa, H., Loucks, T.L., Dobos, J.A., Brett, C.A., Psychosomatics 23, 435436.
Santarsieri, M., Dixon, C.E., Berga, S.L., et al. Wang, H., Qi, H., Wang, B., Cui, Y., Zhu, L., Rong, Z.,
(2011a). Acute serum hormone levels: charac- and Chen, H. (2008). Is acupuncture beneficial in
terization and prognosis after severe traumatic depression? A meta-analysis of 8 randomized
brain injury. J. Neurotrauma 28, 871888. controlled trials. J. Affect. Disord. 111, 125134.
Wagner, A.K., Amin, K.B., Niyonkuru, C., Postal, Wang, H.-C., Wang, P.-M., Lin, Y.-J., Kwan, A.-L.,
B.A., McCullough, E.H., Ozawa, H., Dixon, Lin, W.-C., Tsai, N.-W., Cheng, B.-C., Chang,
C.E., Bayir, H., Clark, R.S., and Kochanek, P.M. W.-N., Su, B.Y.-J., Kung, C.-T., et al. (2013).
Traumatic BrainInjury 451

Serum adhesion molecules, outcome and Wilson, M.S., and Hamm, R.J. (2002). Effects of
neuro-psychological function in acute trau- fluoxetine on the 5-HT1A receptor and recovery
matic brain injury patients. Clin. Chim. Acta Int. of cognitive function after traumatic brain injury
J.Clin. Chem. 423, 122129. in rats. Am. J.Phys. Med. Rehabil. 81, 364372.
Wang, Y., Neumann, M., Hansen, K., Hong, S.M., Wolin, K.Y., Glynn, R.J., Colditz, G.A., Lee, I.-M.,
Kim, S., Noble-Haeusslein, L.J., and Liu, J. and Kawachi, I. (2007). Long-term physical
(2011). Fluoxetine increases hippocampal neuro- activity patterns and health-related quality of life
genesis and induces epigenetic factors but does in U.S.women. Am. J.Prev. Med. 32, 490499.
not improve functional recovery after traumatic Wong, V., Cheuk, D.K., Lee, S., and Chu, V. (2011).
brain injury. J. Neurotrauma 28, 259268. Acupuncture for acute management and reha-
Warner, M.A., de la Plata, C.M., Spence, J., Wang, bilitation of traumatic brain injury. Cochrane
J.Y., Harper, C., Moore, C., Devous, M., and Database Syst. Rev. 5, CD007700.
Diaz-Arrastia, R. (2010). Assessing spatial rela- Wright, D.W., Bauer, M.E., Hoffman, S.W., and Stein,
tionships between axonal integrity, regional D.G. (2001). Serum progesterone levels correlate
brain volumes, and neuropsychological outcomes with decreased cerebral edema after traumatic
after traumatic axonal injury. J. Neurotrauma 27, brain injury in male rats. J. Neurotrauma 18,
21212130. 901909.
Webster, M.J., Herman, M.M., Kleinman, J.E., and Wright, D.W., Kellermann, A.L., Hertzberg,
Shannon Weickert, C. (2006). BDNF and trkB V.S., Clark, P.L., Frankel, M., Goldstein, F.C.,
mRNA expression in the hippocampus and tem- Salomone, J.P., Dent, L.L., Harris, O.A., and
poral cortex during the human lifespan. Gene Ander, D.S. (2007). ProTECT: A randomized
Expr. Patterns 6, 941951. clinical trial of progesterone for acute traumatic
Weiller, C., May, A., Limmroth, V., Jptner, M., brain injury. Ann. Emerg. Med. 49, 391402.
Kaube, H., Schayck, R.V., Coenen, H.H., and Wright, D.W., Yeatts, S.D., Silbergleit, R., Palesch,
Diener, H.C. (1995). Brain stem activation in Y.Y., Hertzberg, V.S., Frankel, M., Goldstein, F.C.,
spontaneous human migraine attacks. Nat. Med. Caveney, A.F., Howlett-Smith, H., Bengelink,
1, 658660. E.M., etal. (2014). Very early administration of
Werner, C., and Engelhard, K. (2007). Pathophysiology progesterone for acute traumatic brain injury. N.
of traumatic brain injury. Br. J. Anaesth. 99,49. Engl. J.Med. 371, 24572466.
White, R.J., and Reynolds, I.J. (1996). Mitochondrial Wroblewski, B.A., Joseph, A.B., and Cornblatt, R.R.
depolarization in glutamate-stimulated neu- (1996). Antidepressant pharmacotherapy and the
rons:An early signal specific to excitotoxin expo- treatment of depression in patients with severe
sure. J. Neurosci. 16, 56885697. traumatic brain injury:Acontrolled, prospective
Whyte, J., Hart, T., Schuster, K., Fleming, M., study. J. Clin. Psychiatry 57, 582587.
Polansky, M., and Coslett, H.B. (1997). Effects Wu, A., Ying, Z., and Gomez-Pinilla, F. (2004).
of methylphenidate on attentional function Dietary omega-3 fatty acids normalize BDNF
after traumatic brain injury: A randomized, levels, reduce oxidative damage, and counteract
placebo-controlled trial. Am. J. Phys. Med. learning disability after traumatic brain injury in
Rehabil. Assoc. Acad. Physiatr. 76, 440450. rats. J. Neurotrauma 21, 14571467.
Whyte, J., Hart, T., Vaccaro, M., Grieb-Neff, P., Wu, J.-N. (1996). A short history of acupuncture. J.
Risser, A., Polansky, M., and Coslett, H.B. (2004). Altern. Complement. Med. 2,1921.
Effects of methylphenidate on attention deficits Xiao, G., Wei, J., Yan, W., Wang, W., and Lu, Z.
after traumatic brain injury: A multidimen- (2008). Improved outcomes from the adminis-
sional, randomized, controlled trial. Am. J.Phys. tration of progesterone for patients with acute
Med. Rehabil. 83, 401420. severe traumatic brain injury: a randomized
Whyte, J., Vaccaro, M., Grieb-Neff, P., Hart, T., controlled trial. Crit. Care 12,R61.
Polansky, M., and Coslett, H.B. (2008). The effects Xiong, Y., Gu, Q., Peterson, P.L., Muizelaar, J.P., and
of bromocriptine on attention deficits after trau- Lee, C.P. (1997). Mitochondrial dysfunction
matic brain injury: A placebo-controlled pilot and calcium perturbation induced by traumatic
study. Am. J.Phys. Med. Rehabil. 87,8599. brain injury. J. Neurotrauma 14,2334.
Williamson, D.J., and Hargreaves, R.J. (2001). Yan, H.Q., Kline, A.E., Ma, X., Hooghe-Peters,
Neurogenic inflammation in the context of E.L., Marion, D.W., and Dixon, C.E. (2001).
migraine. Microsc. Res. Tech. 53, 167178. Tyrosine hydroxylase, but not dopamine
Wilson, B.A. (1991). Long-term prognosis of patients beta-hydroxylase, is increased in rat frontal cor-
with severe memory disorders. Neuropsychol. tex after traumatic brain injury. Neuroreport
Rehabil. 1, 117134. 12, 23232327.
452 Part IV:Homeostatic Therapies

Yan, H.Q., Ma, X., Chen, X., Li, Y., Shao, L., and Glushakova, O., Robicsek, S., et al. (2014).
Dixon, C.E. (2007). Delayed increase of tyrosine Human traumatic brain injury induces auto-
hydroxylase expression in rat nigrostriatal sys- antibody response against glial fibrillary acidic
tem after traumatic brain injury. Brain Res. 1134, protein and its breakdown products. PLoS One
171179. 9, e92698.
Young, B., Rapp, R.P., Norton, J.A., Haack, D., Tibbs, Ziebell, J.M., and Morganti-Kossmann, M.C. (2010).
P.A., and Bean, J.R. (1983). Failure of prophy- Involvement of pro- and anti-inflammatory
lactically administered phenytoin to prevent cytokines and chemokines in the pathophysiol-
late posttraumatic seizures. J. Neurosurg. 58, ogy of traumatic brain injury. Neurotherapeutics
236241. 7,2230.
Zafonte, R.D., Bagiella, E., Ansel, B.M., Novack, Zou, H., Brayer, S.W., Hurwitz, M., Niyonkuru,
T.A., Friedewald, W.T., Hesdorffer, D.C., C., Fowler, L.E., and Wagner, A.K. (2013).
Timmons, S.D., Jallo, J., Eisenberg, H., Hart, Neuroprotective, neuroplastic, and neurobe-
T., et al. (2012). Effect of citicoline on func- havioral effects of daily treatment with leve-
tional and cognitive status among patients tiracetam in experimental traumatic brain
with traumatic brain injury: Citicoline Brain injury. Neurorehabil. Neural Repair 27,
Injury Treatment Trial (COBRIT). JAMA 308, 878888.
19932000. Zuccato, C., and Cattaneo, E. (2009). Brain-derived
Zhang, Z., Zoltewicz, S., Mondello, S., Newsom, K., neurotrophic factor in neurodegenerative dis-
Yang, Z., Yang, B., Kobeissy, F., Guingab, J., eases. Nat. Rev. Neurol. 5, 311322.
25
Adenosine and Alzheimers Disease:
A Possible EpigeneticLink
D AV I D B L U M , U R S U L A S . S A N D A U , O L I V I E R B O U S I G E S ,
L U I S A V. L O P E S , VA N E S S A F L AT E N , E M I L I E FA I V R E ,
LUC BUE , A NNE-L AUR ENCE BOU T ILLIER, A ND DE T LE VBOISON

ALZHEIMERS DISEASE The definitive diagnosis of AD is based on


the observation of characteristic brain lesions,
Generalities, Symptoms, and Lesions usually found during a postmortem examina-
Alzheimers disease (AD) was first described tion: senile plaques and neurofibrillary tangles.
more than 100 years ago. Corresponding to the Neurofibrillary degeneration is due to the patho-
rise in life expectancy, its incidence has increased logical accumulation in the neuron of a naturally
dramatically, and current forecasts speak in present protein, the Tau protein, while the amy-
terms of a doubling of the number of persons loid pathology is characterized by the extracel-
affected every 20 years (Wittchen and Jacobi, lular accumulation of the amyloid-beta peptide
2005). AD is the most frequently encountered (A) that is normally present in low concentra-
form of dementia (about 70% of cases of demen- tions. Soluble oligomeric forms of amyloid pep-
tia). The earliest and most frequent manifesta- tides are thought to promote a significant part of
tions are benign memory disorders relating to their toxic effects at synapses, thereby contribut-
recent facts (Amieva et al., 2005; Ritchie et al., ing to plasticity deficits (Brouillette et al., 2012;
2001). This is followed by a slow evolution of Benilova etal., 2012; and references herein). A
symptoms, which gradually spread to organiza- peptides either are derived from the -amyloid
tional and programming disorders, such asapha- precursor protein (APP) or result from the
sia (Martin and Fedio, 1983; Murdoch et al., combined action of the two distinct proteo-
1987), apraxia (Foundas etal., 1999; Mozaz etal., lytic enzymatic activities of - and -secretase.
2006), and agnosia. In most cases, AD appears as (Checler, 1995; De Strooper et al., 2010). The
a sporadic multifactorial disease resulting from activity of -secretase is now well characterized
the interaction of different environmental, epige- as an acid protease (-site-APP cleaving enzyme
netic, and genetic factors that might facilitate its 1 [BACE1]), which releases the N-terminal frag-
onset. Various epidemiologic studies have identi- ment of the amyloid peptides. In comparison, the
fied risk factors and protective factors (Reitz C-terminal fragment is released by -secretase.
etal., 2011). Not only aging but also cardiovascu- Numerous studies suggest that presenilins
lar factors such as high blood pressure, diabetes, 1 (PS1) and 2 (PS2) are themselves carriers of the
and obesity are examples of the former (Ballard -secretase activity. Depending on the preseni-
et al., 2011; Pasinetti and Eberstein, 2008). lins involved, the -secretase activity appears
Conversely, a healthy lifestyle (physical and intel- to be associated with a multiproteoic complex
lectual activities, fish consumption, among other of high molecular weight implicating at least
factors) seems to have protective effects (Belarbi three other proteins:nicastrin, anterior pharynx
etal., 2011; La Rue, 2010; and references herein). defective 1 homolog, and presenilin enhancer 2
Recently, genome-wide association studies have homolog (De Strooper,2010).
allowed for the identification of genes associated Neurofibrillary degeneration consists of
with the disease (Lambert and Amouyel, 2011; the intraneuronal accumulation of proteina-
Bettens etal.,2013). ceous fibrils forming flame-shaped neurons
454 Part IV:Homeostatic Therapies

into paired helical filaments (for reviews see from several observations. Postmortem charac-
Buee et al., 2000; Sergeant et al., 2008). The terization of brain specimens from human AD
major antigen of paired helical filaments corre- patients reveal that the marker for glial activa-
sponds to Tau protein, which is phosphorylated. tion, S100, is elevated (Sheng et al., 1994; Van
Tau is a neuronal protein essentially located Eldik and Griffin, 1994)and that activated astro-
within the axonal compartment. Its structure cytes are found juxtaposed to amyloid plaques
makes it essential for the organization, stabi- (Wisniewski and Wegiel, 1991). Furthermore,
lization, and dynamics of microtubules (Buee there is an increase in all isoforms of the astro-
et al., 2000; Sergeant et al., 2008), but recent cyte selective marker glial fibrillary acidic pro-
data also emphasize that Tau has other impor- tein during disease progression (Kamphuis etal.,
tant neuronal functions at the dendritic and 2014). Importantly, there is a tight association
nuclear levels (Ittner et al., 2010; Morris et al., between astrogliosis and either aberrant A or
2011 for a review; Sultan etal., 2011). The physi- Tau function. Exogenous application of the A1-42
ologic and pathologic functions of Tau are also peptide to the rodent cortex causes rapid activa-
regulated by posttranslational modifications tion of astrocytes, as well as increased expression
such as phosphorylation. In AD and related of the astroglial adenosine A 2A receptor, lead-
disorders, aggregated Tau proteins are always ing to a decrease in astroglial glutamate uptake,
found hyperphosphorylated. These changes in which may underlie glutamatergic dysfunction
phosphorylation may affect a number of Tau and excitotoxicity in AD (Matos et al., 2012).
functions and facilitate Tau aggregation (Buee Importantly, the astroglial response occurs in
etal., 2000; Sergeant etal., 2008). During nor- the absence of senile plaques, indicative of a
mal aging, Tau hyperphosphorylation occurs direct A-mediated effect on astrocyte func-
in the hippocampal formation. In AD, once tion (Perez et al., 2010). Considering that A,
the hippocampus is involved, amyloid plaques acting as an acute toxin is a potent glial activa-
may be present, and the Tau pathology spreads tor, it remains possible that astrocyte activation
to the basal forebrain and several cortical areas could be an early event in disease onset, occur-
in an anatomically defined pattern (Dujardin ring even in the absence of amyloid deposition
et al., 2014) along neuronal projections defin- (Moreira etal., 2008; Nunomura etal., 2000). In
ing the Braak stages of Tau pathology (Braak line with this, glial activation has been detected
and Braak, 1991). These stages are well corre- at very early stages in the AD brain (Cagnin etal.,
lated to the severity of dementia (Duyckaerts 2001). Furthermore, in a population-based study,
et al., 1997; Grober et al., 1999). Although Tau increased gliosis has been found before the devel-
is normally considered an intracellular protein, opment of AD lesions (Wharton et al., 2009).
even in its abnormal conformational state, it is The presence of reactive astrocytes in early AD
also found in the cerebrospinal fluid (CSF) of is of great importance considering that cogni-
patients as well as in the extracellular space of tive deficits and neurodegeneration precede the
diseased brains, suggesting a spreading mecha- formation of senile plaques in multiple trans-
nism in a prion-like process (Jucker and Walker, genic mouse models of AD (Hsia et al., 1999;
2013). These findings support the instrumental Westerman etal., 2002). Not only are astrocytes
role of Tau pathology in cognitive alterations, activated by A1-42, but they may also contrib-
as evidenced by reductions in several forms of ute to disease progression and plaque formation
long-term potentiation (LTP) and long-term (Nagele etal., 2004; Nagele etal., 2003). Within
depression, two well-known manifestations of the AD brain there is evidence that A accu-
hippocampal synaptic plasticity in transgenic mulates within astrocytes (Kurt et al., 1999).
models mimicking AD-like Tau pathology Furthermore, there is a positive correlation with
(Hoover etal., 2010; Polydoro etal., 2009; Van the amount of astrocytic A42 and AD pathol-
der Jeugd etal., 2011; Burnouf etal.,2013). ogy. Single and double immunohistochemistry
to label plaques and markers for either astrocytes
InflammationandAD or neurons within human AD brain specimens
Besides A load and Tau pathology, both inflam- suggests that astrocytes, which accumulate A42,
matory processes and astrogliosis appear to be may lyse and be the source for small plaques that
integral components of AD onset and progres- form within the subpial portion of the molecular
sion (Heneka and OBanion,2007). layer of the cerebrocortex (Nagele etal.,2003).
Affirmation of the role that reactive astro- Alterations in astrocyte function are not lim-
cytes play in the pathophysiology of AD stems ited to A toxicity but are also associated with
Adenosine and Alzheimers Disease 455

Tau pathology (Belarbi etal., 2011; Schindowski CX3CR1-mediated signaling between neurons
etal., 2006). Hippocampal sclerosis is a neuropa- and microglia results in reduction of -amyloid
thology that has been described in brains from levels and deposition in APP mouse models,
patients with dementia, AD, and, most com- while microglial reaction is increased (Lee etal.,
monly, mesial temporal lobe epilepsy. The pathol- 2010b). In accordance, intrahippocampal injec-
ogy of hippocampal sclerosis is characterized by tion of lipopolysaccharide (LPS), a well-known
astrogliosis and severe loss and redistribution of promoter of inflammation, can reduce A load
neurons (Attems and Jellinger, 2006). Correlative in vivo (Herber et al., 2004; Wyss-Coray et al.,
analysis of case histories and postmortem brain 2001). In line with these findings, recent data
specimens from patients with hippocampal scle- indicate that favoring the M2 microglial pheno-
rosis reveal a strong positive correlation between type by reducing Nlrp3 inflammasomes leads
AD and the presence of both glial and neuronal to increased amyloid clearance and improved
tauopathy within the sclerotic tissue (Beach etal., memory performance in APP mice (Heneka
2003). The putative role for astrocytes in tauopa- etal., 2013). Besides resident microglia, it has also
thies has further been investigated in THY-Tau22 been shown that peripheral monocytes could
transgenic mice. As a tauopathy model, these infiltrate the brain and contribute to amyloid
mice express a double-mutated 4-repeat human deposit reduction (Simard etal., 2006). Depletion
tau, which causes hyperphosphorylated Tau of the chemokine receptor CCR2, known to be
and aggregation throughout the brain. The hip- involved in the accumulation of the mononuclear
pocampal pathology of THY-Tau22 mice includes phagocyte in the brain, results in an increase of
astrogliosis, as evidenced by increased glial fibril- A burden and memory defects in APP mouse
lary acidic protein expression, without neuronal models (El Khoury etal., 2007; Naert and Rivest,
cell loss. THY-Tau 22 mice have altered synap- 2011). However, in opposition to the aforemen-
tic function and plasticity and impairments of tioned results, some studies have concluded that
learning and memory (Van der Jeugd etal., 2011; microglia are unable to remove A deposits but
Schindowski etal., 2006). Considering the asso- instead produce cytotoxic damages (Fang et al.,
ciation of aberrant astrocyte function in both 2010; Wegiel et al., 2003). Puzzlingly, a recent
Tau and amyloid-mediated dementia and AD, study suggests that microglia ablation does not
glial cells may play a pivotal role in regulating modulate plaque formation in an APP transgenic
disease progression and behavioral outcomes. model (Grathwohl etal.,2009).
Thus restoring normal astrocyte function could Therefore, the precise role of innate immu-
be of importance for managing symptoms or dis- nity toward the amyloid side of AD remains
ease progression. unclear. At early stages, microglia and astro-
Inflammatory processes are an important cytes would help to clear amyloid but, with
hallmark of AD (for reviews see Heneka and time, chronic microglial activation and change
OBanion, 2007; Wyss-Coray, 2006). This is sub- in astrocyte functionality would contribute to
stantiated by recent genetic data indicating that the detrimental outcome. Further adding to
variants located in genes notably involved in this complexity, little is known about the rela-
innate immunity regulation such as CR1, TREM2 tionship between inflammation and the Tau
or CD33 modulate AD risk in genome-wide asso- side of AD. Previous work readily described
ciation studies (Lambert et al., 2009; Guerreiro neuroinflammatory processes in several models
et al. 2013; Jonsson et al., 2013; Naj et al., 2011; of AD-like Tau pathology (Bellucci etal., 2004;
Bradshaw et al., 2013). Further, a recent study Lourent et al., 2011; Sasaki et al., 2008; Zilka
points out that both acute and chronic systemic et al., 2009). It has been stressed in particular
inflammation, characterized with increases in that microglial activation would even precede
serum tumor necrosis factor-alpha (TNF), is tangles formation (Yoshiyama et al., 2007).
associated with an increase in cognitive decline Interestingly, microglial activation would be
in AD (Holmes etal., 2009). Microglial cells seem detrimental toward Tau pathology. Indeed, LPS
to play an important role in AD-related central administration in rTg4510 mice exacerbated
inflammatory processes. Whether microglial hippocampal and cortical Tau phosphoryla-
activation is protective or deleterious is still a tion (Lee etal., 2010a). This is in line with other
matter of debate in AD research and has been dis- data showing that LPS significantly induced
cussed elsewhere (e.g., see Wyss-Coray, 2006, for Tau hyperphosphorylation through activation
a review). Some data indicate that microglia may of cyclin-dependent kinase 5 in the 3xTg trans-
promote A clearance. For instance, removing genic model, without affecting APP processing
456 Part IV:Homeostatic Therapies

(Kitazawa et al., 2005). Involvement of micro- in AD. In contrast, the role of glia and of home-
glia in processes underlying Tau pathology has ostatic control mechanisms in the pathogen-
been further emphasized by demonstrating esis of AD have remained understudied. Thus
that removal of the microglial CX3CR1 recep- therapies aimed at reconstructing homeostatic
tor leads to enhanced Tau pathology (Bhaskar network function are of interest to improve
etal., 2010). These observations fit well with the cognitive function in AD. Importantly, cogni-
ability of proinflammatory mediators, known tive dysfunction in AD occurs prior to neu-
to be released by microglial cells, such as IL1 rodegenerative changes in several models of
or TNF, to promote Tau phosphorylation and AD. This implies that neuroprotective strate-
even its neuritic aggregation (Gorlovoy et al., gies might not be sufficient to improve cogni-
2009; Li et al., 2003). Together, these observa- tive function in AD and that strategies aimed
tions fit with recent data demonstrating a good at reinstating normal network activity deserve
correlation between innate immunity and investigation. If cognitive impairment in AD
phosphorylated-Tau levels in the brain of AD is not primarily due to the loss of neurons but
patients (Zotova etal.,2013). can be linked to a deficiency in homeostatic
Overall, the aforementioned data stress that control mechanisms, then cognitive decline in
astrogliosis and neuroinflammatory events, AD might be a therapeutically reversible pro-
especially those mediated by microglial cells, cess. In support of this hypothesis the antie-
may have an instrumental role in AD, while pileptic drug levetiracetam effectively reduced
their respective contribution to the amyloid and abnormal epileptiform activity in hAPP mice.
Tau sides remain unclear so far. It is also indi- Importantly, chronic treatment with this drug
cated, however, that modulation of brain glial also reversed hippocampal remodeling, behav-
cells may affect mechanisms underlying AD ioral abnormalities, synaptic dysfunction,
pathogenesis. and deficits in learning and memory in those
animals (Sanchez et al., 2012). These findings
suggest that aberrant network activity might
Lesions, Seizures, and
causally be linked to synaptic and cognitive
Memory LossinAD
deficitsinAD.
As already outlined, inflammatory processes
appear to be crucial in AD pathophysiology
(Heneka and OBanion, 2007). Several lines of EPIGENETIC CHANGES
evidence now indicate that astrocyte and micro- AND MEMORY LOSS
glial activation could be an early event in the I N A D : F R O M M E T H Y L AT I O N
disease, occurring even in the absence of A T O A C E T Y L AT I O N
deposition (Heneka and OBanion, 2007). In Epigenetic mechanisms are able to influence gene
support of this notion, AD patients have glial expression without altering the DNA sequence,
activation, which is evident at very early stages and the coordinated action of epigenetic programs
of disease progression (Cagnin et al., 2001) and on thousands of genes leads to diverse phenotypes.
appears to contribute to the development of amy- In dividing cells, such changes are heritable, but
loid plaques (Nagele et al., 2004). Importantly, in postmitotic neurons, they participate in the
AD is now considered as a complex syndrome dynamic regulatory mechanisms maintaining
comprised not only of cognitive impairment but their functional status. As the epigenetic profile
also of comorbid conditions that include seizures of young identical twins are essentially indistin-
and changes in sleep homeostasis. Those comor- guishable, older ones show substantial differ-
bid changes cannot readily be explained by amy- ences in their epigenetic marks (Fraga etal., 2005;
loid or Tau deposits, or by neuronal cell loss, and Martin, 2005; Mastroeni etal., 2011). Remarkably,
indicate that a more global disruption of network the onset of AD in identical twins can differ by
homeostasis might contribute to the complex more than 20 years (Cook et al., 1981; Nee and
symptomatologyofAD. Lippa, 1999). Thus epigenetic mechanisms are
AD research and therapy development have thought to mediate the interaction between genetic
traditionally been dominated by the perception and environmental factors (e.g., diet, heavy metal
that cognitive impairment in AD is due to neu- exposure, stress), and they could represent a link
ronal cell loss. Therefore, therapy development between life events, risk factors, and pathophysi-
has focused largely on neuroprotective strate- ological processes underlying the emergence of
gies, which fail to reverse cognitive impairment neurodegenerative disorders suchasAD.
Adenosine and Alzheimers Disease 457

Epigenetic Marks ofthe Chromatin undermethylated and repressed genes undera-


Epigenetic modifications are found either on the cetylated (Cervoni and Szyf, 2001), thereby main-
DNA or the histone proteins. DNA methylation taining cellular homeostasis.
is involved in key cellular processes, includ-
ing X-chromosome inactivation, imprinting, DNA Methylation and Susceptibility
and transcriptional silencing of specific genes. GenesinAD
DNA methylation is a covalent biochemical Basically, DNA methylation perturbations have
modification occurring predominantly on the been shown to affect some susceptibility genes
cytosines (5mC) of so-called CpG islands, cata- involved in AD and hence can drive changes on
lyzed by the family of DNA methyltransferases the pathophysiology of the disease. For example,
(DNMT1, DNMT3A and DNMT3B; Goll and the high CpG content (72%) found in the APP
Bestor, 2005). So far, no direct DNA demethyl- promoter makes it a valuable target for control
ase has been identified, but it was recently found by methylation (Yoshikai et al., 1990). This was
that ten-eleven translocation enzymes (TET1, further confirmed as complete demethylation of
TET2, and TET3) could convert aberrant DNA the APP gene was reported in an AD postmortem
methylation of cytosines (5mC) into hydroxym- cortical sample but not in a normal control or a
ethylation (5hmC), thereby providing a means Picks disease patient (West etal., 1995). Specific
for active demethylation (Tahiliani etal., 2009). demethylation of the APP promoter was also
Such a mechanism has also been found in neu- reported to occur with age, a major risk factor for
rons (Guo et al., 2011). Methylation of CpG AD, in the parietal cortex of 10 neurologically nor-
island promoters correlates with transcrip- mal individuals (Tohgi etal., 1999). Several stud-
tional silencing, either by a direct interference ies showed a general decrease in 5mC amounts
of transcription-factor binding or through the with age (Cooney, 1993; Wilson and Jones, 1983;
recruitment of repressive methyl-binding pro- Wilson etal., 1987). Regarding the mechanisms,
teins such as MeCP2 (Bogdanovic and Veenstra, immunoreactivity for DNMT1 and methylation
2009). Of note, Gadd45b, involved in DNA corepressor complex components were mark-
repairlike mechanisms, has been proposed to edly decreased together with 5mC in some AD
drive activity-dependent demethylation at spe- cases (Mastroeni et al., 2010). In addition, in a
cific promoters (Bdnf and Fgf) in neurons (Ma primate model of AD, DNMT1 was decreased in
etal.,2009). the cortex and associated with hypomethylation
In contrast to DNA, histones can undergo of the APP promoter and increased APP mRNA
multiple posttranslational modifications such expression (Wu et al., 2008). The presenilin-1
as methylation, acetylation, phosphorylation, (PSEN1) promoter was also hypomethylated
sumoylation, ubiquitination, ADP-ribosylation, in late-onset AD brains compared with con-
and others, among which acetylation is the most trols (Wang et al., 2008). These results failed to
well studied. Histone acetylation is tightly regu- be reproduced in another study investigating
lated by opposing enzymes, the histone acetyl- the methylation state of selected loci (including
transferases (HATs) add an acetyl group on lysine microtubule-associated protein Tau MAPT, APP,
residues in the N-terminal histone tails, while and PSEN1) in AD disease cases when compared
the histone deacetylases (HDACs) hydrolyze with controls, which failed to detect any signifi-
the acetyl group. It is believed that such modi- cant modifications, despite a high preservation
fications remove positive charges from histone of CpG methylation of gene promoters with
proteins and lower their interactions with the postmortem delay. However, small changes in
negatively charged DNA structure, thereby con- the methylation of DNA promoters in vulner-
ferring a more relaxed conformation to the chro- able cells might have been missed using total
matin. This has an impact on transcription factor homogenates of frontal cortex and hippocampus
and chromatin regulator accessibility, ultimately (Barrachina and Ferrer,2009).
modulating gene transcription (Kouzarides, By contrast, higher A concentration results
2007; Li etal., 2007). Recent data suggest that the in promoter hypermethylation, particularly that
co-occurrence of histone marks may act as allos- of the neprilysin gene, a major A-degrading
teric regulators of chromatin complexes (Rando, enzyme in the brain, thereby leading to decreased
2012). It is noteworthy that histone- and DNA neprilysin levels in the hippocampus of AD
methylation modifications work tightly together patients (Chen etal., 2009; Gunzburg etal., 2007;
in cells, maintaining, for instance, active genes Iwata et al., 2000; Yasojima et al., 2001). This
458 Part IV:Homeostatic Therapies

suggests that A-induced methylation could etal., 2004)and of Hcy in plasma (Clarke etal.,
exert positive feedback on its own production 1998; Smith, 2008). Deficits in folates and ele-
through such methylation processes. vated Hcy levels in APP mutant mice impaired
In summary, some genes involved in AD DNA repair in hippocampal neurons, sensitiz-
(APP, PSEN1) are found hypomethylated in ing them to oxidative damage induced by A
patients and animal models of AD. Interestingly, (Kruman etal., 2002). The resulting elevated Hcy
in AD patients, global DNA hypomethylation levels and corresponding decrease in SAM in
was observed in the entorhinal cortex, one of AD (Tchantchou etal., 2006)may decrease APP
the first structures to be affected by the disease promoter methylation and increase its expres-
(Mastroeni et al., 2010). Global DNA hypo- sion, thereby leading to increased A load. In
methylation was also found in an AD monozy- vitro, expression of the BACE and PSEN 1 genes
gotic twin compared with his normal sibling is enhanced after folate deprivationinduced
(Mastroeni et al., 2009). Of note, such global hypomethylation and restored to normal when
hypomethylation is also involved in the deregu- folate deprivation is accompanied by SAM sup-
lation of other genes, including those contrib- plementation (Fuso et al., 2005). In vivo, expo-
uting to neurodegeneration (e.g., aberrant cell sure of APP-overexpressing transgenic mice
cycle, apoptosis, and inflammatory processes) in to a folate/B12/B6deficient diet is associated
AD (Mastroeni etal.,2011). with enhanced SAH relative to SAM, as well as
PS1 hypomethylation leading to PS1 upregu-
DNA Methylation and lation but also BACE upregulation, enhanced
theMethionine CycleinAD A deposition, and an accelerated appearance
Both homocysteine (Hcy) and folate are criti- of intraneuronal A in line with the develop-
cal components of a series of biosynthetic path- ment of cognitive deficits (although the latter
ways essential for DNA methylation. S-adenosyl was quite modest; Fuso et al., 2008; Fuso et al.,
methionine (SAM), generated by methionine, is 2011). Furthermore, inhibiting methylation via
a methyl donor for methylation reactions. The vitamin B deficiency is responsible for glycogen
resulting product, S-adenosylhomocysteine synthase kinase 3 beta (GSK3; one of the most
(SAH), can be further processed in Hcy and important protein kinases regulating Tau phos-
adenosine (ADO) by SAH hydrolase. Then rem- phorylation) upregulation, promoting GSK3beta
ethylation of Hcy to methionine (the methionine protein increase and leading to abnormal
cycle) predominates over the catabolic degra- hyperphosphorylated Tau (Nicolia et al., 2010).
dation of Hcy (Finkelstein, 2000). Folate and SAM treatment reversed PS1 hypomethylation
vitamin B12 are essential cofactors required to and consequently inhibited the upregulation
regenerate methionine from Hcy. Chronic eleva- of vitamin B deficiencyinduced PSEN1 and
tion in Hcy levels results in parallel increases in also BACE1 expression. Those changes reduced
SAH, whose intracellular accumulation can be amyloid production and plaque spreading and
pathologic as it is a potent product inhibitor of increased Tau phosphorylation and spatial
DNMTs (Hoffman et al., 1980). Thus sustained memory in TgCRND8 mice (expressing human
hydrolysis of SAH to Hcy and ADO is essential to APP695 with the Swedish [KM670/671/NL] and
maintain normal methylation of DNA, as well as Indiana [V717F] mutations) and wild-type mice
a proper methionine regeneration. (Fuso etal., 2011; Fuso etal., 2012). Several stud-
Vitamin B deficiency in humans and in ies have tested the benefits of high-dose supple-
animal models results by itself in global hypo- ments of folate, vitamin B12, and vitamin B6 in
methylation. In AD patients the levels of Hcy humans (Aisen etal., 2003; Aisen etal., 2008)and
are elevated, and the pathology is associated demonstrated that vitamin B treatment reduced
with folate and vitamin B12 deficiency (Morris, the elevated Hcy levels in AD patients, without
2003; Seshadri et al., 2002). Folate is signifi- any significant benefits in cognitive function,
cantly decreased in AD CSF (Serot etal., 2001), however. It was further reported that a cocktail of
as well as SAM and one of its synthesizing folate, vitamin B6, -tocopherol, SAM, N-acetyl
enzymes, methionine S-adenosyltransferase, in cysteine, and acetyl-L-carnitine in early-stage
the CSF and brain of AD patients (Bottiglieri AD patients led to significant improvements in
et al., 1990; Morrison et al., 1996). DNA meth- all scoring systems (dementia rating scale, neu-
ylation inhibition has been observed together ropsychiatric inventory, and activities of daily
with increased SAH levels in the brain (Kennedy living) for all patients (Chan etal.,2008).
Adenosine and Alzheimers Disease 459

Histone ModificationsinAD broad HDACi sodium butyrate of p25/Cdk5,


Histone acetylation is the major histone modifi- which present neurodegeneration, neurofibril-
cation under the scope of Alzheimers research. lary pathology (Cruz etal., 2003), and intracel-
Other histone modifications, such as methyla- lular A accumulation (Cruz et al., 2006), not
tion, phosphorylation, and ADP-ribosylation only increased H3 and H4 acetylation levels in
have been linked to memory functions but, so mice but also induced sprouting of dendrites,
far, not to AD (Grff etal., 2011; Fischer 2014). increased the number of synapses, and resulted
The exact outcome of the role and the fate of in the reestablishment of learning abilities, as
acetylation in AD is not clear, as many conflict- well as access to long-term memories that had
ing data have been reported at all levels stud- been ablated by prior hyperactivation of p25/
ied (cellular, animal, human pathology), likely Cdk5 (Fischer et al., 2007). Administration of
due to the great heterogeneity of models and the pan-HDACi phenylbutyrate (PBA), a model
approaches that have been used. Unfortunately, of amyloid pathology, in the tg2576 mice was
even if acetyltransferase activities seem to be able to reverse spatial memory loss (Ricobaraza
relatively preserved with postmortem and stor- et al., 2009) and fear conditioningrelated
age duration, acetylated histones are mostly memory (Ricobaraza etal., 2011). Interestingly,
unstable in postmortem tissues and only a PBA also successfully impacted AD physi-
few results are available in the human brain opathology, as it reduced the hyperphosphoryl-
(Barrachina etal., 2012; Monoranu etal., 2011). ated form of Tau via an increase of the GSK3
In addition, brain tissues reflect a mixture of inactive form (Ricobaraza et al., 2009) and
neuronal and glial populations, the ratio of decreased amyloid beta pathology as well as
which is exacerbated by the pathology, so it is inflammation (Ricobaraza etal., 2011). Chronic
difficult to conclude histone acetylation levels PBA treatment also restored the loss of den-
in neurons through global analyses. A recent dritic spines and synaptic plasticity in the
study reported that HDAC2 levels were signifi- hippocampus in 6- and 16-month-old tg2576
cantly elevated in the hippocampal area CA1 mice (Ricobaraza et al., 2011; Ricobaraza
and the entorhinal cortex in AD patients, as et al., 2012)., Importantly, such treatments
early as in Braak and Braak stage I/II, suggest- were efficient against memory dysfunctions
ing HDAC2 increase could be an early event in even in a very severe stage, as described in the
the progression of AD (Grff et al., 2012). As APPPS1-21 mouse strain following sodium
HDAC2 was recently described to negatively butyrate treatment (Govindarajan etal., 2011).
regulate memory and synaptic plasticity in the Recent reports performed with more selective
healthy rodent brain (Guan etal., 2009; Hanson inhibitors also showed an interesting thera-
et al., 2013), elevated levels of HDAC2 in AD peutic potential obtained with an HDAC1
may also accompany the cognitive decline of inhibitor MS-275 (Entinostat) in APPPS1-21
the human neurodegenerating brain. This result mice (behavioral impairment improvement
nicely fits the current literature promoting the and significant reduction of amyloid plaque
use of HDAC inhibitors in memory-related dis- deposition in the hippocampus and corti-
eases (Fischer etal., 2010; Grff and Tsai, 2013b), cal regions; Zhang and Schluesener 2013) or a
because histone acetylation has been associated mercaptoacetamide-based class II inhibitor in
with facilitation of learning and memory (Grff 3xTG AD mice (decrease of A40, A42, and
and Tsai, 2013a). Indeed, many studies have Thr181 phospho-Tau levels and spatial memory
now tested the potential of HDAC inhibitors in performance improvement; Sung etal., 2013). It
AD models for their ability to restore memory is noteworthy that HDACi might affect not only
functions. Injection of trichostatin Ain the hip- histone acetylation but other cellular proteins
pocampus of APP/PS1 mice increased the levels as well. For example, Tau was shown to be acet-
of acetyl H4 and contextual freezing perfor- ylated by the HAT p300 and deacetylated by the
mance upto wild-type values and restored hip- classIII HDAC SiRT1 (Min etal., 2010). ATau
pocampus synaptic dysfunction (Francis etal., intrinsic acetyltransferase activity was also
2009). Administration of various pan-HDACs reported (Cohen etal., 2013). The impact of Tau
inhibitors (HDACi; sodium valproate, sodium acetylation in the pathology seems to lead to an
butyrate, suberanilohydroxamic acid) was able aggravation of the tauopathy (Min etal., 2010;
to reinstate associative memory in APP/PS19 Cohen et al., 2011; Irwin et al., 2012), but this
mice (Kilgore etal., 2010). Treatment with the has been recently debated (Cook et al., 2014).
460 Part IV:Homeostatic Therapies

Ultimately HDACi treatment might lower the in histone acetylation. Deletion of the genes
phosphorylation status of Tau as reported in encoding presenilins PSEN1 and PSEN2 lead to
response to phenylbutyrate (Ricobaraza et al., a downregulation of genes regulated through the
2009)or mercaptoacetamide (Sung etal., 2013). CREB/CBP complex, which are associated with
However, such links remain to be further tested. decreased LTP, long-term memory deficits, and
In contrast to these in vivo beneficial effects hyperphosphorylated Tau (Saura et al., 2004).
of increasing acetylation with HDACis, early evi- In cellular systems, overexpression of wild-type
dence obtained in cellular models rather linked PSEN1, but not PSEN1 variants containing
AD-related genes with upregulation of histone mutations associated with familial AD, stimu-
acetylation. For instance, the APP C-terminal lated the transcriptional activity of CBP and
peptide (AICD), a product of APP cleavage by p300 (Francis et al., 2007; Francis et al., 2006).
the -secretase, was shown to interact with the Moreover, CRE/CBP-dependent gene expression
acetyltransferase TIP60 through the nuclear was altered in PSEN1 and APP mice (Soreghan
adaptor Fe65, and the resulting complex was able et al., 2005). Interestingly, an in vivo study
to enhance gene transcription (Cao and Sudhof, showed that increasing CBP levels with admin-
2001). The AICD fragment induced histone H3 istration of viral particles in triple transgenic
and H4 hyperacetylation and the expression AD mouse hippocampus restored learning and
of cytotoxic genes, an effect further enhanced memory deficits, likely through BDNF upregula-
by high-dose HDAC inhibitor administration tion (Caccamo etal., 2010). Therefore, CBP level
(Kim et al., 2004). Another study reported that preservation seems to be important in patholo-
familial-AD mutations on PS genes inhibited the gies such as AD. In addition, a recent study in
production of an intracellular peptide N-Cad/ humans established that AD patients presented
CTF2 originally cleaved through WT PS1, increased nuclear translocation of an endog-
thereby preventing the degradation of the acetyl- enous p300/CBP inhibitor (EP300-interacting
transferase CBP and resulting in upregulated inhibitor of differentiation 1 [EID1]) in their hip-
CBP/CREB-mediated transcription (Marambaud pocampal neurons (Liu etal., 2012). EID1 over-
et al., 2003). However, whether these mecha- expression in mouse brain reduces hippocampal
nisms exist in vivo remains unknown. Of note, LTP and impairs spatial memory as well as his-
epidemiologic studies show that AD risk is sig- tone H3 acetylation (Liu etal., 2012). Altogether,
nificantly reduced in a population that consumes this series of data supports strategies that could
large quantities of curcumin (Chandra et al., target CBP HAT activity as an interesting thera-
1998; Chandra etal., 2001), a molecule that has peutic pathway to target treatment development
been shown to display acetyltransferase inhibi- in AD (Schneider etal., 2013; Valor etal., 2013).
tor on CBP/P300 proteins (Morimoto et al., Such drugs are currently being developed, and a
2008) and to interfere with the formation of HAT activator treatment in wild-type mice was
amyloid plaques in AD rodent models (Ishrat recently shown to enhance maturation of newly
et al., 2009; Lim et al., 2001; Ono et al., 2004; generated neurons in the adult and sustain spa-
Yang et al., 2005). Overexpression of SIRT1, tial long-term memory (Chatterjee et al., 2013),
which mainly deacetylates nonhistone proteins, but it remains to be tested in AD models.
rescued neurons against neurotoxicity in p25/ In conclusion, multiple epigenetic mecha-
Cdk5 mice (Kim et al., 2007), as well as in the nisms are dysregulated in AD, and it is difficult to
APPswe,PSENdE9 AD mouse strain (Donmez clearly link specific epigenetic changes to patho-
etal.,2010). logical effects, as many molecular players and
In contrast, modulation of the APP sig- mechanisms are involved. However, the thera-
nalling pathway (with a specific APP-targeted peutic modulation of histone acetylation stands
antibody) in primary neurons induced the dea- out as a very promising strategy with proven effi-
cetylation of H3 and H4, together with caspase cacy on plasticity and memory-related symptoms
6-dependent degradation of the acetyltrans- and to a certain degree also on the pathophysiol-
ferases CBP and p300 (Rouaux et al., 2003). It ogy in animal models (Grff etal., 2011; Fischer
is noteworthy that caspase-6 activity has been 2014). Manipulating the global DNA hypometh-
observed in neurofibrillary tangles in the brain ylation observed in AD brains, especially as it
of AD patients (Albrecht etal., 2007), suggesting is also possible with specific diets in the elderly,
that such loss of acetyltransferases could occur may be of great interest in AD pathology as well
in diseased neurons and produce a net decrease (Narayan and Dragunow,2010).
Adenosine and Alzheimers Disease 461

ADENOSINE AS by interfering with other receptors (Ribeiro


A H O M E O S TAT I C and Sebastiao, 2010). Adenosine receptors are
R E G U L AT O R I N A D G-protein coupled receptors that mostly regu-
late, in opposite directions, the second messen-
Adenosine:Origin and Receptors ger cAMP. While A1Rs are coupled to inhibitory
ADO is an endogenous neuromodulator control- Gi/o proteins, thus decreasing cAMP levels, A2A
ling brain function via activation of high-affinity receptors are coupled to excitatory Gs proteins,
A1 or A2A, low-affinity A2B, or low-abundance increasing cAMP levels. Activation of these
A3 adenosine receptors that feed into a multi- receptors also modulates Ca 2+ channels and
tude of different neuronal and glial downstream the phospholipase C pathway. Through these
pathways (Blum etal., 2003; Boison etal., 2009; actions and by modulating both the release and
Sebastiao and Ribeiro, 2000, 2009). Synaptic lev- the uptake of different neurotransmitters, the
els of ADO are regulated by an astrocyte-based balance between adenosine A1 and A2A recep-
ADO cycle and metabolic clearance through tors activation allows the fine-tuning of synaptic
phosphorylation into adenosine monophosphate transmission and plasticity in the hippocampus
(AMP) via adenosine kinase (ADK; Boison, (Sebastiao and Ribeiro, 2009). Upon aging, as
2008). Importantly, the ADK/ADO system is an well as under chronic noxious brain conditions,
upstream regulatory component of complex neu- increased ADO levels are associated with an
ronal interactions. Changes in this system affect increase in A2A receptor density and effects and
brain function on the network level by simulta- a decrease in A1 receptors (Cunha et al., 2001;
neous modulation of several downstream path- Lopes et al., 1999; Rebola et al., 2003). Thus, by
ways (Boison, 2009; Boison etal., 2009; Sebastiao blocking adenosine receptors, caffeine emerges
and Ribeiro, 2000). Evidence that ADO dysregu- as a potential drug to modulate brain function
lation is associated with AD pathology is evident and cognition (de Mendonca and Cunha,2010).
from the observed decrease in hippocampal A1 The widespread consumption of caffeine
receptors in AD patients (Stone etal., 2009). The underlies the importance of studying the conse-
postulated deficiency in ADO signaling in AD is quences of its chronic intake to aging-associated
consistent with cognitive impairment, seizures, cognitive decline, particularly in pathologi-
and sleep disorders, which are all characteristic cal conditions. Caffeine consumption increases
comorbiditiesofAD. alertness and improves cognition in physiologi-
cal conditions (Fisone etal., 2004; Haskell etal.,
Caffeine asa Cognitive 2005; Sawyer etal., 1982; Smit and Rogers, 2000).
NormalizerinAD Caffeine also improves attention and information
Caffeine is one of the most consumed psychoac- processing (Lorist and Tops, 2003). In rodents,
tive drugs. At high millimolar concentrations, evidence from the past few years supports the
irrelevant for normal consumption, caffeine cognitive-enhancing properties of caffeine in a
can act at the level of ryanodine receptors and variety of behavioral tasks that evaluate learning
cyclic nucleotide phosphodiesterases, but it is and memory (Takahashi et al., 2008; Marques
now well established that under normal physi- etal., 2011). More recently, and in line with such
ological conditions, the effects exerted in the experimental evaluations, a recent study points
brain by caffeine depend on its ability to block out that caffeine improves long-term memory in
adenosine A1 and A 2A receptors (Fredholm humans (Borota etal.,2014).
et al., 1999; Fredholm et al., 2005). In humans, However, only recently has the relationship
caffeine is largely demethylated to its dimethyl between the consumption of caffeine and the
metabolic intermediates, with over 80% of orally pathophysiology of many neurodegenerative dis-
delivered caffeine metabolized to paraxanthine eases been firmly established (Rahman, 2009).
(1,7-dimethyl-xanthine), and about 16% is con- Even if some controversy may still exist (Eskelinen
verted to theophylline (3,7-dimethylxanthine) and Kivipelto, 2010), caffeine consumption has
and theobromine (1,3-dimethylxanthine), both beneficial effects not only on aging, reducing the
potent antagonists of adenosine receptors. associated cognitive decline in healthy subjects
Adenosine receptors have a crucial neu- (Johnson-Kozlow etal., 2002; Ritchie etal., 2007;
romodulatory role and regulate both synaptic van Gelder etal., 2007), but also in AD patients.
transmission and plasticity (Linden, 2001)either Indeed, different epidemiological reports high-
by directly modulating synaptic responses or light the inverse correlation between caffeine
462 Part IV:Homeostatic Therapies

consumption and the incidence or severity of to 9.5 months of age with caffeine (300mg/L by
AD. In the Canadian Study of Health and Aging, drinking water corresponding to 500mg/d in
daily coffee drinking decreased the risk of AD humans) were cognitively improved in several
by 31% during a 5-year follow-up (Lindsay etal., behavioral tasks that evaluated working and
2002). In line with this, another study from the spatial memories and exhibited reduction of
same population reports that coffee drinkers at hippocampal A1-40 and A1-42 (Arendash et al.,
midlife had a lower risk of dementia and AD later 2006). Importantly, a similar treatment of APPsw
in life compared with those who drank little or mice at late pathological stages (1819 months)
no little coffee. The lowest risk (65% decreased) for 4 to 5 weeks reversed memory deficits and
was found in people who drank three to five cups reduced amyloid deposition as well as soluble
per day (Eskelinen etal., 2009). Finally, a retro- A levels in both entorhinal cortex and hip-
spective study has shown an inverse correlation pocampus (Arendash et al., 2009). Such benefi-
between coffee intake and the occurrence of cial effects of caffeine against A production has
AD later on in life. Indeed, while age-matched recently been confirmed by another group in an
controls had an average daily caffeine intake experimental model of sporadic AD based on
of 198.7 135.7 mg during the corresponding feeding rabbits a cholesterol-enriched diet that
20years of their lifetimes, patients with AD had elevates both A levels and Tau phosphoryla-
a lower average daily caffeine intake of 73.9 97.9 tion in the brain (Prasanthi etal., 2010). In this
mg during the 20years that preceded diagnosis study, rabbits fed a cholesterol-enriched diet were
of AD (Maia and de Mendonca, 2002). Other ret- treated with low doses of caffeine (0.5 to 30 mg/d)
rospective data proposed that high plasma caf- through drinking water, corresponding to a max-
feine levels were associated with a reduced risk imal 60mg/d consumption in humans. In this
of dementia or a delayed onset in patients with paradigm, caffeine significantly decreased A
mild cognitive impairment (Cao etal., 2012). It is production in accordance with Arendash etal.s
also noteworthy that the recent Honolulu-Asian results (Prasanthi et al., 2010). Interestingly,
Aging Study did not find a significant associa- reduced production of A140 and A142 was also
tion between caffeine intake and dementia risk observed in a neuroblastoma model overexpress-
(Gelber etal., 2011). However, the authors inter- ing mutated APP following treatment with con-
estingly reported that, at autopsy, patients in the centrations of caffeine below 10 M (Arendash
highest quartile of caffeine intake (>277.5 mg/d) etal., 2006), further supporting the idea that caf-
were less likely to have any of the neuropathol- feine impacts on mechanisms underlying amy-
gical lesions, such as AD-related lesions, micro- loid peptide production. In accordance, chronic
vascular ischemic lesions, cortical Lewy bodies, caffeine treatment of APPsw mice has been asso-
hippocampal sclerosis, or generalized atrophy. ciated with decreased PS1 and BACE1 protein
Therefore, available epidemiological data support expression as well as increased insulin-degrading
that caffeine consumption is able to slow down enzyme levels, the latter presumably contribut-
cognitive decline in the elderly and reduces the ing to enhanced A degradation (Arendash etal.,
risk of developing AD. Of note, while this looks 2006; Prasanthi et al., 2010). Effects of caffeine
also to be the case in Parkinsons disease (Ross on BACE1 expression could relate to its ability
etal., 2001), caffeine has been recently suggested to reduce c-Raf1 activity, possibly through PKA
to exhibit detrimental effects in Huntingtons activation (Arendash et al., 2009). In addition,
disease (Simonin et al., 2013), suggesting that caffeine could reduce GSK3 expression and/or
caffeine is not protective in all neurodegenerative activity and thereby influence A production
conditions and that its effects depends on under- (Arendash et al., 2009). However, a direct effect
lying instrumental mechanisms. of caffeine on -secretase activity remains elu-
sive, and mechanisms linking caffeine and A
Caffeine, Adenosine Receptor production/clearance deserve further evaluation.
Targeting, and AD:From Lesions Finally, it is noteworthy that although the ben-
toInflammation eficial effects of coffee on cognitive decline and
Caffeine mitigates cognitive decline induced by decreased AD risk in humans has been mostly
A and reduces amyloid burden in transgenic ascribed to caffeine, other coffee constituents may
mice overexpressing mutated APP (APPsw) play an important role. Indeed, two recent stud-
in preventive, but also therapeutic paradigms. ies support that noncaffeine components of cof-
Indeed, APPsw mice chronically treated from 4 fee display neuroprotective effects in drosophila
Adenosine and Alzheimers Disease 463

and C.elegans amyloid models though activation as LTP impairments (Lopes et al., unpublished
of the Nrf2 detoxification pathway (Dostal etal., data). Interestingly, other detrimental condi-
2010; Trinh etal.,2010). tions associated with cognitive impairment, such
Doses used in experimental studies describ- as hypoxia, diabetes, or epilepsy, share similar
ing beneficial effects of caffeine achieve plasma A 2A receptor overactivation (Lopes et al., 1999;
concentrations of caffeine and metabolites Batalha etal., 2013; Lopes etal., 2011, for review).
below 10M (Georgiev etal., 1993). Taking into Recently, we demonstrated decreased adult hip-
account the brain-to-plasma ratio (Kaplan etal., pocampal LTP and cognitive/memory impair-
1989), and even if the involvement of other tar- ment in a chronic stress induced aging-like
gets cannot be excluded (Fredholm et al., 1999; model, generated by maternal separation during
Guerreiro etal., 2008), such levels are essentially the early postnatal period, in association with
compatible with adenosine receptor inhibition increased hippocampal A 2A receptor expression
(Fredholm et al., 1999; Muller and Jacobson, (Bathala etal., 2013). Strikingly, we observed, in
2011). Therefore, beneficial effects of caffeine adults, a normalization of synaptic and cognitive
could then be mostly ascribed to its effects on dysfunctions following A2A receptor blockade
adenosine receptors. Previous studies indicate with the selective antagonist KW6002, support-
that A1 receptor activation increases soluble APP ing an instrumental role of A2A receptor dys-
production, a nonamyloidogenic form of APP regulation in the genesis of synaptic dysfunction
(Angulo etal., 2003). Thus A1 receptor blockade underlying cognitive impairment within the
by caffeine could reduce the nonamyloidogenic context ofaging.
pathway, an effect considered deleterious in the As A2A receptors are dysregulated in AD
AD context. However, the involvement of the A1 (Albasanz et al., 2008), and given the impact
receptor blockade by caffeine in the modulation of caffeine toward both cognitive and patho-
of amyloid production and toxicity remains to be logical processes, the blockade of A 2A receptors
thoroughly evaluated, as does the potential role appears to be a rational strategy to mitigate sev-
of adaptive changes of A1 receptors promoted by eral detrimental components in AD. Besides
chronic caffeine consumption (Jacobson et al., A-production, convergent data indicate that caf-
1996; Johansson et al., 1997). Indeed, adaptive feine protects against the synapto-neurotoxicity
increases in A1 receptor expression are expected induced by A through blockade of A 2A recep-
to promote the neuroprotective potential of tors. Caffeine indeed protected against the death
endogenous ADO (Cunha, 2008) and have been of rat primary hippocampal neurons induced
suggested to be involved in protection afforded by an adenovirus carrying a mutated APP gene
by caffeine in ischemia or traumatic brain injury (Stoppelkamp et al., 2011). These data are in
(Jacobson et al., 1996; Johansson et al., 1997; Li accordance with former work showing that, in
etal.,2008). primary cultures of cerebellar granule cells, low
Caffeine could also mediate its long-term doses of caffeine (125M) are able to counteract
effects through A2A receptor blockade. During A25-35 toxicity, an effect mimicked by ZM241385,
aging, we and others have found compelling evi- an A2A receptor antagonist but not CPT, a selec-
dence of cortical and hippocampal increases of tive A1 receptor antagonist (DallIgna et al.,
A2A receptor expression/function. Specifically, 2003). These protective effects were confirmed
in the hippocampus of aged rats, A2A receptor in vivo. Subchronic treatment with caffeine at a
expression is nearly two-fold higher compared to dose of 30mg/kg protected against aversive and
young rats (Cunha etal., 1995; Lopes etal., 1999). working- memory deficits induced by intrac-
More important, the A2A receptor-dependent erebroventricular (ICV) injection of A2535 in
activation of glutamate release becomes more mice (DallIgna et al., 2007) and mimicked by
pronounced as aging progresses and shifts administration of SCH58261, a selective A2A
from protein kinase C-mediated signaling to receptor antagonist. A2A receptor blockade,
cAMP-dependent effects (Lopes et al., 1999; through intraperitoneal injection of SCH58261
Rebola etal., 2005). This is accompanied by clear and KW6002 or genetic knockout, also pre-
behavioral deficits in hippocampus-dependent vented working-memory impairment as well as
tasks, such as spatial memory in rats (Diogenes synaptic loss induced by ICV injection of A142
etal., 2011). Accordingly, rats overexpressing hip- (Canas etal., 2009; Cunha etal., 2008). Improved
pocampal A2A receptors also exhibit behavioral working-memory following A 2A receptor block-
deficits, including spatial memory defects as well ade was thought to be related to prevention of
464 Part IV:Homeostatic Therapies

synaptotoxicity promoted by A-through modu- recently, we established that reducing A2A recep-
lation of p38 MAPK and mitochondrial function tor function improves memory and Tau pathol-
(Canas etal.,2009). ogy in a Tau transgenic mouse model (Laurent
Interestingly, memory improvement pro- etal., 2014b). This would suggest that A 2A receptor
moted by A2A receptor blockade following ICV modulation impacts on both pathological sides
injection of A was not observed in amnestic of AD, supporting interest toward A 2AR-based
conditions induced by MK801 or scopolamine strategiesinAD.
(Cunha etal., 2008). This reinforces the idea that Data from DallIgna et al., (2003) and Canas
A2A receptor blockade targets specific synaptic et al. (2009) suggest that protection afforded by
mechanisms rather than general mechanisms A2A receptor blockade against A is ascribed to
controlling memory (Cunha et al., 2008; and neuron-autonomous mechanisms. As discussed
Gomes et al., 2011, for discussion). However, earlier in this chapter, astroglial and microglial
this possibility should be tempered since, physi- reactions are AD hallmarks, and both type of
ologically, the A2A receptor blockade is procog- cells express A2A receptors. Therefore, nonneu-
nitive and particularly favors working memory, ronal autonomous mechanisms are also expected.
but apparently not reference memory that needs The role of A2A receptors expressed by astro- and
memory consolidation (Wei et al., 2011; Zhou microglial cells is far from understood (for a
et al., 2009). This raises the possibility that the review see Hasko etal., 2005). A2A receptors may
overall cognitive effect observed in the para- promote activation and proliferation of astroglial
digms discussed previously may not be entirely cells (Hindley etal., 1994; Ke etal., 2009)as well as
related to normalization of synaptic defects their ability to release glutamate (Nishizaki, 2004),
induced by A, but rather involve more com- while A2A receptor blockade counteracts the astro-
plex regulation. Further, the extent to which A2A cyte induction driven by basic fibroblast growth
receptor modulation impacts on amyloidogenesis factor (Brambilla etal., 2003). However, other stud-
remains totally unknown sofar. ies indicate that A2A receptor stimulation inhibits
Contrasting with the available encouraging nitric oxide and TNF release by astrocytes (Brodie
data supporting that caffeine and A 2A receptor et al., 1998). Concerning microglial cells, it has
blockade impacts on the amyloid side of AD, been shown that A2A receptor stimulation causes
much less is known about the impact of adeno- microglial activation (Orr etal., 2009)and potenti-
sine receptor modulation on Tau pathology. ates the release of nitric oxide as well as prostaglan-
So far, few data are available, and most studies dine E2 release from these cells (Fiebich etal., 1996;
have focused on the effects of caffeine. Caffeine Saura etal., 2005). Also, different experimental evi-
protected against the death of rat primary hip- dence support the anti-inflammatory effect of A2A
pocampal neurons expressing a mutated Tau receptor blockade in different neuropathological
protein (Stoppelkamp etal., 2011). Furthermore, situations (Dai etal., 2010a; Rebola etal., 2011; Yu
caffeine mitigated Tau hyperphosphorylation et al., 2008). In particular, A2A receptor blockade
induced by a cholesterol-rich diet in rabbits mitigates LTP defects as well as microglial activa-
(Prasanthi et al., 2010). In line with this, caf- tion and IL1 release following LPS administra-
feine, at doses irrelevant of human consump- tion (Rebola etal., 2011). Of interest, the role of A2A
tion, reduced Tau phoshorylation in rat primary receptors may vary depending on the pathophysio-
cortical neurons (Currais et al., 2011). Caffeine logical context and in particular with regard to the
effects would rely at least on GSK3 modula- local environment. Indeed, although A2A receptors
tion, but the underlying mechanisms remain may have anti-inflammatory functions under low
largely unknown. However, we recently obtained glutamate conditions, their function shifts toward
results supporting the notion that caffeine miti- a proinflammatory function under high glutamate
gates memory defects and Tau pathology in a concentrations following a transduction shift (Dai
transgenic mouse model of AD- Tau pathology etal., 2010b).
(Laurent etal., 2014a). Whether these effects are A 2AR upregulation has also been observed
mediated through adenosine receptors remains in the microglia from AD patients (Angulo
unclear. Several years ago, A1 receptor activation etal., 2003). Further, experimental studies point
was found to promote Tau phosphoryation in a out that amyloid peptides can promote an A2A
neuroblastoma model (Angulo etal., 2003), sup- receptor upsurge in both micro- and astroglial
porting that the effect of caffeine toward Tau may cells (Orr etal., 2009; Matos etal., 2012). Recent
be mediated through this receptor subtype. More observations indicate that inhibitory effects of
Adenosine and Alzheimers Disease 465

A on astrocytic glutamate uptake is prevented Astrogliosis and the associated increase


by A2A receptor blockade (Matos et al., 2012). in ADK leading to focal adenosine deficiency
Together, this would suggest a tight relationship is sufficient to trigger seizures (Boison, 2008).
between glial A2A receptors and pathological pro- Given the tight association between astrogliosis
cesses underlying AD, which needs to be deeply and neuropathological lesions found in AD, it is
investigated. tempting to hypothesize that ADO dysfunction
may be instrumental for AD-related seizures. In
Adenosine, Adenosine Receptors, line with this idea, ADO deficiency as a result
and Seizures of transgenic overexpression of ADK is suffi-
Seizures and epilepsy are a common comorbid- cient to promote severe learning deficits in the
ity of AD (Menendez, 2005; Palop and Mucke, Morris water maze and in Pavlovian condition-
2009). Within the sporadic AD population there ing (Yee etal., 2007). The role of ADO deficiency
is an 87% increase in the incidence of unpro- and ADK on the generation of AD-related
voked clinical seizures compared to the aged seizures and memory deficits remains to be
matched reference population (Amatniek et al., established in reliable models and human
2006; Menendez, 2005). In addition to these brain tissue. Importantly, this possibility sug-
behavioral seizures, it is postulated that the gests that reduction of the adenosinergic tone
numerous episodes of severe confusion reported by increasing its metabolic clearance through
by AD patients is the occurrence of silent (i.e., overexpressed ADK is deleterious. Although
subclinical, electrographic) seizures (Thomas, this notion apparently contrasts with the pos-
1997). So far, seizure occurrence in AD has sible protective effect expected from adenosine
been ascribed to the amyloid side through the receptor blockade, especially of the A 2A subtype,
use of several APP transgenic mouse models. it needs to be stressed that the net effects of dis-
Transgenic mice that carry the human APP with rupted ADO homeostasis affect bioenergetic
the Swedish and Indiana mutations (hAPP J20) network regulation and encompass adenosine
exhibited spontaneous nonconvulsive seizures in receptor dependent, as well as adenosine recep-
cortical and hippocampal networks associated tor independent, aspects of homeostatic net-
with synaptic plasticity deficits (Minkeviciene work control by ADO. Future work needs to
et al., 2009; Palop, 2007). In addition, seizure address adenosine receptor independent and
thresholds in the hAPP transgenic mice were epigenetic functions of ADO within the context
reduced in response to pentylentetrazol (Del of AD, integrate those functions with the bet-
Vecchio etal., 2004; Palop, 2007). The spontane- ter characterized adenosine receptor depend-
ous and chemically induced seizure phenotypes ent functions of adenosine, and ascribe specific
in the APP mutant were attenuated by genetically roles of adenosine-dependent homeostatic net-
removing the endogenous Tau protein, thereby work disruption to cognitive and seizure phe-
restoring synaptic function and reducing hyper- notypes of AD. Eventually, it might be possible
excitability (Roberson et al., 2007; Roberson to treat or possibly cure AD synergistically by
etal., 2011). These data raise the possibility that reconstructing homeostatic bioenergetic net-
amyloid may be instrumental in seizure occur- work functions that depend on correct ADO
rence observed in AD patients but also that these homeostasis. Therefore, carefully tailored focal
seizures relate to A-Tau interaction. To this end, ADO augmentation strategies, perhaps in com-
targeted gene therapy with a Tau antisense oligo- bination with A 2A receptor blockade, might hold
nucleotide designed to decrease endogenous Tau promise for the future therapyofAD.
throughout the brain reduced seizure severity
in two independent chemically induced seizure Adenosine asa Potential Epigenetic
models (DeVos etal., 2013). Interestingly, epilep- Modulator forMemory Improvement
tic activity and reduced seizure thresholds have As already described in detail, DNA methylation
been observed in different tauopathies, includ- status is dependent on a biochemical transmeth-
ing fronto-temporal dementia linked to chro- ylation reaction whereby SAM donates a methyl
mosome 17 and progressive supranuclear palsy group to unmethylated cytosines in DNA and is
(Garcia-Cabero et al., 2013; Beach et al., 2003; converted to SAH. SAH is further hydrolyzed to
Nygaard etal., 1989). However, whether AD-like ADO and Hcy. ADO is cleared by ADK-mediated
Tau pathology is also able to promote seizure phosphorylation to AMP and Hcy is converted
occurrence has yet to be established. to methionine in a folate-dependent manner.
466 Part IV:Homeostatic Therapies

Importantly, DNA methylation is dependent on tone nonspecifically drives the transmethylation


the continuous removal and subsequent equi- pathway by regulating substrate availability. Any
librium constants of SAH, ADO, and Hcy (Lu, site-specific DNA methylation changes would be
2000; Boison etal., 2002). Thus an accumulation mediated by DNMT1, 3a or 3b complexes (Goll
of ADO or Hcy prevents the biochemical conver- and Bestor, 2005; Caiafa etal., 2009; Feng etal.,
sion of SAM to SAH and consequentially inhibits 2010; Zampieri etal., 2012). Thus the ADO tone
DNA methylation. does not regulate site-specific DNA methylation
The effects of ADO tone on the DNA meth- but instead the homeostasis of the methylome.
ylation status are evident from humans with Therefore, any modification of this tone is likely
a missense mutation in the Adk gene. Loss of to impact on gene expression. Ultimately, this
ADK function prevents ADO clearance, which tone may have a significant impact on the expres-
consequently blocks the methionine cycle lead- sion of AD susceptibility genes or their regulators
ing to an increase in plasma SAH and SAM lev- or of those implicated in memory and plasticity
els, while Hcy levels remain normal or mildly (see previous discussion).
elevated (Bjursell etal., 2011). These findings are Albeit a global biochemical shift in the trans-
recapitulated in two rodent models of hyper- methylation pathway has profound physiological
methioninemia involving either global or focal, effects, humans without ADK have abnormal
forebrain restricted, ADK knockout. Global liver function, develop encephalopathy, and have
ADK knockout mice have a pathological shift severe progressive neurological deficits includ-
in the transmethylation pathway as evidenced ing learning and memory impairments and sei-
by decreased blood adenine levels and increased zures (Bjursell etal., 2011). Similarly, mice with
Hcy levels (Boison et al., 2002). Similarly, focal a global ADK knockout develop the fatal liver
ADK knockout increases hippocampal ADO disease neonatal hepatic steatosis (Boison et al.,
3.3-fold (Shen et al., 2011) and decreases DNA 2002). Transgenic mice with ADK overexpres-
methylation by 30% (Williams-Karnesky et al., sion throughout the brain have working and ref-
2013). Independent of permanent genetic muta- erence memory impairments in the Morris water
tions that influence the methionine cycle, both maze (Yee et al., 2007). Furthermore, patients
systemic and focal administration of pharmaco- with mesial temporal lobe epilepsy including hip-
logical agents also influence the DNA methyla- pocampal sclerosis frequently have dementia as a
tion status. Systemic administration of the ADK comorbidity and are characterized by increased
inhibitor 5-iodotubercidin (3.1 mg/kg ip once astrogliosis and ADK expression.
every day for 5 days) decreases the global DNA The association between pathological changes
methylation status by 35% in the hippocam- in ADK expression and memory deficits may be
pus of wild-type mice. Likewise, focal deliv- through the inability to mount de novo changes
ery of ADO and Hcy to nave rats (ICV, single in the DNA methylation status. It is now read-
bolus) decreases hippocampal DNA methyla- ily accepted that the epigenome of mature cells
tion (Williams-Karnesky etal., 2013). Thus DNA within the central nervous system normally
methylation is responsive to de novo changes in undergoes modifications that involve both his-
ADO tissuetone. tones and DNA methylation (Feng et al., 2010;
While low ADK and high ADO can decrease Ma et al., 2010). These changes occur regularly
DNA methylation, the transmethylation path- and rapidly and can include both active meth-
way can also be shifted to increased meth- ylation and demethylation. For example, an
ylation as a result of high ADK and low ADO episode of neural synchronization induces hip-
tone. DNA hypermethylation is observed with pocampal DNA methylationdependent changes
both a pharmacological, focal delivery of SAM in the transcription of immediate early genes
to nave rats (ICV, single bolus) and a patho- and initiates a cascade of transcription factors
logical, injury-induced increase in ADK (kainic contributing to long-term neuronal and circuit
acidinduced status epilepticus). Importantly, plasticity (Nelson et al., 2008). Furthermore,
using cultured ADK knockout cells transiently activity-induced adult neurogenesis is depend-
transfected with either the short or long isoform of ent on GADD45b-mediated active DNA
ADK, the nuclear (long) isoform has the greatest demethylation of the Bdnf and Fgf1 genes within
effect on increasing the DNA methylation status dentate granular neurons (Ma et al., 2009).
(Williams-Karnesky et al., 2013). As an obliga- Aproper regulation of DNA methylation may be
tory end product of transmethylation, the ADO of prime importance for fear-memory formation,
Adenosine and Alzheimers Disease 467

particularly hypermethylation of the PP1 gene DISTALZ (Development of Innovative Strategies


and demethylation of the reelin gene (Miller and for a Transdisciplinary approach to Alzheimers
Sweatt, 2007). Altered PP1 levels (through ADO disease), Inserm, CNRS, Universit Lille 2, Rgion
tone-induced methylation deregulation) might Nord/Pas-de-Calais, ANR (ADORATAU),
then subsequently impact histone modifications ERA-Net (ABeta-ID) and FUI MEDIALZ. Olivier
(phosphorylation but also acetylation) related to Bousiges and Anne-Laurence Boutillier are sup-
long-term memory (Koshibu etal., 2009). Thus by ported by CNRS, Universit de Strasbourg,
manipulating the endogenous ADO tone to shift Hopital Universitaire Strasbourg, ANR (ANR-12-
the transmethylation pathway to either increase MALZ-0002-01 ATACTAD), the Indo-French
or decrease the DNA methylation status, it may Centre for the Promotion of Advanced Research
be feasible to restore learning and memory defi- (IFCPAR No.4803-3) and Alsace Alzheimer 67
cits by promoting neurogenesis, neuronal plastic- association. Vanessa Flaten holds a grant from
ity, and memory signaling. Inserm/Region Nord pas de Calais. Detlev Boison
is supported through grants from the National
CONCLUSION Institutes of Health (NS065957, MH083973,
Network homeostasis is strongly compromised in NS061844), the US Department of the Army
AD, and there is an interest to reinstate network (W81XWH-12-1-0283), and the Legacy Good
function as well as neuroglial communication Samaritan Foundation. L.V. Lopes is an iFCT
to improve cognition as well as mitigate lesions. investigator. David Blum is an Inserm investigator.
The adenosinergic system is a regulatory system
impaired in AD that deserves interest as a drug- References
gable entity. On one hand, ADO itself is a critical Aisen, P.S., Egelko, S., Andrews, H., Diaz-Arrastia, R.,
modulator of both network function and epige- Weiner, M., DeCarli, C., Jagust, W., Miller, J.W.,
netic regulation. Reconstitution of ADO-based Green, R., Bell, K., etal. (2003). A pilot study of
homeostatic network functions would be of ther- vitamins to lower plasma homocysteine levels in
apeutic interest and thus deserves preclinical and Alzheimer disease. Am J Geriatr Psychiatry 11,
clinical evaluations. On the other hand, ADO 246249.
A2A receptor dysregulation may be instrumental Aisen, P.S., Schneider, L.S., Sano, M., Diaz-Arrastia, R.,
in AD. In line with this notion, several lines of van Dyck, C.H., Weiner, M.F., Bottiglieri, T.,
evidence indicate that caffeine administration Jin, S., Stokes, K.T., Thomas, R.G., et al. (2008).
and A2A receptor blockade constitute promising High-dose B vitamin supplementation and cog-
therapeutic approaches for AD. So far, no clini- nitive decline in Alzheimer disease:Arandom-
ized controlled trial. JAMA 300, 17741783.
cal trials have been performed to date to evalu-
Albasanz, J.L., Perez, S., Barrachina, M., Ferrer, I.,
ate the extent by which caffeine can slow down
and Martin, M. (2008). Up-regulation of adenos-
disease progression in AD patients. Further,
ine receptors in the frontal cortex in Alzheimers
although A2A antagonists appear safe and toler-
disease. Brain Pathol 18, 211219.
able in Parkinsons disease patients (for reviews
Albrecht, S., Bourdeau, M., Bennett, D., Mufson, E.J.,
see Morelli et al., 2009; Pinna, 2009; Simola
Bhattacharjee, M., and LeBlanc, A.C. (2007).
et al., 2008), no study has evaluated effects in Activation of caspase-6 in aging and mild cog-
AD patients, and the major drawback for the use nitive impairment. Am J Pathol 170, 12001209.
of A2A receptor antagonists in AD is the lack of Amatniek, J.C., Hauser, W.A., DelCastillo-Castaneda,
experimental studies and pilot clinical studies C., Jacobs, D.M., Marder, K., Bell, K., Albert,
assessing their effects on cognitive impairment M., Brandt, J., and Stern, Y. (2006). Incidence
during aging or in AD patients. Overall, modu- and predictors of seizures in patients with
lation of the adenosinergic system is certainly Alzheimers disease. Epilepsia 47, 867872.
important to consider for the treatment of AD, Amieva, H., Jacqmin-Gadda, H., Orgogozo, J.M.,
but precise evaluations of its importance for Le Carret, N., Helmer, C., Letenneur, L.,
pathological and memory processes await fur- Barberger-Gateau, P., Fabrigoule, C., and
ther experimental work in the future. Dartigues, J.F. (2005). The 9 year cognitive
decline before dementia of the Alzheimer
ACK NOWLEDGMENTS type: A prospective population-based study.
David Blum and Luc Bue are supported by Brain 128, 10931101.
France Alzheimer, LECMA/Alzheimer Forschung Angulo, E., Casado, V., Mallol, J., Canela, E.I., Vinals, F.,
Initiative, LabEx (Excellence Laboratory) Ferrer, I., Lluis, C., and Franco, R. (2003). A1
468 Part IV:Homeostatic Therapies

adenosine receptors accumulate in neurode- Benilova, I., Karran, E., and De Strooper, B. (2012).
generative structures in Alzheimer disease and The toxic A oligomer and Alzheimers dis-
mediate both amyloid precursor protein process- ease:an emperor in need of clothes. Nat Neurosci.
ing and tau phosphorylation and translocation. 15, 349357.
Brain Pathol 13, 440451. Bettens, K., Sleegers, K., and Van Broeckhoven, C.
Arendash, G.W., Mori, T., Cao, C., Mamcarz, M., (2013). Genetic insights in Alzheimers disease.
Runfeldt, M., Dickson, A., Rezai-Zadeh, K., Lancet Neurol. 12, 92104.
Tane, J., Citron, B.A., Lin, X., et al. (2009). Bhaskar, K., Konerth, M., Kokiko-Cochran, O.N.,
Caffeine reverses cognitive impairment and Cardona, A., Ransohoff, R.M., and Lamb,
decreases brain amyloid-beta levels in aged B.T. (2010). Regulation of tau pathology by
Alzheimers disease mice. J Alzheimers Dis 17, the microglial fractalkine receptor. Neuron
661680. 68,1931.
Arendash, G.W., Schleif, W., Rezai-Zadeh, K., Bjursell, M.K., Blom, H.J., Cayuela, J.A., Engvall,
Jackson, E.K., Zacharia, L.C., Cracchiolo, J.R., M.L., Lesko, N., Balasubramaniam, S.,
Shippy, D., and Tan, J. (2006). Caffeine protects Brandberg, G., Halldin, M., Falkenberg, M.,
Alzheimers mice against cognitive impairment Jakobs, C., et al. (2011). Adenosine kinase defi-
and reduces brain beta-amyloid production. ciency disrupts the methionine cycle and causes
Neuroscience 142, 941952. hypermethioninemia, encephalopathy, and
Attems, J., and Jellinger, K.A. (2006). Hippocampal abnormal liver function. Am J Hum Genet 89,
sclerosis in Alzheimer disease and other demen- 507515.
tias. Neurology 66,775. Blum, D., Hourez, R., Galas, M.C., Popoli, P., and
Ballard, C., Gauthier, S., Corbett, A., Brayne, C., Schiffmann, S.N. (2003). Adenosine receptors and
Aarsland, D., and Jones, E. (2011). Alzheimers Huntingtons disease:Implications for pathogen-
disease. Lancet 377, 10191031. esis and therapeutics. Lancet Neurol 2, 366374.
Barrachina, M., and Ferrer, I. (2009). DNA methyla- Bogdanovic, O., and Veenstra, G.J. (2009). DNA
tion of Alzheimer disease and tauopathy-related methylation and methyl-CpG binding pro-
genes in postmortem brain. J Neuropathol Exp teins: Developmental requirements and func-
Neurol 68, 880891. tion. Chromosoma 118, 549565.
Barrachina, M., Moreno, J., Villar-Menendez, I., Bottiglieri, T., Godfrey, P., Flynn, T., Carney,
Juves, S., and Ferrer, I. (2012). Histone tail acety- M.W., Toone, B.K., and Reynolds, E.H. (1990).
lation in brain occurs in an unpredictable fash- Cerebrospinal fluid S-adenosylmethionine in
ion after death. Cell Tissue Bank 13, 597606. depression and dementia: Effects of treatment
Batalha, V.L., Pego, J.M., Fontinha, B.M., Costenla, with parenteral and oral S-adenosylmethionine.
A.R., Valadas, J.S., Baqi, Y., Radjainia, H., J Neurol Neurosurg Psychiatry 53, 10961098.
Muller, C.E., Sebastiao, A.M., and Lopes, L.V. Boison, D. (2008). The adenosine kinase hypothesis
(2013). Adenosine A(2A) receptor blockade of epileptogenesis. Prog Neurobiol 84, 249262.
reverts hippocampal stress-induced deficits and Boison, D. (2009). Adenosine-based modulation of
restores corticosterone circadian oscillation. Mol brain activity. Curr Neuropharmacol 7, 158159.
Psychiatry 18, 320331. Boison, D., Chen, J.F., and Fredholm, B.B. (2009).
Beach, T.G., Sue, L., Scott, S., Layne, K., Newell, A., Adenosine signalling and function in glial cells.
Walker, D., Baker, M., Sahara, N., Yen, S.H., Cell Death Differ 17, 10711082.
Hutton, M., et al. (2003). Hippocampal sclero- Boison, D., Scheurer, L., Zumsteg, V., Rulicke, T.,
sis dementia with tauopathy. Brain Pathol 13, Litynski, P., Fowler, B., Brandner, S., and Mohler,
263278. H. (2002). Neonatal hepatic steatosis by disrup-
Belarbi, K., Burnouf, S., Fernandez-Gomez, F.J., tion of the adenosine kinase gene. Proc Natl
Laurent, C., Lestavel, S., Figeac, M., Sultan, A., Acad Sci USA 99, 69856990.
Troquier, L., Leboucher, A., Caillierez, R., etal. Borota, D., Murray, E., Keceli, G., Chang, A.,
(2011). Beneficial effects of exercise in a trans- Watabe, J.M., Ly, M., Toscano, J.P., and Yassa,
genic mouse model of Alzheimers disease-like M.A. (2014). Post-study caffeine administration
Tau pathology. Neurobiol Dis 43, 486494. enhances memory consolidation in humans. Nat
Bellucci, A., Westwood, A.J., Ingram, E., Casamenti, F., Neurosci 17, 201203.
Goedert, M., and Spillantini, M.G. (2004). Braak, H., and Braak, E. (1991). Neuropathological
Induction of inflammatory mediators and stageing of Alzheimer-related changes. Acta
microglial activation in mice transgenic for Neuropathol. 82, 239259.
mutant human P301S tau protein. Am J Pathol Bradshaw, E.M., Chibnik, L.B., Keenan, B.T.,
165, 16431652. Ottoboni, L., Raj, T., Tang, A., Rosenkrantz,
Adenosine and Alzheimers Disease 469

L.L., Imboywa, S., Lee, M., Von Korff, A., etal. G., Cai, J., etal. (2012). High blood caffeine levels
(2013). CD33 Alzheimers disease locus: altered in MCI linked to lack of progression to dementia.
monocyte function and amyloid biology. Nat J Alzheimers Dis 30, 559572.
Neurosci. 16, 848880. Cao, X., and Sudhof, T.C. (2001). A transcriptionally
Brambilla, R., Cottini, L., Fumagalli, M., Ceruti, S., [correction of transcriptively] active complex
and Abbracchio, M.P. (2003). Blockade of A2A of APP with Fe65 and histone acetyltransferase
adenosine receptors prevents basic fibroblast Tip60. Science 293, 115120.
growth factor-induced reactive astrogliosis in rat Cervoni, N., and Szyf, M. (2001). Demethylase activ-
striatal primary astrocytes. Glia 43, 190194. ity is directed by histone acetylation. J Biol Chem
Brodie, C., Blumberg, P.M., and Jacobson, K.A. 276, 4077840787.
(1998). Activation of the A2A adenosine recep- Chan, A., Paskavitz, J., Remington, R., Rasmussen, S.,
tor inhibits nitric oxide production in glial cells. and Shea, T.B. (2008). Efficacy of a vitamin/nutri-
FEBS Lett 429, 139142. ceutical formulation for early-stage Alzheimers
Brouillette, J., Caillierez, R., Zommer, N., Alves-Pires, disease:A1-year, open-label pilot study with an
C., Benilova, I., Blum, D., De Strooper, B., and 16-month caregiver extension. Am J Alzheimers
Buee, L. (2012). Neurotoxicity and memory defi- Dis 23, 571585.
cits induced by soluble low-molecular-weight Chandra, V., Ganguli, M., Pandav, R., Johnston, J.,
amyloid-beta142 oligomers are revealed in vivo Belle, S., DeKosky, S.T. (1998). Prevalence of
by using a novel animal model. J Neurosci 32, Alzheimers disease and other dementias in
78527861. rural India: The Indo-US study. Neurology 51,
Burnouf, S., Martire, A., Derisbourg, M., Laurent, C., 10001008.
Belarbi, K., Leboucher, A., Fernandez-Gomez, F.J., Chandra, V., Pandav, R., Dodge, H.H., Johnston,
Troquier, L., Eddarkaoui, S., Grosjean, M.E., J.M., Belle, S.H., DeKosky, S.T., Ganguli, M.
et al. (2013). NMDA receptor dysfunction con- (2001). Incidence of Alzheimers disease in a
tributes to impaired brain-derived neurotrophic rural community in India: The Indo-US study.
factor-induced facilitation of hippocampal syn- Neurology 57, 985989.
aptic transmission in a Tau transgenic model. Chatterjee, S., Mizar, P., Cassel, R., Neidl, R., Selvi, B.R.,
Aging Cell 12,1123. Mohankrishna, D.V., Vedamurthy, B.M.,
Buee, L., Bussiere, T., Buee-Scherrer, V., Delacourte, A., Schneider, A., Bousiges, O., Mathis, C., et al.
and Hof, P.R. (2000). Tau protein isoforms, phos- (2013). A novel activator of CBP/p300 acetyl-
phorylation and role in neurodegenerative disor- transferases promotes neurogenesis and extends
ders. Brain Res Brain Res Rev 33, 95130. memory duration in adult mice. J Neurosci 33,
Caccamo, A., Maldonado, M.A., Bokov, A.F., 1069810712.
Majumder, S., and Oddo, S. (2010). CBP gene Checler, F. (1995). Processing of the beta-amyloid pre-
transfer increases BDNF levels and ameliorates cursor protein and its regulation in Alzheimers
learning and memory deficits in a mouse model disease. J Neurochem 65, 14311444.
of Alzheimers disease. Proc Natl Acad Sci USA Chen, K.L., Wang, S.S., Yang, Y.Y., Yuan, R.Y., Chen,
107, 2268722692. R.M., and Hu, C.J. (2009). The epigenetic effects
Cagnin, A., Brooks, D.J., Kennedy, A.M., Gunn, R.N., of amyloid-beta(1-40) on global DNA and nepri-
Myers, R., Turkheimer, F.E., Jones, T., and lysin genes in murine cerebral endothelial cells.
Banati, R.B. (2001). In-vivo measurement of Biochem Biophys Res Commun 378,5761.
activated microglia in dementia. Lancet 358, Clarke, R., Smith, A.D., Jobst, K.A., Refsum, H.,
461467. Sutton, L., and Ueland, P.M. (1998). Folate, vita-
Caiafa, P., Guastafierro. T., and Zampieri, M. (2009). min B12, and serum total homocysteine levels in
Epigenetics: Poly(ADP-ribosyl)ation of PARP-1 confirmed Alzheimer disease. Arch Neurol 55,
regulates genomic methylation patterns. FASEB 14491455.
J 23, 672678. Cohen, T.J., Friedmann, D., Hwang, A.W.,
Canas, P.M., Porciuncula, L.O., Cunha, G.M., Silva, Marmorstein, R., Lee, V.M. (2013). The
C.G., Machado, N.J., Oliveira, J.M., Oliveira, microtubule-associated tau protein has intrinsic
C.R., and Cunha, R.A. (2009). Adenosine A2A acetyltransferase activity. Nat Struct Mol Biol 20,
receptor blockade prevents synaptotoxicity and 756762.
memory dysfunction caused by beta-amyloid Cohen, T.J., Guo, J.L., Hurtado, D.E., Kwong, L.K.,
peptides via p38 mitogen-activated protein Mills, I.P., Trojanowski, J.Q., and Lee, V.M.
kinase pathway. J Neurosci 29, 1474114751. (2011). The acetylation of tau inhibits its function
Cao, C., Loewenstein, D.A., Lin, X., Zhang, C., and promotes pathological tau aggregation. Nat
Wang, L., Duara, R., Wu, Y., Giannini, A., Bai, Commun 2,252.
470 Part IV:Homeostatic Therapies

Cook, C., Carlomagno, Y., Gendron, T.F., Dunmore, J., A2A receptor regulation of neuroinflamma-
Scheffel, K., Stetler, C., Davis, M., Dickson, D., tion and traumatic brain injury. J Neurosci 30,
Jarpe, M., DeTure, M., and Petrucelli, L. (2014). 58025810.
Acetylation of the KXGS motifs in tau is a criti- DallIgna, O.P., Fett, P., Gomes, M.W., Souza, D.O.,
cal determinant in modulation of tau aggrega- Cunha, R.A., and Lara, D.R. (2007). Caffeine and
tion and clearance. Hum Mol Genet 23, 104116. adenosine A(2a) receptor antagonists prevent
Cook, R.H., Schneck, S.A., and Clark, D.B. (1981). beta-amyloid (2535)-induced cognitive deficits
Twins with Alzheimers disease. Arch Neurol 38, in mice. Exp Neurol 203, 241245.
300301. DallIgna, O.P., Porciuncula, L.O., Souza, D.O.,
Cooney, C.A. (1993). Are somatic cells inherently Cunha, R.A., and Lara, D.R. (2003).
deficient in methylation, metabolism? A pro- Neuroprotection by caffeine and adenosine A2A
posed mechanism for DNA methylation loss, receptor blockade of beta-amyloid neurotoxicity.
senescence and aging. Growth Dev Aging 57, Br J Pharmacol 138, 12071209.
261273. Del Vecchio, R.A., Gold, L.H., Novick, S.J., Wong, G.,
Cruz, J.C., Kim, D., Moy, L.Y., Dobbin, M.M., Sun, X., and Hyde, L.A. (2004). Increased seizure thresh-
Bronson, R.T., and Tsai, L.H. (2006). p25/ old and severity in young transgenic CRND8
cyclin-dependent kinase 5 induces production mice. Neurosci Lett 367, 164167.
and intraneuronal accumulation of amyloid beta de Mendonca, A., and Cunha, R.A. (2010).
in vivo. J Neurosci 26, 1053610541. Therapeutic opportunities for caffeine in
Cruz, J.C., Tseng, H.C., Goldman, J.A., Shih, H., Alzheimers disease and other neurodegenerative
and Tsai, L.H. (2003). Aberrant Cdk5 activation disorders. J Alzheimers Dis 20 Suppl 1,S1S2.
by p25 triggers pathological events leading to De Strooper, B. (2010). Proteases and proteolysis in
neurodegeneration and neurofibrillary tangles. Alzheimer disease:Amultifactorial view on the
Neuron 40, 471483. disease process. Physiol Rev 90, 465494.
Cunha, G.M., Canas, P.M., Melo, C.S., Hockemeyer, J., De Strooper, B., Vassar, R., and Golde, T. (2010). The
Muller, C.E., Oliveira, C.R., and Cunha, R.A. secretases: Enzymes with therapeutic potential
(2008). Adenosine A2A receptor block- in Alzheimer disease. Nat Rev Neurol 6, 99107.
ade prevents memory dysfunction caused by DeVos, S.L., Goncharoff, D.K., Chen, G., Kebodeaux,
beta-amyloid peptides but not by scopolamine or C.S., Yamada, K., Stewart, F.R., Schuler, D.R.,
MK-801. Exp Neurol 210, 776781. Maloney, S.E., Wozniak, D.F., Rigo, F., et al.
Cunha, R.A. (2008). [Caffeine, adenosine recep- (2013). Antisense reduction of tau in adult
tors, memory and Alzheimer disease]. Med Clin mice protects against seizures. J Neurosci 33,
(Barc) 131, 790795. 1288712897.
Cunha, R.A., Almeida, T., and Ribeiro, J.A. (2001). Diogenes, M.J., Costenla, A.R., Lopes, L.V.,
Parallel modification of adenosine extracellular Jeronimo-Santos, A., Sousa, V.C., Fontinha, B.M.,
metabolism and modulatory action in the hippo- Ribeiro, J.A., and Sebastiao, A.M. (2011).
campus of aged rats. J Neurochem 76, 372382. Enhancement of LTP in aged rats is dependent on
Cunha, R.A., Constantino, M.C., Sebastiao, A.M., endogenous BDNF. Neuropsychopharmacology
and Ribeiro, J.A. (1995). Modification of A1 36, 18231836.
and A2a adenosine receptor binding in aged Donmez, G., Wang, D., Cohen, D.E., and Guarente, L.
striatum, hippocampus and cortex of the rat. (2010). SIRT1 suppresses beta-amyloid pro-
Neuroreport 6, 15831588. duction by activating the alpha-secretase gene
Currais, A., Kato, K., Canuet, L., Ishii, R., Tanaka, T., ADAM10. Cell 142, 320332.
Takeda, M., and Soriano, S. (2011). Caffeine Dostal, V., Roberts, C.M., and Link, C.D. (2010).
modulates tau phosphorylation and affects Akt Genetic mechanisms of coffee extract pro-
signaling in postmitotic neurons. J Mol Neurosci tection in a Caenorhabditis elegans model of
43, 326332. beta-amyloid peptide toxicity. Genetics 186,
Dai, S.S., Li, W., An, J.H., Wang, H., Yang, N., Chen, X.Y., 857866.
Zhao, Y., Li, P., Liu, P., Chen, J.F., etal. (2010a). Duyckaerts, C., Bennecib, M., Grignon, Y., Uchihara,
Adenosine A2A receptors in both bone mar- T., He, Y., Piette, F., and Hauw, J.J. (1997).
row cells and non-bone marrow cells contrib- Modeling the relation between neurofibrillary
ute to traumatic brain injury. J Neurochem 113, tangles and intellectual status. Neurobiol Aging
15361544. 18, 267273.
Dai, S.S., Zhou, Y.G., Li, W., An, J.H., Li, P., Yang, N., El Khoury, J., Toft, M., Hickman, S.E., Means, T.K.,
Chen, X.Y., Xiong, R.P., Liu, P., Zhao, Y., et al. Terada, K., Geula, C., and Luster, A.D. (2007).
(2010b). Local glutamate level dictates adenosine Ccr2 deficiency impairs microglial accumulation
Adenosine and Alzheimers Disease 471

and accelerates progression of Alzheimer-like lifetime of monozygotic twins. Proc Natl Acad
disease. Nat Med 13, 432438. Sci USA 102, 1060410609.
Eskelinen, M.H., and Kivipelto, M. (2010). Francis, Y.I., Diss, J.K., Kariti, M., Stephanou,
Caffeine as a protective factor in dementia and A., Latchman, D.S. (2007). p300 activation
Alzheimers disease. J Alzheimers Dis 20 Suppl 1, by Presenilin 1 but not by its M146L mutant.
S167S174. Neurosci Lett 413, 137140.
Eskelinen, M.H., Ngandu, T., Tuomilehto, J., Francis, Y.I., Fa, M., Ashraf, H., Zhang, H.,
Soininen, H., and Kivipelto, M. (2009). Midlife Staniszewski, A., Latchman, D.S., and Arancio,
coffee and tea drinking and the risk of late-life O. (2009). Dysregulation of histone acetylation
dementia: A population-based CAIDE study. J in the APP/PS1 mouse model of Alzheimers dis-
Alzheimers Dis 16,8591. ease. J Alzheimers Dis 18, 131139.
Fang, F., Lue, L.F., Yan, S., Xu, H., Luddy, J.S., Chen, D., Francis, Y.I., Stephanou, A., and Latchman, D.S.
Walker, D.G., Stern, D.M., Schmidt, A.M., Chen, (2006). CREB-binding protein activation by
J.X., etal. (2010). RAGE-dependent signaling in presenilin 1 but not by its M146L mutant.
microglia contributes to neuroinflammation, Neuroreport 17, 917921.
Abeta accumulation, and impaired learning/ Fredholm, B.B., Battig, K., Holmen, J., Nehlig, A.,
memory in a mouse model of Alzheimers dis- and Zvartau, E.E. (1999). Actions of caffeine in
ease. FASEB J 24, 10431055. the brain with special reference to factors that
Feng, J., Zhou, Y., Campbell, S.L., Le, T., Li, E., Sweatt, contribute to its widespread use. Pharmacol Rev
J.D., Silva, A.J., and Fan, G. (2010). Dnmt1 and 51, 83133.
Dnmt3a maintain DNA methylation and regu- Fredholm, B.B., Chen, J.F., Cunha, R.A.,
late synaptic function in adult forebrain neurons. Svenningsson, P., and Vaugeois, J.M. (2005).
Nat Neurosci 13, 423430. Adenosine and brain function. Int Rev Neurobiol
Fiebich, B.L., Biber, K., Lieb, K., van Calker, D., 63, 191270.
Berger, M., Bauer, J., and Gebicke-Haerter, P.J. Fuso, A., Nicolia, V., Cavallaro, R.A., and Scarpa, S.
(1996). Cyclooxygenase-2 expression in rat (2011). DNA methylase and demethylase activi-
microglia is induced by adenosine A2a-receptors. ties are modulated by one-carbon metabolism in
Glia 18, 152160. Alzheimers disease models. J Nurtri Biochem
Finkelstein, J.D. (2000). Pathways and regulation of 22, 242251.
homocysteine metabolism in mammals. Semin Fuso, A., Nicolia, V., Cavallaro, R.A., Ricceri, L.,
Thromb Hemost 26, 219225. DAnselmi, F., Coluccia, P., Calamandrei, G.,
Fischer, A. (2014). Targeting histone-modifications and Scarpa, S. (2008). B-vitamin depriva-
in Alzheimers disease. What is the evidence tion induces hyperhomocysteinemia and
that this is a promising therapeutic avenue? brain S-adenosylhomocysteine, depletes brain
Neuropharmacology 80, 95102. S-adenosylmethionine, and enhances PS1 and
Fischer, A., Sananbenesi, F., Mungenast, A., and Tsai, BACE expression and amyloid-beta deposition
L.H. (2010). Targeting the correct HDAC(s) to in mice. Mole Cell Neurosci 37, 731746.
treat cognitive disorders. Trends Pharmacol Sci Fuso, A., Nicolia, V., Ricceri, L., Cavallaro, R.A., Isopi,
31, 605617. E., Mangia, F., Fiorenza, M.T., and Scarpa, S.
Fischer, A., Sananbenesi, F., Wang, X., Dobbin, M., (2012). S-adenosylmethionine reduces the prog-
and Tsai, L.H. (2007). Recovery of learning and ress of the Alzheimer-like features induced by
memory is associated with chromatin remodel- B-vitamin deficiency in mice. Neurobiol Aging
ling. Nature 447, 178182. 33, 1482.e116.
Fisone, G., Borgkvist, A., and Usiello, A. (2004). Fuso, A., Seminara, L., Cavallaro, R.A., DAnselmi,
Caffeine as a psychomotor stimulant:Mechanism F., and Scarpa, S. (2005). S-adenosylmethionine/
of action. Cell Mol Life Sci 61, 857872. homocysteine cycle alterations modify DNA
Foundas, A.L., Macauley, B.L., Raymer, A.M., Maher, methylation status with consequent deregulation
L.M., Rothi, L.J., and Heilman, K.M. (1999). of PS1 and BACE and beta-amyloid production.
Ideomotor apraxia in Alzheimer disease and left Mole Cell Neurosci 28, 195204.
hemisphere stroke:Limb transitive and intransi- Garcia-Cabrero, A.M., Guerrero-Lopez, R., Giraldez,
tive movements. Neuropsychiatry Neuropsychol B.G., Llorens-Martin, M., Avila, J., Serratosa,
Behav Neurol 12, 161166. J.M., and Sanchez, M.P. (2013). Hyperexcitability
Fraga, M.F., Ballestar, E., Paz, M.F., Ropero, S., and epileptic seizures in a model of frontotempo-
Setien, F., Ballestar, M.L., Heine-Suner, D., ral dementia. Neurobiol Dis 58, 200208.
Cigudosa, J.C., Urioste, M., Benitez, J., et al. Gelber, R.P., Petrovitch, H., Masaki, K.H., Ross, G.W.,
(2005). Epigenetic differences arise during the and White, L.R. (2011). Coffee intake in midlife
472 Part IV:Homeostatic Therapies

and risk of dementia and its neuropathologic Paraxanthine, the primary metabolite of caf-
correlates. J Alzheimers Dis 23, 607615. feine, provides protection against dopaminergic
Georgiev, V., Johansson, B., and Fredholm, B.B. (1993). cell death via stimulation of ryanodine receptor
Long-term caffeine treatment leads to a decreased channels. Mol Pharmacol 74, 980989.
susceptibility to NMDA-induced clonic seizures Guerreiro, R., Wojtas, A., Bras, J., Carrasquillo, M.,
in mice without changes in adenosine A1 recep- Rogaeva, E., Majounie, E., Cruchaga, C., Sassi, C.,
tor number. Brain Res 612, 271277. Kauwe, J.S., Younkin, S., et al. (2013). TREM2
Goll, M.G., and Bestor, T.H. (2005). Eukaryotic cyto- variants in Alzheimers disease. N Engl J Med.
sine methyltransferases. Annu Rev Biochem 74, 368, 117127.
481514. Gunzburg, M.J., Perugini, M.A., and Howlett, G.J.
Gomes, C.V., Kaster, M.P., Tome, A.R., Agostinho, P.M., (2007). Structural basis for the recognition and
and Cunha, R.A. (2011). Adenosine receptors and cross-linking of amyloid fibrils by human apoli-
brain diseases: Neuroprotection and neurodegen- poprotein E. J Biol Chem 282, 3583135841.
eration. Biochim Biophys Acta 1808, 13801399. Guo, J.U., Su, Y., Zhong, C., Ming, G.L., and Song,
Gorlovoy, P., Larionov, S., Pham, T.T., and Neumann, H. H. (2011). Hydroxylation of 5-methylcytosine by
(2009). Accumulation of tau induced in neu- TET1 promotes active DNA demethylation in
rites by microglial proinflammatory mediators. the adult brain. Cell 145, 423434.
FASEB J 23, 25022513. Hanson, J.E., Deng, L., Hackos, D.H., Lo, S.C.,
Govindarajan, N., Agis-Balboa, C., Walter, J., Lauffer, B.E., Steiner, P., and Zhou, Q. (2013).
Sananbenesi, F., and Fischer, A. (2011). Sodium Histone deacetylase 2 cell autonomously sup-
butyrate improves memory function in an presses excitatory and enhances inhibitory
Alzheimers disease mouse model when adminis- synaptic function in CA1 pyramidal neurons. J
tered at an advanced stage of disease progression. Neurosci 33, 59245929.
J Alzheimers Dis 26, 187197. Haskell, C.F., Kennedy, D.O., Wesnes, K.A., and
Grff, J., Kim, D., Dobbin, M.M., and Tsai, L.H. Scholey, A.B. (2005). Cognitive and mood
(2011). Epigenetic regulation of gene expression improvements of caffeine in habitual consum-
in physiological and pathological brain pro- ers and habitual non-consumers of caffeine.
cesses. Physiol Rev 91, 603649. Psychopharmacology (Berl) 179, 813825.
Grff, J., Rei, D., Guan, J.S., Wang, W.Y., Seo, J., Hasko, G., Pacher, P., Vizi, E.S., and Illes, P. (2005).
Hennig, K.M., Nieland, T.J., Fass, D.M., Kao, P.F., Adenosine receptor signaling in the brain
Kahn, M., et al. (2012). An epigenetic blockade immune system. Trends Pharmacol Sci 26,
of cognitive functions in the neurodegenerating 511516.
brain. Nature 483, 222226. Heneka, M.T., Kummer, M.P., Stutz, A., Delekate, A.,
Grff, J., and Tsai, L.H. (2013a). Histone acetyla- Schwartz, S., Vieira-Saecker, A., Griep, A., Axt,
tion: Molecular mnemonics on the chromatin. D., Remus, A., Tzeng, T.C., etal. (2013). NLRP3 is
Nature Rev Neurosci 14, 97111. activated in Alzheimers disease and contributes to
Grff, J., and Tsai, L.H. (2013b). The potential of pathology in APP/PS1 mice. Nature. 493, 674678.
HDAC inhibitors as cognitive enhancers. Annu Heneka, M.T., and OBanion, M.K. (2007).
Rev Pharmacol Toxicol 53, 311330. Inflammatory processes in Alzheimers disease.
Grathwohl, S.A., Kalin, R.E., Bolmont, T., Prokop, S., J Neuroimmunol 184,6991.
Winkelmann, G., Kaeser, S.A., Odenthal, J., Herber, D.L., Roth, L.M., Wilson, D., Wilson, N.,
Radde, R., Eldh, T., Gandy, S., et al. (2009). Mason, J.E., Morgan, D., and Gordon, M.N.
Formation and maintenance of Alzheimers (2004). Time-dependent reduction in Abeta lev-
disease beta-amyloid plaques in the absence of els after intracranial LPS administration in APP
microglia. Nat Neurosci 12, 13611363. transgenic mice. Exp Neurol 190, 245253.
Grober, E., Dickson, D., Sliwinski, M.J., Buschke, H., Hindley, S., Herman, M.A., and Rathbone, M.P.
Katz, M., Crystal, H., and Lipton, R.B. (1999). (1994). Stimulation of reactive astrogliosis in
Memory and mental status correlates of modi- vivo by extracellular adenosine diphosphate or
fied Braak staging. Neurobiol Aging 20, 573579. an adenosine A2 receptor agonist. J Neurosci Res
Guan, J.S., Haggarty, S.J., Giacometti, E., Dannenberg, 38, 399406.
J.H., Joseph, N., Gao, J., Nieland, T.J., Zhou, Y., Hoffman, D.R., Marion, D.W., Cornatzer, W.E.,
Wang, X., Mazitschek, R., et al. (2009). HDAC2 and Duerre, J.A. (1980). S-adenosylmethionine
negatively regulates memory formation and syn- and S-adenosylhomocystein metabolism in
aptic plasticity. Nature 459,5560. isolated rat liver. Effects of L-methionine,
Guerreiro, S., Toulorge, D., Hirsch, E., Marien, M., L-homocystein, and adenosine. J Biol Chem
Sokoloff, P., and Michel, P.P. (2008). 255, 1082210827.
Adenosine and Alzheimers Disease 473

Holmes, C., Cunningham, C., Zotova, E., Woolford, Huttenlocher, J., Levey, A.I., Lah, J.J., etal. (2013).
J., Dean, C., Kerr, S., Culliford, D., and Perry, Variant of TREM2 associated with the risk of
V.H. (2009). Systemic inflammation and disease Alzheimers disease. N Engl J Med. 368, 107116.
progression in Alzheimer disease. Neurology 73, Jucker, M., and Walker, L.C. (2013). Self-propagation
768774. of pathogenic protein aggregates in neurodegen-
Hoover, B.R., Reed, M.N., Su, J., Penrod, R.D., erative diseases. Nature. 501,4551.
Kotilinek, L.A., Grant, M.K., Pitstick, R., Kamphuis, W., Middeldorp, J., Kooijman, L., Sluijs,
Carlson, G.A., Lanier, L.M., Yuan, L.L., et al. J.A., Kooi, E.J., Moeton, M., Freriks, M., Mizee,
(2010). Tau mislocalization to dendritic spines M.R., and Hol, E.M. (2014). Glial fibrillary acidic
mediates synaptic dysfunction independently of protein isoform expression in plaque related
neurodegeneration. Neuron 68, 10671081. astrogliosis in Alzheimers disease. Neurobiol
Hsia, A.Y., Masliah, E., McConlogue, L., Yu, G.Q., Aging 35, 492510.
Tatsuno, G., Hu, K., Kholodenko, D., Malenka, Kaplan, G.B., Greenblatt, D.J., Leduc, B.W.,
R.C., Nicoll, R.A., and Mucke, L. (1999). Thompson, M.L., and Shader, R.I. (1989).
Plaque-independent disruption of neural circuits Relationship of plasma and brain concentra-
in Alzheimers disease mouse models. Proc Natl tions of caffeine and metabolites to benzodiaz-
Acad Sci USA 96, 32283233. epine receptor binding and locomotor activity. J
Irwin, D.J., Cohen, T.J., Grossman, M., Arnold, S.E., Pharmacol Exp Ther 248, 10781083.
Xie, S.X., Lee, V.M., and Trojanowski, J.Q. (2012). Ke, R.H., Xiong, J., Liu, Y., and Ye, Z.R. (2009).
Acetylated tau, a novel pathological signature in Adenosine A2a receptor induced gliosis via Akt/
Alzheimers disease and other tauopathies. Brain NF-kappaB pathway in vitro. Neurosci Res 65,
135, 807818. 280285.
Ishrat, T., Hoda, M.N., Khan, M.B., Yousuf, S., Kennedy, B.P., Bottiglieri, T., Arning, E., Ziegler,
Ahmad, M., Khan, M.M., Ahmad, A., and Islam, M.G., Hansen, L.A., and Masliah, E. (2004).
F. (2009). Amelioration of cognitive deficits and Elevated S-adenosylhomocysteine in Alzheimer
neurodegeneration by curcumin in rat model of brain:Influence on methyltransferases and cog-
sporadic dementia of Alzheimers type (SDAT). nitive function. J Neural Trans 111, 547567.
Eur Neuropsychopharmacol 19, 636647. Kilgore, M., Miller, C.A., Fass, D.M., Hennig, K.M.,
Ittner, L.M., Ke, Y.D., Delerue, F., Bi, M., Gladbach, A., Haggarty, S.J., Sweatt, J.D., and Rumbaugh, G.
van Eersel, J., Wolfing, H., Chieng, B.C., Christie, (2010). Inhibitors of class 1 histone deacety-
M.J., Napier, I.A., et al. (2010). Dendritic func- lases reverse contextual memory deficits
tion of tau mediates amyloid-beta toxicity in in a mouse model of Alzheimers disease.
Alzheimers disease mouse models. Cell 142, Neuropsychopharmacology 35, 870880.
387397. Kim, H.S., Kim, E.M., Kim, N.J., Chang, K.A., Choi,
Iwata, N., Tsubuki, S., Takaki, Y., Watanabe, K., Y., Ahn, K.W., Lee, J.H., Kim, S., Park, C.H., and
Sekiguchi, M., Hosoki, E., Kawashima-Morishima, Suh, Y.H. (2004). Inhibition of histone deacety-
M., Lee, H.J., Hama, E., Sekine-Aizawa, Y., lation enhances the neurotoxicity induced by the
et al. (2000). Identification of the major C-terminal fragments of amyloid precursor pro-
Abeta1-42-degrading catabolic pathway in brain tein. J Neurosci Res 75, 117124.
parenchyma: Suppression leads to biochemical Kim, D., Nguyen, M.D., Dobbin, M.M., Fischer, A.,
and pathological deposition. Nat Med 6, 143150. Sananbenesi, F., Rodgers, J.T., Delalle, I., Baur,
Jacobson, K.A., von Lubitz, D.K., Daly, J.W., and J.A., Sui, G., Armour, S.M., etal. (2007). SIRT1
Fredholm, B.B. (1996). Adenosine receptor deacetylase protects against neurodegeneration
ligands: Differences with acute versus chronic in models for Alzheimers disease and amyo-
treatment. Trends Pharmacol Sci 17, 108113. trophic lateral sclerosis. EMBO J 26, 31693179.
Johansson, B., Georgiev, V., Lindstrom, K., and Kitazawa, M., Oddo, S., Yamasaki, T.R.,
Fredholm, B.B. (1997). A1 and A2A adenosine Green, K.N., and LaFerla, F.M. (2005).
receptors and A1 mRNA in mouse brain:Effect Lipopolysaccharide-induced inflammation exac-
of long-term caffeine treatment. Brain Res 762, erbates tau pathology by a cyclin-dependent
153164. kinase 5-mediated pathway in a transgenic model
Johnson-Kozlow, M., Kritz-Silverstein, D., Barrett- of Alzheimers disease. J Neurosci 25, 88438853.
Connor, E., and Morton, D. (2002). Coffee con- Koshibu, K., Grff, J., Beullens, M., Heitz, F.D.,
sumption and cognitive function among older Berchtold, D., Russig, H., Farinelli, M., Bollen,
adults. Am J Epidemiol 156, 842850. M., and Mansuy, I.M. (2009). Protein phospha-
Jonsson, T., Stefansson, H., Steinberg, S., Jonsdottir, I., tase 1 regulates the histone code for long-term
Jonsson, P.V., Snaedal, J., Bjornsson, S., memory. J Neurosci 29, 1307913089.
474 Part IV:Homeostatic Therapies

Kouzarides, T. (2007). Chromatin modifications and attenuates traumatic brain injury in the mouse
their function. Cell 128, 693705. cortical impact model. Neuroscience 151,
Kruman, I.I., Kumaravel, T.S., Lohani, A., Pedersen, 11981207.
W.A., Cutler, R.G., Kruman, Y., Haughey, N., Li, Y., Liu, L., Barger, S.W., and Griffin, W.S. (2003).
Lee, J., Evans, M., and Mattson, M.P. (2002). Interleukin-1 mediates pathological effects of
Folic acid deficiency and homocysteine impair microglia on tau phosphorylation and on synap-
DNA repair in hippocampal neurons and sen- tophysin synthesis in cortical neurons through a
sitize them to amyloid toxicity in experimental p38-MAPK pathway. J Neurosci 23, 16051611.
models of Alzheimers disease. J NeuroscI 22, Lim, G.P., Chu, T., Yang, F., Beech, W., Frautschy,
17521762. S.A., and Cole, G.M. (2001). The curry spice cur-
Kurt, M.A., Davies, D.C., and Kidd, M. (1999). cumin reduces oxidative damage and amyloid
Beta-amyloid immunoreactivity in astrocytes in pathology in an Alzheimer transgenic mouse. J
Alzheimers disease brain biopsies: An electron Neurosci 21, 83708377.
microscope study. Exp Neurol 158, 221228. Linden, J. (2001). Molecular approach to adenosine
Lambert, J.C., and Amouyel, P. (2011). Genetics of receptors:Receptor-mediated mechanisms of tis-
Alzheimers disease: New evidences for an old sue protection. Annu Rev Pharmacol Toxicol 41,
hypothesis? Curr Opin Genet Dev 21, 295301. 775787.
Lambert, J.C., Heath, S., Even, G., Campion, D., Lindsay, J., Laurin, D., Verreault, R., Hebert, R.,
Sleegers, K., Hiltunen, M., Combarros, O., Helliwell, B., Hill, G.B., and McDowell, I. (2002).
Zelenika, D., Bullido, M.J., Tavernier, B., et al. Risk factors for Alzheimers disease:Aprospec-
(2009). Genome-wide association study identi- tive analysis from the Canadian Study of Health
fies variants at CLU and CR1 associated with and Aging. Am J Epidemiol 156, 445453.
Alzheimers disease. Nat Genet 41, 10941099. Liu, R., Lei, J.X., Luo, C., Lan, X., Chi, L., Deng, P.,
La Rue, A. (2010). Healthy brain aging:Role of cogni- Lei, S., Ghribi, O., and Liu, Q.Y. (2012). Increased
tive reserve, cognitive stimulation, and cognitive EID1 nuclear translocation impairs synaptic
exercises. Clin Geriatr Med 26, 99111. plasticity and memory function associated with
Laurent, C., Burnouf, S., Ferry, B., Batalha, V.L., pathogenesis of Alzheimers disease. Neurobiol
Coelho, J.E., Baqi, Y., Malik, E., Mariciniak, E., Dis 45, 902912.
Parrot, S., Van der Jeugd, A., et al. (2014). A2A Lopes, L.V., Cunha, R.A., and Ribeiro, J.A. (1999).
adenosine receptor deletion is protective in a Cross talk between A(1) and A(2A) adenos-
mouse model of Tauopathy. Mol Psychiatry. ine receptors in the hippocampus and cortex
[Epub ahead of print]. of young adult and old rats. J Neurophysiol 82,
Laurent, C., Eddarkaoui, S., Derisbourg, M., 31963203.
Leboucher, A., Demeyer, D., Carrier, S., Lopes, L.V., Sebastiao, A.M., and Ribeiro, J.A.
Schneider, M., Hamdane, M., Mller, C.E., (2011). Adenosine and related drugs in brain dis-
Bue, L., and Blum, D. (2014). Beneficial effects eases:Present and future in clinical trials. Curr
of caffeine in a transgenic model of Alzheimers Top Med Chem 11, 10871101.
disease-like tau pathology. Neurobiol Aging. 35, Lorist, M.M., and Tops, M. (2003). Caffeine, fatigue,
20792090. and cognition. Brain Cogn 53,8294.
Lee, D.C., Rizer, J., Selenica, M.L., Reid, P., Kraft, C., Lourent, C., Leboucher, A., Troquier, L., Monnet, Y.,
Johnson, A., Blair, L., Gordon, M.N., Dickey, C.A., Burnouf, S., Figeac, M., Caillierez, R., Zommer,
and Morgan, D. (2010a). LPS- induced inflam- N., Demeyer, D., Hamdane, M., et al. (2011).
mation exacerbates phospho-tau pathology in Inflammatory processes in the THY-Tau22
rTg4510 mice. J Neuroinflammation7,56. model of AD-like Tau pathology. Paper pre-
Lee, S., Varvel, N.H., Konerth, M.E., Xu, G., Cardona, sented at the Society for Neuroscience meeting,
A.E., Ransohoff, R.M., and Lamb, B.T. (2010b). Washington,DC.
CX3CR1 deficiency alters microglial activation Lu, S.C., Huang, Z.Z., Yang, H., Mato, J.M., Avila,
and reduces beta-amyloid deposition in two M.A., and Tsukamoto, H. (2000). Changes
Alzheimers disease mouse models. Am J Pathol in methionine adenosyltransferase and
177, 25492562. S-adenosylmethionine homeostasis in alcoholic
Li, B., Carey, M., and Workman, J.L. (2007). The role rat liver. Am J Physiol Gastrointest Liver Physiol
of chromatin during transcription. Cell 128, 279, G178G185.
707719. Ma, D.K., Jang, M.H., Guo, J.U., Kitabatake, Y.,
Li, W., Dai, S., An, J., Li, P., Chen, X., Xiong, R., Liu, Chang, M.L., Pow-Anpongkul, N., Flavell,
P., Wang, H., Zhao, Y., Zhu, M., et al. (2008). R.A., Lu, B., Ming, G.L., and Song, H. (2009).
Chronic but not acute treatment with caffeine Neuronal activity-induced Gadd45b promotes
Adenosine and Alzheimers Disease 475

epigenetic DNA demethylation and adult neuro- Minkeviciene, R., Rheims, S., Dobszay, M.B.,
genesis. Science 323, 10741077. Zilberter, M., Hartikainen, J., Fulop, L., Penke,
Ma, D.K., Marchetto, M.C., Guo, J.U., Ming, G.L., B., Zilberter, Y., Harkany, T., Pitkanen, A., and
Gage, F.H., and Song, H (2010). Epigenetic cho- Tanila, H. (2009). Amyloid beta-induced neu-
reographers of neurogenesis in the adult mam- ronal hyperexcitability triggers progressive epi-
malian brain. Nat Neurosci 13, 13381344. lepsy. J Neurosci 29, 34533462.
Maia, L., and de Mendonca, A. (2002). Does caffeine Monoranu, C.M., Grunblatt, E., Bartl, J., Meyer,
intake protect from Alzheimers disease? Eur J A., Apfelbacher, M., Keller, D., Michel, T.M.,
Neurol 9, 377382. Al-Saraj, S., Schmitt, A., Falkai, P., etal. (2011).
Marambaud, P., Wen, P.H., Dutt, A., Shioi, J., Methyl- and acetyltransferases are stable epigen-
Takashima, A., Siman, R., and Robakis, N.K. etic markers postmortem. Cell Tissue Bank 12,
(2003). A CBP binding transcriptional repres- 289297.
sor produced by the PS1/epsilon-cleavage of Moreira, P.I., Santos, M.S., Oliveira, C.R., Shenk,
N-cadherin is inhibited by PS1 FAD mutations. J.C., Nunomura, A., Smith, M.A., Zhu, X., and
Cell 114, 635645. Perry, G. (2008). Alzheimer disease and the role
Marques, S., Batalha, V.L., Lopes, L.V., and Outeiro, T.F. of free radicals in the pathogenesis of the disease.
(2011). Modulating Alzheimers disease through CNS Neurol Disord Drug Targets 7,310.
caffeine: A putative link to epigenetics. J Morelli, M., Carta, A.R., and Jenner, P. (2009).
Alzheimers Dis 24 Suppl 2, 161171. Adenosine A2A receptors and Parkinsons dis-
Martin, A., and Fedio, P. (1983). Word production ease. Handb Exp Pharmacol. 193, 589615.
and comprehension in Alzheimers disease:The Morimoto, T., Sunagawa, Y., Kawamura, T., Takaya,
breakdown of semantic knowledge. Brain Lang T., Wada, H., Nagasawa, A., Komeda, M., Fujita,
19, 124141. M., Shimatsu, A., Kita, T., et al. (2008). The
Martin, G.M. (2005). Epigenetic drift in aging dietary compound curcumin inhibits p300 his-
identical twins. Proc Natl Acad Sci USA 102, tone acetyltransferase activity and prevents heart
1041310414. failure in rats. J Clin Invest 118, 868878.
Mastroeni, D., Grover, A., Delvaux, E., Whiteside, C., Morris, M., Maeda, S., Vossel, K., and Mucke,
Coleman, P.D., and Rogers, J. (2010). Epigenetic L. (2011). The many faces of tau. Neuron 70,
changes in Alzheimers disease: Decrements 410426.
in DNA methylation. Neurobiol Aging 31, Morris, M.S. (2003). Homocysteine and Alzheimers
20252037. disease. Lancet Neurol 2, 425428.
Mastroeni, D., Grover, A., Delvaux, E., Whiteside, C., Morrison, L.D., Smith, D.D., and Kish, S.J. (1996).
Coleman, P.D., and Rogers, J. (2011). Epigenetic Brain S-adenosylmethionine levels are severely
mechanisms in Alzheimers disease. Neurobiol decreased in Alzheimers disease. J Neurochem
Aging 32, 11611180. 67, 13281331.
Mastroeni, D., Mckee, A., Grover, A., Rogers, J., and Mozaz, M., Garaigordobil, M., Gonzalez Rothi, L.J.,
Coleman, P.D. (2009). Epigenetic differences Anderson, J., Crucian, G.P., and Heilman, K.M.
in cortical neurons from a pair of monozygotic (2006). Posture recognition in Alzheimers dis-
twins discordant for Alzheimers disease. PLoS ease. Brain Cogn 62, 241245.
One 4,e6617. Muller, C.E., and Jacobson, K.A. (2011). Recent
Matos, M., Augusto, E., Machado, N.J., dos developments in adenosine receptor ligands and
Santos-Rodrigues, A., Cunha, R.A., and Agostinho, their potential as novel drugs. Biochim Biophys
P. (2012). Astrocytic adenosine A2A receptors con- Acta 1808, 12901308.
trol the amyloid-beta peptide-induced decrease of Murdoch, B.E., Chenery, H.J., Wilks, V., and Boyle, R.S.
glutamate uptake. J Alzheimers Dis 31, 555567. (1987). Language disorders in dementia of the
Menendez, M. (2005). Down syndrome, Alzheimers Alzheimer type. Brain Lang 31, 122137.
disease and seizures. Brain Dev 27, 246252. Naert, G., and Rivest, S. (2011). CC chemokine recep-
Miller, C.A., and Sweatt, J.D. (2007). Covalent modi- tor 2 deficiency aggravates cognitive impair-
fication of DNA regulates memory formation. ments and amyloid pathology in a transgenic
Neuron 53, 857869. Erratum in: Neuron 59, mouse model of Alzheimers disease. J Neurosci
1051 (2008). 31, 62086220.
Min, S.W., Cho, S.H., Zhou, Y., Schroeder, S., Nagele, R.G., DAndrea, M.R., Lee, H., Venkataraman,
Haroutunian, V., Seeley, W.W., Huang, E.J., V., and Wang, H.Y. (2003). Astrocytes accumu-
Shen, Y., Masliah, E., Mukherjee, C., etal. (2010). late A beta 42 and give rise to astrocytic amyloid
Acetylation of tau inhibits its degradation and plaques in Alzheimer disease brains. Brain Res
contributes to tauopathy. Neuron 67, 953966. 971, 197209.
476 Part IV:Homeostatic Therapies

Naj, A.C., Jun, G., Beecham, G.W., Wang, L.S., Palop, J.J., and Mucke, L. (2009). Epilepsy and cog-
Vardarajan, B.N., Buros, J., Gallins, P.J., nitive impairments in Alzheimer disease. Arch
Buxbaum, J.D., Jarvik, G.P., Crane, P.K., et al. Neurol 66, 435440.
(2011). Common variants at MS4A4/MS4A6E, Pasinetti, G.M., and Eberstein, J.A. (2008). Metabolic
CD2AP, CD33 and EPHA1 are associated with syndrome and the role of dietary lifestyles
late-onset Alzheimers disease. Nat Genet. 43, in Alzheimers disease. J Neurochem 106,
436441. 15031514.
Narayan, P., and Dragunow, M. (2010). Pharmacology Perez, J.L., Carrero, I., Gonzalo, P., Arevalo-Serrano, J.,
of epigenetics in brain disorders. Br J Pharmacol Sanz-Anquela, J.M., Ortega, J., Rodriguez, M.,
159, 285303. and Gonzalo-Ruiz, A. (2010). Soluble oligomeric
Nagele, R.G., Wegiel, J., Venkataraman, V., Imaki, H., forms of beta-amyloid (Abeta) peptide stimu-
Wang, K.C., and Wegiel, J. (2004). Contribution late Abeta production via astrogliosis in the rat
of glial cells to the development of amyloid brain. Exp Neurol 223, 410421.
plaques in Alzheimers disease. Neurobiol Aging Pinna, A. (2009). Novel investigational adenosine
25, 663674. A2A receptor antagonists for Parkinsons dis-
Nee, L.E., and Lippa, C.F. (1999). Alzheimers disease ease. Expert Opin Investig Drugs. 18, 16191631.
in 22 twin pairs13-year follow-up:Hormonal, Polydoro, M., Acker, C.M., Duff, K., Castillo, P.E.,
infectious and traumatic factors. Dement Geriatr and Davies, P. (2009). Age-dependent impair-
Cogn Disord 10, 148151. ment of cognitive and synaptic function in the
Nelson, E.D., Kavalali, E.T., and Monteggia, L.M. htau mouse model of tau pathology. J Neurosci
(2008). Activity-dependent suppression of min- 29, 1074110749.
iature neurotransmission through the regulation Prasanthi, J.R., Dasari, B., Marwarha, G., Larson,
of DNA methylation. J Neurosci 28, 395406. T., Chen, X., Geiger, J.D., and Ghribi, O. (2010).
Nicolia, V., Fuso, A., Cavallaro, R.A., Di Luzio, A., Caffeine protects against oxidative stress and
and Scarpa, S. (2010). B vitamin deficiency pro- Alzheimers disease-like pathology in rabbit hip-
motes tau phosphorylation through regulation pocampus induced by cholesterol-enriched diet.
of GSK3beta and PP2A. J Alzheimers Dis 19, Free Radic Biol Med 49, 12121220.
895907. Rahman, A. (2009). The role of adenosine in
Nishizaki, T. (2004). ATP- and adenosine-mediated Alzheimers disease. Curr Neuropharmacol 7,
signaling in the central nervous sys- 207216.
tem: Adenosine stimulates glutamate release Rando, O.J. (2012). Combinatorial complexity in
from astrocytes via A2a adenosine receptors. J chromatin structure and function:Revisiting the
Pharmacol Sci 94, 100102. histone code. Curr Opin Genet Dev 22, 148155.
Nunomura, A., Perry, G., Pappolla, M.A., Friedland, Rebola, N., Rodrigues, R.J., Lopes, L.V.,
R.P., Hirai, K., Chiba, S., and Smith, M.A. (2000). Richardson, P.J., Oliveira, C.R., and Cunha,
Neuronal oxidative stress precedes amyloid-beta R.A. (2005). Adenosine A1 and A2A receptors
deposition in Down syndrome. J Neuropathol are co-expressed in pyramidal neurons and
Exp Neurol 59, 10111017. co-localized in glutamatergic nerve terminals of
Nygaard, T.G., Duvoisin, R.C., Manocha, M., and the rat hippocampus. Neuroscience 133,7983.
Chokroverty, S. (1989). Seizures in progressive Rebola, N., Sebastiao, A.M., de Mendonca, A.,
supranuclear palsy. Neurology 39, 138140. Oliveira, C.R., Ribeiro, J.A., and Cunha, R.A.
Ono, K., Hasegawa, K., Naiki, H., and Yamada, M. (2003). Enhanced adenosine A2A receptor facili-
(2004). Curcumin has potent anti-amyloidogenic tation of synaptic transmission in the hippocam-
effects for Alzheimers -amyloid fibrils in vitro. pus of aged rats. J Neurophysiol 90, 12951303.
J Neurosci Res 75, 742750. Rebola, N., Simoes, A.P., Canas, P.M., Tome, A.R.,
Orr, A.G., Orr, A.L., Li, X.J., Gross, R.E., and Andrade, G.M., Barry, C.E., Agostinho, P.M.,
Traynelis, S.F. (2009). Adenosine A(2A) recep- Lynch, M.A., and Cunha, R.A. (2011). Adenosine
tor mediates microglial process retraction. Nat A2A receptors control neuroinflammation and
Neurosci 12, 872878. consequent hippocampal neuronal dysfunction.
Palop, J.J., Chin, J., Roberson, E.D., Wang, J., Thwin, J Neurochem 117, 100111.
M.T., Bien-Ly, N., Yoo, J., Ho, K.O., Yu, G.Q., Reitz, C., Brayne, C., and Mayeux, R. (2011).
Kreitzer, A., et al. (2007). Aberrant excitatory Epidemiology of Alzheimer disease. Nature Rev
neuronal activity and compensatory remodel- Neurol 7, 137152.
ing of inhibitory hippocampal circuits in mouse Ribeiro, J.A., and Sebastiao, A.M. (2010). Modulation
models of Alzheimers disease. Neuron 55, and metamodulation of synapses by adenosine.
697711. Acta Physiol (Oxf) 199, 161169.
Adenosine and Alzheimers Disease 477

Ricobaraza, A., Cuadrado-Tejedor, M., and and Nakazato, Y. (2008). Microglial activation
Garcia-Osta, A. (2011). Long-term phenylbutyr- in brain lesions with tau deposits: Comparison
ate administration prevents memory deficits in of human tauopathies and tau transgenic mice
Tg2576 mice by decreasing Abeta. Front Biosci TgTauP301L. Brain Res 1214, 159168.
(Elite Ed) 3, 13751384. Saura, C.A., Choi, S.Y., Beglopoulos, V., Malkani, S.,
Ricobaraza, A., Cuadrado-Tejedor, M., Marco, S., Zhang, D., Shankaranarayana Rao, B.S.,
Prez-Otao, I., and Garca-Osta, A. (2012). Chattarji, S., Kelleher, R.J., 3rd, Kandel, E.R.,
Phenylbutyrate rescues dendritic spine loss asso- Duff, K., etal. (2004). Loss of presenilin function
ciated with memory deficits in a mouse model of causes impairments of memory and synaptic
Alzheimer disease. Hippocampus 22, 10401050. plasticity followed by age-dependent neurode-
Ricobaraza, A., Cuadrado-Tejedor, M., generation. Neuron 42,2336.
Prez-Mediavilla, A., Frechilla, D., Del Ro, J., Saura, J., Angulo, E., Ejarque, A., Casado, V., Tusell,
and Garca-Osta, A. (2009). Phenylbutyrate ame- J.M., Moratalla, R., Chen, J.F., Schwarzschild,
liorates cognitive deficit and reduces tau pathol- M.A., Lluis, C., Franco, R., et al. (2005).
ogy in an Alzheimers disease mouse model. Adenosine A2A receptor stimulation potenti-
Neuropsychopharmacology 34, 17211732. ates nitric oxide release by activated microglia. J
Ritchie, K., Artero, S., and Touchon, J. (2001). Neurochem 95, 919929.
Classification criteria for mild cognitive impair- Sawyer, D.A., Julia, H.L., and Turin, A.C. (1982).
ment: A population-based validation study. Caffeine and human behavior:Arousal, anxiety,
Neurology 56,3742. and performance effects. J Behav Med 5, 415439.
Ritchie, K., Carriere, I., de Mendonca, A., Portet, F., Schindowski, K., Bretteville, A., Leroy, K., Begard,
Dartigues, J.F., Rouaud, O., Barberger-Gateau, P., S., Brion, J.P., Hamdane, M., and Buee, L. (2006).
and Ancelin, M.L. (2007). The neuroprotec- Alzheimers disease-like tau neuropathology
tive effects of caffeine: A prospective popula- leads to memory deficits and loss of functional
tion study (the Three City Study). Neurology 69, synapses in a novel mutated tau transgenic
536545. mouse without any motor deficits. Am J Pathol
Roberson, E.D., Halabisky, B., Yoo, J.W., Yao, J., Chin, 169, 599616.
J., Yan, F., Wu, T., Hamto, P., Devidze, N., Yu, Schneider, A., Chatterjee, S., Bousiges, O., Selvi,
G.Q., et al. (2011). Amyloid-beta/Fyn-induced B.R., Swaminathan, A., Cassel, R., Blanc, F.,
synaptic, network, and cognitive impairments Kundu, T.K., and Boutillier, A.L. (2013).
depend on tau levels in multiple mouse models Acetyltransferases (HATs) as targets for neu-
of Alzheimers disease. J Neurosci 31, 700711. rological therapeutics. Neurotherapeutics 10,
Roberson, E.D., Scearce-Levie, K., Palop, J.J., Yan, F., 568588.
Cheng, I.H., Wu, T., Gerstein, H., Yu, G.Q., and Sebastiao, A.M., and Ribeiro, J.A. (2000).
Mucke, L. (2007). Reducing endogenous tau Fine-tuning neuromodulation by adenosine.
ameliorates amyloid beta-induced deficits in an Trends Pharmacol Sci 21, 341346.
Alzheimers disease mouse model. Science 316, Sebastiao, A.M., and Ribeiro, J.A. (2009). Tuning and
750754. fine-tuning of synapses with adenosine. Curr
Ross, G.W., and Petrovitch, H. (2001). Current evi- Neuropharmacol 7, 180194.
dence for neuroprotective effects of nicotine Sergeant, N., Bretteville, A., Hamdane, M.,
and caffeine against Parkinsons disease. Drugs Caillet-Boudin, M.L., Grognet, P., Bombois,
Aging 18, 797806. S., Blum, D., Delacourte, A., Pasquier, F.,
Rouaux, C., Jokic, N., Mbebi, C., Boutillier, S., Vanmechelen, E., et al. (2008). Biochemistry of
Loeffler, J.P., and Boutillier, A.L. (2003). Critical Tau in Alzheimers disease and related neurologi-
loss of CBP/p300 histone acetylase activity by cal disorders. Expert Rev Proteomics 5, 207224.
caspase-6 during neurodegeneration. EMBO J Serot, J.M., Christmann, D., Dubost, T., Bene, M.C.,
22, 65376549. and Faure, G.C. (2001). CSF-folate levels are
Sanchez, P.E., Zhu, L., Verret, L., Vossel, K.A., Orr, decreased in late-onset AD patients. J Neural
A.G., Cirrito, J.R., Devidze, N., Ho, K., Yu, G.Q., Transm 108,9399.
Palop, J.J., et al. (2012). Levetiracetam suppresses Seshadri, S., Beiser, A., Selhub, J., Jacques, P.F.,
neuronal network dysfunction and reverses synap- Rosenberg, I.H., DAgostino, R.B., Wilson, P.W.,
tic and cognitive deficits in an Alzheimers disease and Wolf, P.A. (2002). Plasma homocysteine as a
model. Proc Natl Acad Sci USA 109, E2895E2903. risk factor for dementia and Alzheimers disease.
Sasaki, A., Kawarabayashi, T., Murakami, T., New Engl J Med 346, 476483.
Matsubara, E., Ikeda, M., Hagiwara, H., Shen, H.Y., Lusardi, T.A., Williams-Karnesky, R.L.,
Westaway, D., George-Hyslop, P.S., Shoji, M., Lan, J.Q., Poulsen, D.J., and Boison, D. (2011).
478 Part IV:Homeostatic Therapies

Adenosine kinase determines the degree of brain Tahiliani, M., Koh, K.P., Shen, Y., Pastor, W.A.,
injury after ischemic stroke in mice. J Cereb Bandukwala, H., Brudno, Y., Agarwal, S.,
Blood Flow Metab 31, 16481659. Iyer, L.M., Liu, D.R., Aravind, L., et al.
Sheng, J.G., Mrak, R.E., and Griffin, W.S. (1994). (2009). Conversion of 5-methylcytosine to
S100 beta protein expression in Alzheimer dis- 5-hydroxymethylcytosine in mammalian DNA
ease: Potential role in the pathogenesis of neu- by MLL partner TET1. Science 324, 930935.
ritic plaques. J Neurosci Res 39, 398404. Takahashi, R.N., Pamplona, F.A., and Prediger, R.D.
Simard, A.R., Soulet, D., Gowing, G., Julien, J.P., and (2008). Adenosine receptor antagonists for cog-
Rivest, S. (2006). Bone marrow-derived microglia nitive dysfunction: A review of animal studies.
play a critical role in restricting senile plaque forma- Front Biosci 13, 26142632.
tion in Alzheimers disease. Neuron 49, 489502. Tchantchou, F., Graves, M., Ortiz, D., Chan, A., Rogers, E.,
Simola, N., Morelli, M., and Pinna, A. (2008). and Shea, T.B. (2006). S-adenosyl methio-
Adenosine A2A receptor antagonists and nine:Aconnection between nutritional and genetic
Parkinsons disease: state of the art and future risk factors for neurodegeneration in Alzheimers
directions. Curr Pharm Des. 14, 14751489. disease. J Nutr Health Aging 10, 541544.
Simonin, C., Duru, C., Salleron, J., Hincker, P., Tohgi, H., Utsugisawa, K., Nagane, Y., Yoshimura,
Charles, P., Delval, A., Youssov, K., Burnouf, S., M., Genda, Y., and Ukitsu, M. (1999). Reduction
Azulay, J.P., Verny, C., etal. (2013). Association with age in methylcytosine in the promoter
between caffeine intake and age at onset in region -224 approximately -101 of the amyloid
Huntingtons disease. Neurobiol Dis 58, 179182. precursor protein gene in autopsy human cortex.
Smit, H.J., and Rogers, P.J. (2000). Effects of low doses Brain Res Mol Brain Res 70, 288292.
of caffeine on cognitive performance, mood and Thomas, R.J. (1997). Seizures and epilepsy in the
thirst in low and higher caffeine consumers. elderly. Arch Intern Med 157, 605617.
Psychopharmacology (Berl) 152, 167173. Trinh, K., Andrews, L., Krause, J., Hanak, T., Lee, D.,
Smith, A.D. (2008). The worldwide challenge of the Gelb, M., and Pallanck, L. (2010). Decaffeinated
dementias:Arole for B vitamins and homocyste- coffee and nicotine-free tobacco provide neuro-
ine? Food Nutr Bull 29, S143S172. protection in drosophila models of Parkinsons
Soreghan, B.A., Lu, B.W., Thomas, S.N., Duff, K., disease through an NRF2-dependent mecha-
Rakhmatulin, E.A., Nikolskaya, T., Chen, T., and nism. J Neurosci 30, 55255532.
Yang, A.J. (2005). Using proteomics and network Valor, L.M., Viosca, J., Lopez-Atalaya, J.P., and
analysis to elucidate the consequences of synap- Barco, A. (2013). Lysine acetyltransferases CBP
tic protein oxidation in a PS1 + AbetaPP mouse and p300 as therapeutic targets in cognitive and
model of Alzheimers disease. J Alzheimers Dis neurodegenerative disorders. Curr Pharma Des
8, 227241. 19, 50515064.
Stone, T.W., Ceruti, S., and Abbracchio, M.P. (2009). Van der Jeugd, A., Ahmed, T., Burnouf, S.,
Adenosine receptors and neurological dis- Belarbi, K., Hamdame, M., Grosjean, M.E.,
ease: Neuroprotection and neurodegeneration. Humez, S., Balschun, D., Blum, D., Buee, L.,
Handb Exp Pharmacol 193, 535587. et al. (2011). Hippocampal tauopathy in tau
Stoppelkamp, S., Bell, H.S., Palacios-Filardo, J., Shewan, transgenic mice coincides with impaired
D.A., Riedel, G., and Platt, B. (2011). In vitro mod- hippocampus-dependent learning and memory,
elling of Alzheimers disease: Degeneration and and attenuated late-phase long-term depression
cell death induced by viral delivery of amyloid and of synaptic transmission. Neurobiol Learn Mem
tau. Exp Neurol 229, 226237. 95, 296304.
Sultan, A., Nesslany, F., Violet, M., Begard, S., Van Eldik, L.J., and Griffin, W.S. (1994). S100 beta
Loyens, A., Talahari, S., Mansuroglu, Z., Marzin, expression in Alzheimers disease: Relation
D., Sergeant, N., Humez, S., etal. (2011). Nuclear to neuropathology in brain regions. Biochim
tau, a key player in neuronal DNA protection. J Biophys Acta 1223, 398403.
Biol Chem 286, 45664575. van Gelder, B.M., Buijsse, B., Tijhuis, M., Kalmijn, S.,
Sung, Y.M., Lee, T., Yoon, H., DiBattista, A.M., Song, Giampaoli, S., Nissinen, A., and Kromhout, D.
J., Sohn, Y., Moffat, E., Turner, R.S., Jung, M., (2007). Coffee consumption is inversely associ-
Kim, J., et al. (2013). Mercaptoacetamide-based ated with cognitive decline in elderly European
class II HDAC inhibitor lowers Ab levels and men:The FINE Study. Eur J Clin Nutr 61, 226232.
improves learning and memory in a mouse Wang, S.C., Oelze, B., and Schumacher, A. (2008).
model of Alzheimers disease. Exp Neurol 239, Age-specific epigenetic drift in late-onset
192201. Alzheimers disease. PLoS One 3,e2698.
Adenosine and Alzheimers Disease 479

Wegiel, J., Imaki, H., Wang, K.C., Wronska, A., Wyss-Coray, T., Lin, C., Yan, F., Yu, G.Q., Rohde,
Osuchowski, M., and Rubenstein, R. (2003). M., McConlogue, L., Masliah, E., and Mucke,
Origin and turnover of microglial cells in fibril- L. (2001). TGF-beta1 promotes microglial
lar plaques of APPsw transgenic mice. Acta amyloid-beta clearance and reduces plaque bur-
Neuropathol 105, 393402. den in transgenic mice. Nat Med 7, 612618.
Wei, C.J., Li, W., and Chen, J.F. (2011). Normal and Yang, F., Lim, G.P., Begum, A.N., Ubeda, O.J.,
abnormal functions of adenosine receptors in the Simmons, M.R., Ambegaokar, S.S., Chen, P.P.,
central nervous system revealed by genetic knock- Kayed, R., Glabe, C.G., Frautschy, S.A., et al.
out studies. Biochim Biophys Acta 1808, 13581379. (2005). Curcumin inhibits formation of amy-
West, R.L., Lee, J.M., and Maroun, L.E. (1995). loid beta oligomers and fibrils, binds plaques,
Hypomethylation of the amyloid precursor pro- and reduces amyloid in vivo. J Biol Chem 280,
tein gene in the brain of an Alzheimers disease 58925901.
patient. J Mol Neurosci 6, 141146. Yasojima, K., Akiyama, H., McGeer, E.G., and
Westerman, M.A., Cooper-Blacketer, D., Mariash, A., McGeer, P.L. (2001). Reduced neprilysin in high
Kotilinek, L., Kawarabayashi, T., Younkin, L.H., plaque areas of Alzheimer brain:Apossible rela-
Carlson, G.A., Younkin, S.G., and Ashe, K.H. tionship to deficient degradation of beta-amyloid
(2002). The relationship between Abeta and mem- peptide. Neurosci Lett 297, 97100.
ory in the Tg2576 mouse model of Alzheimers Yee, B.K., Singer, P., Chen, J.F., Feldon, J., and Boison,
disease. J Neurosci 22, 18581867. D. (2007). Transgenic overexpression of adenos-
Wharton, S.B., OCallaghan, J.P., Savva, G.M., Nicoll, ine kinase in brain leads to multiple learning
J.A., Matthews, F., Simpson, J.E., Forster, G., impairments and altered sensitivity to psycho-
Shaw, P.J., Brayne, C., and Ince, P.G. (2009). mimetic drugs. Eur J Neurosci 26, 32373252.
Population variation in glial fibrillary acidic Yoshikai, S., Sasaki, H., Doh-ura, K., Furuya, H.,
protein levels in brain ageing: Relationship and Sakaki, Y. (1990). Genomic organization of
to Alzheimer-type pathology and dementia. the human amyloid beta-protein precursor gene.
Dement Geriatr Cogn Disord 27, 465473. Gene 87, 257263.
Williams-Karnesky, R.L., Sandau, U.S., Lusardi, T.A., Yoshiyama, Y., Higuchi, M., Zhang, B., Huang, S.M.,
Lytle, N.K., Farrell, J.M., Pritchard, E.M., Kaplan, Iwata, N., Saido, T.C., Maeda, J., Suhara, T.,
D.L., and Boison, D. (2013). Epigenetic changes Trojanowski, J.Q., and Lee, V.M. (2007). Synapse
induced by adenosine augmentation therapy pre- loss and microglial activation precede tangles
vent epileptogenesis. J Clin Invest 123, 35523563. in a P301S tauopathy mouse model. Neuron 53,
Wilson, V.L., and Jones, P.A. (1983). DNA methyla- 337351.
tion decreases in aging but not in immortal cells. Yu, L., Shen, H.Y., Coelho, J.E., Araujo, I.M., Huang,
Science 220, 10551057. Q.Y., Day, Y.J., Rebola, N., Canas, P.M., Rapp,
Wilson, V.L., Smith, R.A., Ma, S., and Cutler, E.K., Ferrara, J., et al. (2008). Adenosine A2A
R.G. (1987). Genomic 5-methyldeoxycytidine receptor antagonists exert motor and neuropro-
decreases with age. J Biol Chem 262, 99489951. tective effects by distinct cellular mechanisms.
Wisniewski, H.M., and Wegiel, J. (1991). Spatial rela- Ann Neurol 63, 338346.
tionships between astrocytes and classical plaque Zampieri, M., Guastafierro, T., Calabrese, R.,
components. Neurobiol Aging 12, 593600. Ciccarone, F., Bacalini, M.G., Reale, A., Perilli, M.,
Wittchen, H.U., and Jacobi, F. (2005). Size and Passananti, C., and Caiafa, P. (2012). ADP-ribose
burden of mental disorders in Europea criti- polymers localized on Ctcf-Parp1-Dnmt1 com-
cal review and appraisal of 27 studies. Eur plex prevent methylation of Ctcf target sites.
Neuropsychopharmacol 15, 357376. Biochem J 441, 645652.
Wu, J., Basha, M.R., Brock, B., Cox, D.P., Zhang, Z., and Schluesener, Y.H.J. (2013). Oral
Cardozo-Pelaez, F., McPherson, C.A., Harry, J., administration of histone deacetylase inhibi-
Rice, D.C., Maloney, B., Chen, D., et al. (2008). tor MS-275 ameliorates neuroinflammation and
Alzheimers disease (AD)-like pathology in aged cerebral amyloidosis and improves behavior in
monkeys after infantile exposure to environ- a mouse model. J Neuropathol Exp Neurol 72,
mental metal lead (Pb):Evidence for a develop- 178185.
mental origin and environmental link for AD. J Zhou, S.J., Zhu, M.E., Shu, D., Du, X.P., Song, X.H.,
Neurosci 28,39. Wang, X.T., Zheng, R.Y., Cai, X.H., Chen, J.F.,
Wyss-Coray, T. (2006). Inflammation in Alzheimer and He, J.C. (2009). Preferential enhancement
disease: Driving force, bystander or beneficial of working memory in mice lacking adenosine
response? Nat Med 12, 10051015. A(2A) receptors. Brain Res 1303,7483.
480 Part IV:Homeostatic Therapies

Zilka, N., Stozicka, Z., Kovac, A., Pilipcinec, E., Bugos, Zotova, E., Bharambe, V., Cheaveau, M., Morgan,
O., and Novak, M. (2009). Human misfolded W., Holmes, C., Harris, S., Neal, J.W., Love, S.,
truncated tau protein promotes activation of Nicoll, J.A., and Boche, D. (2013). Inflammatory
microglia and leukocyte infiltration in the trans- components in human Alzheimers disease and
genic rat model of tauopathy. J Neuroimmunol after active amyloid-42 immunization. Brain.
209,1625. 136, 26772696.
26
Brain Homeostasis and Parkinsons Disease
DEEP TILALL

PA R K I N S O N S D I S E A S E : gives these neurons their distinct color. Loss of


AN OVERVIEW these neurons produces the classic gross neu-
Parkinsons disease (PD) is a chronic, progres- ropathological finding of SNpc depigmenta-
sive, neurodegenerative disorder of the basal tion, highly characteristic of PD. The pattern
ganglia. In its worldwide prevalence it is second of the SNpc neuronal cell loss also parallels the
only to Alzheimers disease, affecting 5 million expression of dopamine transporter (Uhl etal.,
people globally, and its occurrence is expected to 1994)and is in line with the findings that deple-
markedly increase in the coming decades due to tion of dopamine (DA) is most pronounced in
an increase in lifespan of the population (Olanow the dorsolateral putamen (Bernheimer et al.,
and Schapira, 2013). As with Alzheimers, the 1973), which is the main site for the projections
principal risk factor for acquiring PD increases of these neurons. At the onset of PD symptoms,
with age with incidence rates rising exponen- approximately 80% of the putamental DA is
tially after 50 years of age (Bower et al., 2000; depleted and approximately 60% of the SNpc
Driver etal., 2009). The disorder does not show DAergic neurons arelost.
any geographical and/or gender-specific perva- The PD-associated loss of DAergic neu-
siveness; it is a common disabling disorder and is rons displays a characteristic pattern that is
often seen in people from all races and geograph- highly distinct and different from neuronal
ical locations with clinical signs emerging also in loss in normal aging. Fearnley and Lees (1991)
the young (Lees etal., 2009; Massano and Bhatia, reported that while in normal aging the dor-
2012; Tolosa etal.,2006). somedial part of the SNpc is affected, in PD
the cell loss is more concentrated in the ven-
PAT H O P H Y S I O L O G Y trolateral and caudal regions. Also, the stria-
AND NEUROCHEMICAL tum terminal loss is more pronounced than
F E AT U R E S O F P D the magnitude of SNpc DAergic neuron loss
PD results primarily from the loss of the nigros- (Bernheimer etal.,1973).
triatal dopaminergic (DAergic) neurons in the Apart from neuronal cell loss, PD patients
substantia nigra pars compacta (SNpc), although also show formation of proteinaceous intraneu-
additional neuronal systems such as catechola- ronal or intraglial inclusions, known as Lewy
minergic, serotonergic, and brainstem nuclei are bodies (LBs) composed of a dense core of filamen-
also affected, albeit less severely. Atime-course tous material enshrouded by filaments of 10 to
analysis of the pathological process in PD 20 nm in diameter (Goedert, 2001a, 2001b). LBs
showed that the neurodegeneration reaches are found in all affected regions, have an organ-
the dorsal motor nuclei of the hypoglossus and ized structure of a dense hyaline core surrounded
vagus nerves and raphe nuclei, as well as the by a clear halo, and contain cytoplasmic aggre-
anterior olfactory nucleus and olfactory bulb, gates composed of numerous proteins including
before it extends to the mesencephalic areas, -synuclein, parkin, ubiquitin, and neurofila-
which includes the substantia nigra (SN) where ments (Forno, 1996; Spillantini etal., 1998). The
a majority of the cell damage takes place (Braak influences that lead to this neuronal cell loss and
etal., 2003). The cell bodies of the nigrostriatal the formation of LBs is still unknown. However,
neurons reside in the SNpc and project primar- a common perception points toward complex
ily to the putamen. These neurons contain the interactions between genetic (nonsporadic) and
pigment neuromelanin (Marsden, 1983), which environmental (sporadic)cues.
482 Part IV:Homeostatic Therapies

CLINICAL SYMPTOMS OF PD several monogenic (a form of disease for which


PD is characterized by four cardinal motor fea- a mutation in a single gene is sufficient to cause
tures:bradykinesia (slowness of movements with the disease) forms of the disorder and several risk
a progressive loss of amplitude or speed during factors that increase susceptibility to PD. They
alternating rapid body movements; Jankovic, collectively account for about 30% of the famil-
2008; Marsden, 1982; Rodriguez-Oroz et al., ial and 3% to 5% of the sporadic cases (Klein and
2009), rest tremor, rigidity, and postural and Westenberger, 2012). Today we know of about 18
gait impairment. Numerous non-motor symp- distinct chromosomal regions that are more or
toms are also associated with PD. These include less convincingly related to PD. Only six of these
neuropsychiatric features such as anxiety, panic specific regions contain genes with mutations
attacks, depression, apathy, dysautonomia (i.e., that conclusively cause monogenicPD.
constipation, urinary dysfunction, excessive Chromosomal locations associated with PD
sweating), sleep disorders (e.g., insomnia, rest- are designated as PARK, and they are numbered
less leg syndrome, excessive daytime sleepiness), in chronological order of their identification
sensory dysfunction (e.g., loss of smell, decreased (PARK1, PARK2, PARK3, and so on; Klein and
color and contrast discrimination), pain, and Westenberger, 2012). All of the currently known
fatigue. Since PD is a progressive neurological monogenic PD forms are autosomal, either dom-
disease, these non-motor features may be present inant or recessive. In autosomal-dominant disor-
before presentation of any classical motor symp- ders, one mutated allele of the gene is enough to
tom phenotype and therefore present a poten- cause the disease, whereas in autosomal-recessive
tial utility for diagnosis and determination of disorders two mutations (the samehomozygous
disease-progressionstage. or differentcompound heterozygous), one on
each gene copy (allele), are necessary to cause the
ETIOLOGYOFPD phenotype. Mutations in SNCA (PARK1/4) and
PD is a multifactorial disorder caused by a LRRK2 (leucine-rich repeat kinase 2 or darda-
combination of genes and environmental fac- rin,/PARK8), VPS35 (vacuolar protein sorting
tors. Until 1997, the notion that heritability has 35), EIF4G1 (eukaryotic translation initiation
a role in PD was contentious; indeed, PD was factor 4 gamma 1), and GBA (glucocerebrosi-
considered to be the archetypal nongenetic dase) are responsible for autosomal-dominant
disorder (Farrer, 2006). However, in the past PD forms, and mutations in parkin (PARK2),
decade, multiple mutations in genes have been PTEN-induced putative kinase 1 (PINK1/PARK6),
described in families with a Mendelian pattern DJ-1 (PARK7), ATP13A2 (PARK9), FBXO7
of PD inheritance (Bonifati et al., 2003; Kitada (F-box only protein 7 gene), and PLA2G6 (phos-
etal., 1998; Leroy etal., 1998a; Leroy etal., 1998b; pholipase A2, group VI) are accountable for PD
Paisan-Ruiz et al., 2004; Polymeropoulos et al., that displays an autosomal recessive mode of
1997; Singleton etal., 2003; Valente etal., 2004a; inheritance.
Zimprich etal., 2004). Genome-wide association
as well as linkage mapping studies have identified AUTOSOM AL DOMINANT
many causal mutations and candidate genes as FORMSOFPD
well as multiple susceptibility variants that might
trigger PD (Maraganore etal.,2005). SNCA (PARK1/PARK4)
SNCA was the first pathogenic PD gene dis-
GENETIC FORMSOFPD covered that had a missense mutation. SNCA
PD was long considered a sporadic disorder, and codes for a protein called -synuclein, which is
the hypothesis that heredity underlies the etiol- expressed throughout the brain and is involved
ogy of PD traditionally found little support from in learning, synaptic plasticity, vesicle dynam-
epidemiological data. The majority of PD cases ics, and DA synthesis (Lotharius and Brundin,
are still sporadic: only about 10% (Thomas and 2002; Sidhu et al., 2004). The SNCA gene has
Beal, 2007) of patients report a positive family six exons encoding for -synuclein. Natively
history. Aseminal study by Leroux in 1880 first unfolded -synuclein has no secondary structure
recognized familial aggregation of parkinson- and is nontoxic. Once bound to the phospholipid
ism and suggested that heritable factors might membrane, -synuclein forms the helical struc-
increase disease susceptibility. The past decade ture (Giasson et al., 2001). The point mutants
of research in PD has led to the discovery of of SNCA tend to form stable sheets and thus
Brain Homeostasis and Parkinsons Disease 483

exacerbate the formation of toxic oligomers, dementia and cognitive decline and sometimes
protofibrils, and fibrils. Therefore, it is believed with atypical features such as central hypoven-
that the missense SNCA mutations cause PD tilation and myoclonus. LBs are present and
through a toxic gain of function (Bertoncini spread through the SN, locus ceruleus, hypo-
etal., 2005), and LBs may represent the attempt thalamus, and cerebral cortex (Polymeropoulos
to purge the cell of toxic damaged -synuclein etal.,1996).
(Chen and Feany, 2005). Transgenic overexpres-
sion of -synuclein causes neurotoxicity with LRRK2 (PARK8)
aggregation and accumulation of -synuclein. Mutations in LRRK2 are the most common and
In vivo, mice demonstrate various neuropatho- known cause of autosomal dominant PD with a
logical changes, including neuronal atrophy, frequency ranging from 2% to 40% in different
dystrophic neurites, and astrogliosis, accompa- populations with the highest occurrence among
nied by -synuclein-positive LB-like inclusions North Africans and Jewish individuals (Brice,
(Fernagut and Chesselet, 2004). Similarly, viral 2005; Lesage etal., 2006; Ozelius etal., 2006). The
delivery of -synuclein within the SN of adult LRRK2 gene encodes for a protein called darda-
rats results in marked loss of DA neurons, dys- rin with two enzymatic domains (GTPase and
trophic changes, and aggregated -synuclein kinase) and multiple proteinprotein interaction
pathology (Yamada et al., 2004). In human PD domains. One segment of the dardarin protein is
cases, -synuclein can also be phosphorylated called a leucine-rich region because it contains a
and found within LB inclusions (Fujiwara etal., large amount of a amino acid known as leucine.
2002; Nonaka etal., 2005). However, it is a mat- Proteins with leucine-rich regions appear to
ter of debate whether the phosphorylated mon- play a role in activities that require interactions
omer disrupts the amphipathic association of with other proteins, such as transmitting signals
-synuclein with lipids or is especially toxic if not or helping to assemble the cytoskeleton. Other
sequestered and degraded. parts of the dardarin protein are also thought to
PARK1/PARK4 contributes to SNCA muta- be involved in proteinprotein interactions. The
tions (missense) and duplications/triplications, LRRK2 gene is active in the brain and other tis-
respectively. The SNCA 209G>A (Ala53Thr) sues throughout thebody.
mutation was identified through genome-wide Seven mutations (Asn1437His, Arg1441Cys,
linkage studies in several families with autoso- Arg1441Gly, Arg1441His, Tyr1699Cys, Gly
mal dominant parkinsonism (Athanassiadou 2019Ser, and Ile2020Thr) are known to cause
et al., 1999; Bostantjopoulou et al., 2001; Choi PD. Patients respond favorably to levo-
etal., 2008; Ki etal., 2007; Polymeropoulos etal., dopa therapy, and dementia is not common.
1997; Puschmann etal., 2009; Spira etal., 2001), Neuropathological findings are mostly inconsist-
and two further missense mutations 88G>C ent, showing both LB (and sometimes tau- and
(Ala30Pro; Kruger et al., 1998) and 188G>A ubiquitin-containing inclusions) pathology and
(Glu46Lys; Zarranz et al., 2004) were subse- pure nigral degeneration without LBs, with or
quently identified. Seventeen duplications of the without neurofibrillary tangles. The pathogenic
entire coding region of SNCA have been reported mechanism leading to PD caused by LRRK2
to date, 13 in PD families and 4 in sporadic cases, mutations is still uncertain. Since LRRK2 is a
one in which it was shown that the mutation big protein interacting with so many proteins,
arose de novo (Ahn et al., 2008; Brueggemann any changes in these interactions due to muta-
et al., 2008; Chartier-Harlin et al., 2004; Fuchs tions can have profound effects on its functions
etal., 2009; Fuchs etal., 2007; Ibanez etal., 2004; and thereby can change the susceptibility to the
Ibanez etal., 2009; Ikeuchi etal., 2008; Nishioka disease.
etal., 2006; Troiano etal., 2008; Uchiyama etal.,
2008). Triplications of the SNCA gene were found VPS35
in three independent families (Farrer etal., 2004; VPS35 is a protein involved in retrograde
Ibanez etal., 2009; Singleton etal.,2003). transport of proteins from endosomes to the
Patients with SNCA mutations usually have trans-Golgi network. Recently the p.D620N
early-onset PD (age of onset 50years) with an mutation in VPS35 was discovered as a new cause
initially good response to levodopa treatment of PD in two independent exome sequencing
to increase DA levels. However, the disease has studies on Swiss (Vilarino-Guell etal., 2011)and
a rapid progression and often presents with Austrian families (Zimprich et al., 2011).
484 Part IV:Homeostatic Therapies

Frequency of this mutation carrier is low and has 21years). Pathology underlying PARK2-related
been estimated to represent about only 0.1% of PD does not generally show LB pathology.
the PD population (Kumar etal., 2012). Patients However, thepatients do have substantia nigral
afflicted with this mutation develop classical cell loss and gliosis. Marked cognitive or vegeta-
late-onset, levodopa-responsive Parkinsonism tive disturbances are rare. PD patients suffering
reminiscent of sporadic PD, albeit with a slightly from this mutation show excellent and sustained
earlier age ofonset. response to levodopa (Lohmann etal.,2003).

EIF4G1 PINK1 (PARK6)


EIF4G1 is a eukaryotic translation initiator fac- PINK1 is a mitochondrial serine/threonine-
tor. The p.R1205H mutation in the EIF4G1 was protein kinase encoded by the PINK1 gene and
discovered in a French family inheriting famil- is believed to protect cells from stress-induced
ial parkinsonism (Chartier-Harlin etal., 2011). It mitochondrial dysfunction. Mutation in the
is dominantly inherited and has a very low fre- PINK1 protein is responsible for causing the
quency of 0.02% to 0.2% in the PD population second most common form of autosomal reces-
(Tucci etal.,2012). sive PD. Frequency of PINK1 mutations is in the
range of 1% to 9% with considerable variation
GBA across different ethnic groups (Bonifati et al.,
GBA is an enzyme with glucosylceramidase 2005; Healy et al., 2004; Klein et al., 2005; Li
activity that is needed to cleave, by hydroly- et al., 2005; Rogaeva et al., 2004; Valente et al.,
sis, the beta-glucosidic linkage of the chemical 2004b). It is characterized by early onset in most
glucocerebroside, an intermediate in glycolipid patients (mean age of onset = 30 years) with
metabolism. It is localized in the lysosome. symptoms ranging from dystonia to hyperre-
Heterozygous mutations in this gene are consid- flexia. Psychiatric disturbances such as depres-
ered to be a major risk factor for PD (Sidransky sion, anxiety, and psychosis are also found
etal., 2009). The mutations in the GBA gene deter- frequently. However, patients respond excellently
mined to date are Asn370Ser and Leu444Pro. to Levopoda treatment.
The Asn370Ser mutation is commonly found
in Ashkenazi Jews with a carrier frequency of DJ-1 (PARK7)
5% (Lwin et al., 2004). However, the mutations The third gene associated with autosomal reces-
have a much lower penetrance than the classical sive PD is mutated in about 1% to 2% of patients
Mendelian mutations. Patients with GBA patho- (Pankratz etal., 2006). This seems to be a rare dis-
genic mutations have typical PD with possibly a order with a clinical phenotype similar to PARK6.
slightly earlier onsetage. PARK7 gene encodes for the protein DJ-1, which
functions as a redox-sensitive chaperone and as a
AUTOSOM AL R ECESSIV E sensor for oxidative stress, and protects neurons
FORMSOFPD against oxidative stress and cell death. About 10
Homozygous or compound heterozygous muta- different point mutations and exonic deletions
tions in each of five genesparkin (PRKN/ have been described for this gene, mostly in the
PARK2), PTEN induced putative kinase 1 homozygous or compound-heterozygous state.
(PINK1/PARK6), Parkinson protein 7 (DJ-1/ Given that the mutations are so uncommon,
PARK7), F box only protein 7 gene (FBX07/ there is not enough data to draw any conclusion
PARK15), and Phospholipase A2 (PLA2G6)can about the possible role of heterozygous mutations
cause autosomal recessive forms of early-onset in thisgene.
parkinsonism, usually without atypical clinical
signs (Bonifati,2014). ATP13A2 (PARK9)
ATP13A2 encodes a large lysosomal P-type
Parkin (PRKN/PARK2) ATPase with 1180 amino acids and 10 trans-
PARK2 gene encodes for the protein parkin, membrane domains. Since the lysosomal degra-
which is involved in ubiquitin-mediated deg- dation pathway can clear -synuclein aggregates
radation of proteins. Mutation in parkin causes by macroautophagy, lysosomal dysfunction,
the most common autosomal recessive form of caused by mutations in ATP13A2, might con-
PD, which accounts for most PD cases in people tribute to the pathogenesis of parkinsonism (Pan
under 30years of age (juvenile PD, age of onset etal., 2008). Malfunctioning of this gene causes a
Brain Homeostasis and Parkinsons Disease 485

juvenile atypical parkinsonism characterized by More males are affected than females (in a ratio
pyramidal degeneration, dementia, and supra- of 1.5:1.0), although whether this reflects work-
nuclear gaze palsy, which is levodopa-responsive. place exposure, sex-linked genetic variability,
or the protective effect of estrogen is unknown
FBXO7 (PARK15) (Farrer, 2006). In principle, the progressive
The FBXO7 gene encodes the F-box only protein neurodegeneration of PD can be produced by
7 protein, which plays a role in phosphorylation- chronic neurotoxic exposure or limited expo-
dependent ubiquitination. Mutations in this sure, which then perpetuates a cascade of del-
gene cause a recessive form of juvenile parkin- eterious events. Examples include exposure to
sonism with pyramidal disturbances. The brain 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
pathology in patients with PARK15 remains (MPTP; Langston et al., 1983), which produces
unknown. symptoms identical to PD. Rotenonea mito-
chondrial poison in the environmenthas also
PLA2G6 been indicted in human epidemiological stud-
The PLA2G6 gene encodes for the ies with an elevated risk of PD (Tanner, 1992).
calcium-independent phospholipase A2 enzyme By contrast, protective effects have been noted
that plays a role in phospholipid remodeling and for cigarette smoking and alcohol and caffeine
apoptosis. Recessive mutations in this gene results intake (Hernan et al., 2002), but it is not clear
in levodopa-responsive dystonia-parkinsonism, how these agents influence disease risk. These
pyramidal signs, and cognitive/psychiatric fea- examples reinforce the concept that environmen-
tures with onset in early adulthood (Khateeb tal factors do modify PD susceptibility.
et al., 2006; Morgan et al., 2006; Paisan-Ruiz
etal., 2009). Magnetic resonance imaging in PD EFFECTS
patients showed brain atrophy with or without OFENDOGENOUSTOXINS
iron accumulation. Brain pathology in patients Apart from genetic and environmental factors
with PLA2G6 mutations also shows widespread that are implicated in the etiology of PD, there
accumulation of LBs in different rain regions, is also evidence that endogenous toxins such
suggesting links with typical PD (Paisan-Ruiz as distortion of metabolic pathways may be
etal.,2012). responsible for PD neurodegeneration. Changes
Despite all the evidence available confirming in normal metabolism might create toxic sub-
the hereditary basis of PD, the field remains highly stances due to environmental factors or inher-
controversial. While studies cited here and data ited differences in metabolic pathways (Dauer
from monozygotic and dizygotic disease in longi- and Przedborski, 2003). Cohen (1984) showed
tudinal twin studies have indicated that PD has a that endogenous toxins, which may be the nor-
genetic contribution, larger cross-sectional twin mal metabolism products of DA pathway, can
studies have argued against heritability (Piccini generate harmful reactive oxygen species that
etal., 1999; Tanner etal., 1999; Wirdefeldt etal., can prove detrimental to the cells (Cohen, 1984).
2004). As PD is not a homogeneous, penetrant Also consistent with this hypothesis is a report
trait (Simon et al., 2002), even within families that patients harboring specific polymorphisms
that are now known to be affected by monogenic in the gene encoding xenobiotic detoxifying
PD, the age of onset, symptoms, and end-stage enzyme cytochrome P450 may be at a greater risk
pathology can be quite variable. of developing early-onset PD (Sandy etal., 1996).
Furthermore, isoquinoline-derivative neurotox-
INFLUENCE ins that act similarly like MPTP and are toxic to
O F E N V I R O N M E N TA L FA C T O R S DAergic neurons have been recovered from PD
Studies analyzing the effects of environmental brains (Nagatsu,1997).
toxins on PD are in a nascent stage, although Apart from these examples, other causes of
examples are available in history that points PD include heterogenerative disorders, in which
toward a possible link. Epidemiological research parkinsonism is a manifestation of the disease
indicates that rural living, pesticide use, rather than a direct effect itself, such as demen-
well-water consumption, and certain occupa- tia with LBs, fragile-X-associated tremor/ataxia
tions, including mining and welding, are associ- syndrome, and so on; structural brain lesions
ated with an increased risk of PD (Firestone etal., (cerebrovascular disease, infectious brain lesions,
2005; Jankovic, 2005; Priyadarshi et al., 2001). postencephalitic, etc.) drug use (antipsychotics
486 Part IV:Homeostatic Therapies

such as haloperidol, olanzapince, etc.), and vari- harmful, soluble misfolded proteins from the cel-
ous psychogenicforms. lular milieu (Kopito, 2000). Several lines of study
now indicate that protein aggregation, sequestra-
PAT H O G E N E S I S O F P D tion, and inclusion body formation is perhaps
The pathogenesis of PD remains a mystery in a defensive measure aimed at removing toxic
itself. Different hypotheses have been put for- soluble misfolded proteins (Auluck et al., 2002;
ward to explain the pathogenesis of the disease, Cummings et al., 1999; Cummings et al., 2001;
the most common being misfolding and aggrega- Warrick et al., 1999). The question therefore
tion of proteins, mitochondrial dysfunction, and remains as to what causes these aggregated pro-
consequent oxidative stress, impairment of the teins and inclusion bodies to become toxic. One
ubiquitin-proteasome system, and neuroinflam- answer could be protein-damaging modifications
mation. However, these factors are not mutually as well as dysfunction of chaperones and other
exclusive, and one of the major aims of current proteosomal machinery that may indirectly con-
PD research is to find a common connection tribute to the toxicity. The cause of PD in patients
between them. The finding that oxidative dam- with either -synuclein mutations or DAergic
age to -synuclein enhance its ability to mis- neurodegeneration is thought to be related to
fold and aggregate (Giasson et al., 2000) is one (i) mutant -synuclein, (ii) excess levels of nor-
example of such an interaction. Another highly mal -synuclein adopting toxic conformations
uncertain issue in the field is whether the multi- (Bussell and Eliezer, 2001), or (iii) interference
ple cell-death related pathways activated during with the cellular handling of misfolded proteins.
PD neurodegeneration lead to a common cell fate In sporadic PD, there is a similar focus on both
such as apoptosis or remain highly divergent. protein-damaging modifications and dysfunc-
tion of protein-degradation systems. However,
Protein Aggregation and Misfolding irrefutable evidence that misfolded proteins and
Several age-related neurodegenerative disor- related aggregates are toxic to DAergic neurons
ders share a common pathology of abnormal in PD is still lacking.
deposition of misfolded and aggregated proteins,
although the composition and localization of Ubiquitination and RelatedStress
these aggregates varies in different disorders. The ubiquitin-proteasome system (UPS) is one of
Aggregated or soluble misfolded proteins can the major protein-degradation systems believed
become neurotoxic through a variety of mecha- to go awry in PD. UPS also degrades proteins that
nisms. They can directly cause cell damage by are misfolded and/or damaged, possibly caused
deforming the cell or interfering with the intra- by mutation, environmental stress, or intrin-
cellular trafficking in neurons. Protein inclusions sic folding inefficiency apart from degradation
might also sequester proteins that are essential of key regulatory proteins that control signal
for cell survival. Strict quality-control mecha- transduction, cell cycle progression, apoptosis, as
nisms determine the rate of protein synthesis as well as cellular differentiation (Ciechanover and
well as its degradation (Balch etal., 2008; Powers Brundin, 2003; Ciechanover etal., 2000; Hershko
etal., 2009)and normally prevent such aggregates and Ciechanover, 1998). Ubiquitinated proteins,
from forming. However, prolonged exposure to heat shock proteins/chaperones, and components
various stressors place an incredible burden on of the UPS have all been shown to accumulate
these mechanisms. When these mechanisms fail, within LBs, and both protein misfolding and UPS
aggregation-prone proteins abnormally accumu- dysfunction are implicated in neurodegenerative
late, as observed in neurodegenerative diseases processes in sporadic PD (Ii etal., 1997; Lennox
such as PD (Ross and Poirier, 2004). The common et al., 1989; Lowe et al., 1990; Schlossmacher
observation of protein aggregation in different etal., 2002). The presence of nondegraded, stable
diseases suggests that the protein deposition per ubiquitin conjugates in the brains of PD patients,
se is toxic to the neurons and that there should be as well as in genetic and toxic models of PD, sug-
a correlation between inclusion body formation gests that misfolded proteins in sporadic PD are
and neurodegeneration. This relation, however, properly recognized and ubiquitinated but not
is not convincingly shown in postmortem tis- efficiently degraded. Such inefficient degradation
sue samples from sporadic PD patients. Instead, may be the result of intrinsic resistance to pro-
the formation of aggregates may reflect a state of teasomal degradation of -synuclein-containing
cellular distress and represent a way to sequester aggregates (Lee etal., 2004)or a reflection of the
Brain Homeostasis and Parkinsons Disease 487

difficulty in unfolding stable protein aggregates OxidativeStress


by the 19S subunits of the proteasome complex Oxidative stress has been long implicated in the
(Voges etal.,1999). pathogenesis of PD. Mitochondrial dysfunction
The tissue content of abnormally oxidized/ and oxidative stresses are not mutually exclusive
aggregated proteins (which may misfold) events, and one plays a role in the formation of
increases with age (Beckman and Ames, 1998), the other (Cardoso et al., 1999; Cecarini et al.,
and neurons are particularly susceptible because 2007; Turrens, 2003). Reactive oxygen species
they are postmitotic. Cells respond to the mis- (ROS), which form a major component of oxida-
folded proteins by increasing the production tive stress, are generated within mitochondria
of chaperones. However, with aging the abil- in several sites of mitochondrial electron trans-
ity of cells to produce a variety of chaperones is port chains. Excessive production of ROS and/or
impaired, as is the activity of the proteosomal defective ROS removal can potentially damage
system. This provokes a vicious cycle, with the cellular lipids, proteins, and DNA. Mitochondrial
excess misfolded proteins further inhibiting an dysfunction is closely related to the increased
already compromised proteasome (Dauer and ROS formation in PD. Postmortem studies have
Przedborski, 2003). All of these factors converge consistently shown high levels of oxidation of
and generate a proteotoxic insult to thecells. lipids, proteins, and nucleic acids in the SNpc
of sporadic PD brains (Alam etal., 1997; Dexter
Mitochondrial Dysfunction etal., 1989; Floor and Wetzel, 1998; Jenner, 2003;
Another factor involved in the pathogenesis of Tsang and Chung, 2009; Yoritaka et al., 1996).
PD is mitochondrial dysfunction (Mandemakers The deterioration of DAergic neurons is triggered
et al., 2007; Parker and Swerdlow, 1998). by the pathogenic mitochondrial mechanism
Mitochondrial dysfunction due to oxidative enabling ROS production. Coincidentally, the
stress, mitochondrial DNA deletions, or altered postmortem brain tissue of the SN in PD patients
mitochondrial morphology and the interaction indicates stimulated levels of lipid peroxida-
of pathogenic proteins with mitochondria all tion, high oxidation of proteins and DNA, and
result in DAergic neurodegeneration. However, reduction of glutathione, which strengthens the
the most definitive evidence of mitochondrial notion that this type of stress is involved (Sian
dysfunction in PD has come from studies using et al., 1994). Also, significant alterations of the
MPTP, a toxicant that causes parkinsonian antioxidant defense system, in particular reduc-
symptoms in humans, rodents, and primates by tion in glutathione, were found in the SNpc of PD
inhibiting the mitochondrial complex-I of the patients (Sian etal.,1994).
electron transport chain (Sandy et al., 1993).
Similar to MPTP, other complex-I inhibitors B R A I N H O M E O S TAT I C
such as rotenone, maneb, paraquat, fenzaquin, S Y S T E M S I N V O LV E D I N P D
and trichloroethylene result in the loss of nigral Symptoms in PD can be broadly classified into
DAergic neurons in the mouse model of PD, two groups:(i)motor symptoms and (ii) nonmo-
implicating mitochondrial dysfunction in its tor symptoms. Motor symptoms are those that
pathogenesis (Betarbet et al., 2000; Gash et al., relate to how one moves and include tremor,
2008). Also, it has been shown that PD patients bradykinesia, rigidity, and postural instabil-
have significantly decreased complex-I activity ity. Non-motor symptoms are those that do
within the SN (Heo et al., 2012; Schapira et al., not involve movement, coordination, physical
1990). Recently, more convincing evidence of tasks, or mobility. Many studies have shown
mitochondrial dysfunction in PD has been that nonmotor symptoms may precede motor
reported in conditional knockout MitoPark symptomsand a Parkinsons diagnosisby
mice, which have a disrupted mitochondrial years. The most recognizable early nonmotor
transcription factor A (Tfam) gene in DAergic symptoms include loss of sense of smell, con-
neurons (Ekstrand et al., 2007). These mice stipation, sleep disturbances, mood disorders,
exhibited reduced mitochondrial DNA expres- depression, fear, and anxiety. Conceivably, the
sion and attenuated expression of respiratory smooth operation of routine motor and nonmo-
chain function in DAergic neurons in SN, along tor functions requires a structurally complex
with behavioral impairments and striatal DA neuronal circuitry with a high degree of inter-
depletion, mimicking progressive PD pheno- connectivity that utilizes a number of different
types, starting from 18 weeks ofage. neurotransmitters, including DA, glutamate,
488 Part IV:Homeostatic Therapies

acetylcholine, norepinephrine, serotonin, and a loss of DA in the striatum. This DA loss also
-aminobutyric acid (GABA). Traditionally extends to other parts of the basal ganglia, mani-
research in PD has focused mainly on neurode- festing the motor symptoms observed in PD. In
generation of DAergic nigrostriatal cells as the support of this hypothesis, postmortem DA lev-
key cause of motor dysfunctions. On the other els are extensively reduced in the globus pallidus,
hand, nonmotor symptoms associated with PD with a possible correlation between the pattern of
are not fully associated with respect to specific DA depletion and the presence of tremor in PD
neurotransmitter dysfunctions. (Rajput etal., 2008). Different kinds of dopamine
The basal ganglia is not only a group of func- receptors are widely present in the brain, which
tionally related and strongly interconnected nuclei are classified as D1 to D5 receptors. D1 mRNA
that form the key components of the complicated expression levels are highest in the dorsal stria-
circuitry that mediates motor and cognitive func- tum and nucleus accumbens but are also found
tion and behavior; it is also connected with differ- in the cortex and at lower levels in the hippocam-
ent parts of the cerebral cortex and the thalamus, pus, thalamus, and hypothalamus. Similar to
as well as with various structures leading to the D1, the D2 receptor is highly expressed in spiny
red nucleus, brainstem reticular formation, and projection neurons (SPNs) in the dorsal and ven-
spinal cord (Pollack, 2001), all of which play a tral striatum. D2 mRNA is also expressed in the
role in both normal motor and nonmotor func- cortex, hippocampus, and hypothalamus. D3 is
tions of the body. The role of the basal ganglia in present in particular in the nucleus accumbens,
motor function has been extensively investigated, with only very low levels in the dorsal striatum,
and the pathological changes to these structures hippocampus, and cortex. Low levels of D3 auto-
in PD are well known (Galvan and Wichmann, receptors have been detected in SN and ventral
2008; Halliday et al., 2005). However, its role in tegmental area neurons. D4 shows the lowest
nonmotor functions is not as widely understood. expression levels among all DA receptors in the
Due to the interconnectivity of different neu- basal ganglia. The D5 receptor has been detected
rotransmitter systems, disruption in any one of in the cortex, dorsal and ventral striatum, hypo-
them will have consequences in the symptoma- thalamus, and hippocampus. In the dorsal stria-
tology of PD. For example, the motor function tum, D5 receptors are localized in SPNs and in
of the striatum is dependent on the equilibrium cholinergic interneurons.
reached between DA and acetylcholine (Zhou There are two distinct pathways that process
et al., 2003), while DAergic activity within the signals through the basal ganglia:the direct path-
SNpc is itself modulated by glutamatergic and way and the indirect pathway. These two pathways
GABAergic innervations (Fink and Gothert, have opposite net effects on thalamic target struc-
2007). Disturbances in central noradrenergic sys- tures. Excitation of the direct pathway has the net
tems may be causative in the onset of depression effect of exciting thalamic neurons (which in turn
and anxiety by interactions with serotonin and make excitatory connections onto cortical neu-
DA (Richard, 2005). The observation that selective rons). Excitation of the indirect pathway has the
serotonin reuptake inhibitors (SSRIs), adminis- net effect of inhibiting thalamic neurons (render-
tered to treat the depressive symptoms associated ing them unable to excite motor cortex neurons).
with PD, can affect the motor symptoms suggests The normal functioning of the basal ganglia appar-
that there may be an interaction between sero- ently involves a proper balance between the activi-
tonin and dopamine in PD (Vajda and Solinas, ties of these two pathways. One hypothesis is that
2005). Thus, although loss of DA neurons in the the direct pathway selectively facilitates certain
basal ganglia is most often associated with the motor (or cognitive) programs in the cerebral cor-
disease and its characteristic motor symptoms, it tex that are adaptive for the present task, whereas
is clear that the changes in DA interactions with the indirect pathway simultaneously inhibits the
other neurotransmitters and subsequent effects execution of competing motor programs. An
both within and outside the basal ganglia need to upset of the balance between the direct and indi-
be considered when looking at the wider (nonmo- rect pathways results in motor dysfunctions.
tor) symptoms reported in patients withPD. Therapeutic strategies to relieve PD
symptoms are mostly focused on increas-
Dopaminergic Neurotransmission ing the DA levels in the brain. DA receptor
The pathological hallmark of PD is the preferen- stimulation in the dorsal striatum is the pri-
tial loss of DAergic neurons in SN resulting in mary mechanism by which these drugs help
Brain Homeostasis and Parkinsons Disease 489

alleviate the motor features associated with PD. in various motor symptoms of PD (Lange etal.,
L-3,4-dihydroxyphenylalanine (L-DOPA) ther- 1997), particularly in those patients that do not
apy is most commonly used to treat motor symp- respond to L-DOPA therapy (Rodriguez et al.,
toms of PD. L-DOPA is a precursor to DA, and 1998). Binding studies in human postmor-
once it crosses the blood-brain barrier (which DA tem tissue revealed that the levels of the NR1/
cannot), it nonspecifically increases the concen- NR2B NMDA receptor, the AMPA receptor,
tration of DA at dopamine receptors in the brain, and mGluR5 are increased in the putamen of
thereby relieving some of the cardinal signs patients with levodopa-induced dyskinesia (LID)
of PD. In some cases, dopamine receptor ago- compared to those without (Calon et al., 2003;
nists are used for symptomatic treatment of PD. Ouattara etal., 2011). These findings support the
Recently monoamine oxidase-B (MAO-B) inhib- hypothesis of enhanced glutamatergic neuro-
itors have been used to treat some symptoms of transmission in LID and that the selective inhi-
PD. MAO-B inhibitors increase the level of DA bition of glutamatergic hyperactivity may be an
in the basal ganglia by blocking its metabolism. effective strategy for the treatment of some PD
They inhibit MAO-B, which breaks down DA symptoms. Other studies have shown that mod-
secreted by the DAergic neurons. The reduction ulation of glutamatergic neural tone in patients
in MAO-B activity results in increased L-DOPA with PD may also have a beneficial effect on cog-
in the striatum. nitive impairment (ONeill and Dix,2007).
However, DA replacement therapies often Apart from iGluRs, mGluRs, which are
have little or no effect on nonmotor manifesta- expressed abundantly in basal ganglia, are also
tions of PD. Since the DAergic system is spared in involved in some PD symptoms, although their
the early stages of PD, diagnosis and treatment of direct effects are not properly understood. It is
nonmotor symptoms are of clinical importance hypothesized that antagonism of subtypes 1 and
at early stages of the disease, even before motor 5 can protect against nigrostriatal degenera-
symptoms have become apparent. tion, whereas enhancing the other receptor sub-
types may help relieve parkinsonian symptoms
Glutamatergic Neurotransmission by inhibiting glutamate release and/or reduc-
Glutamate is the most abundant excitatory neuro- ing inhibition of lateral globus pallidus neurons
transmitter in the central nervous system. It exerts (Conn etal.,2005).
its effects through ionotropic glutamate receptors Overall, it is safe to postulate that activation
(iGluRs, which include N-methyl-D-aspartate of glutamatergic receptors may be effective in pre-
[NMDA], -amino-3-hydroxy-5-methylisoxazole clinical models of anxiety, pain, depression (via
propionic acid [AMPA], and kainate receptors), AMPA), and sleep dysfunction, whereas inhibi-
which are involved in fast synaptic transmission tion of glutamatergic neurotransmission may
and metabotropic glutmate receptors (mGluRs, result in increased anxiety and executive dys-
subtypes 18) that mediate slow synaptic trans- function but also lessen depression (via NMDA)
mission. All of these receptors subtypes are and improve cognitive function. However, stud-
expressed throughout the brain, including the ies are needed to confirm this hypothesis and to
basal ganglia, and in peripheral organs, suggest- design novel strategies based on this to treatPD.
ing that glutamate is also involved in biological
processes other than neuronal transmission. GABAergic Neurotransmission
The striatum is the primary input nucleus GABA is the major inhibitory neurotransmitter
in the basal ganglia. Besides dense innervation in the brain. GABA signaling occurs via iono-
from DAergic SN neurons, the striatum receives tropic GABA A and metabotropic GABA B recep-
excitatory afferents from the glutamatergic neu- tors, which are expressed in all human basal
rons in the cerebral cortex, the thalamus, and the ganglia structures. In the striatum, the two dif-
amygdala. The striatum then modulates signal- ferent types of GABAergic SPNs express DA
ing to other brain regions through the basal gan- receptors and give rise to the direct and indirect
glia output nuclei, the internal part of the globus signaling pathways (Gerfen and Surmeier, 2011).
pallidus (GPi) and the SN pars reticulata. The loss Therefore, SPNs are affected by postsynaptic
of DA neurons is believed to cause an increase changes, resulting in an altered activity pat-
in glutamatergic activity in the basal ganglia. tern for both pathways. This signaling alteration
Preclinical studies using NMDA and AMPA changes the output of the basal ganglia, causing
antagonists have demonstrated improvements some of the typical motor features seen in PD
490 Part IV:Homeostatic Therapies

patients, including stiffness and bradykinesia. increasing the susceptibility of DAergic neurons
GABAergic neurotransmission itself is altered in to degeneration or by inhibiting the repair of
PD. Binding studies in postmortem tissue indi- neurons that are already damaged (Fornai etal.,
cate that LID is associated with increased levels 2007). In addition, it has been reported that loss
of GABA A receptors in the internal globus palli- of noradrenergic innervation facilitates the onset
dus and decreased density of GABAB receptors in of dyskinesia during DA replacement therapy
the putamen and in the external globus pallidus (Colosimo and Craus,2003).
in PD patients compared to controls. (Day etal., Among other nonmotor symptoms associ-
2006). The description of these region-specific ated with PD, some studies have shown that sig-
receptor alterations is interesting. However, nificantly more patients with PD reported pain
follow-up studies are required to extend these as compared to healthy controls. Like serotoner-
findings to the development of therapeutic gic and DAergic systems, noradrenaline is a key
strategies. neurotransmitter of the endogenous pain system.
Advanced PD is also associated with As sustained pain induces noradrenergic feed-
GABAergic SPNs characterized by truncated back inhibition (Pertovaara, 2006), lower levels
dendrites and a lower number of spines (McNeill of noradrenaline would most likely be associated
et al., 1988; Stephens et al., 2005) due to deple- with increased symptoms of pain as observed in
tion of DA. Although several of the nonmotor PD patients. However, since other neurotrans-
symptoms associated with PD have been shown mitter systems are also involved in the pathology
to involve GABAergic neurotransmission, such of pain, it would be safer to suggest that there may
as pain, sleep disorders, and depression, there is be some interplay between these various neuro-
a lack of evidence as to how these symptoms are transmitter systems in the propagation ofpain.
modulated by changes in endogenous or circulat-
ing GABA concentrations in PD. Studies in ani- Cholinergic Neurotransmission
mals and in individuals without PD suggest that Cholinergic neurons are found in the basal gan-
decreases in GABAergic neurotransmission gen- glia (i.e., pedunculopontine nucleus [PPN]) and
erally result in the reduction of these symptoms brainstem, as well as in the striatum (a major-
(Barone,2010). ity of interneurons). These neurons are a major
source of acetylcholine, which mediates its
Adrenergic Neurotransmission effects via nicotinic and muscarinic receptors.
Alpha 2-adrenergic (-2a and 2c) receptors are Whereas neurons in the basal ganglia project-
widely distributed throughout the basal ganglia. ing to the SNpc, subthalamic nucleus, globus
Studies in mice have demonstrated that these pallidus, and striatum contain only muscarinic
receptors modulate the sensitivity of DAergic receptors, both subtypes of cholinergic recep-
receptors, and activation of -2 receptors facili- tors are found on neurons projecting to the
tates movements produced by activation of the substantia nigra pars reticulata (Zhou et al.,
direct pathway of the basal ganglia motor circuit 2003). In accordance with their expression,
(Colosimo et al., 2006). Noradrenergic systems, deficits in the cholinergic system are believed
in conjunction with serotonergic and DAergic to play a role in the etiology of cognitive dys-
mechanisms, appear to play an important role function and dementia in PD (Williams-Gray
in the etiology of depression in PD. A recent etal., 2006). Reduction in cholinergic activity in
positron emission tomography study has shown the cortex and degeneration of cholinergic neu-
that noradrenergic innervations in depressed PD rons in the nucleus of Meynert is closely asso-
patients are much lower than those in healthy ciated with declining cognitive functions in PD
controls (Remy etal., 2005). Keeping in mind this (Perry et al., 1985). Recent studies also impli-
observation, a number of selective noradrenergic cate the role of PPN neurons in the etiology of
reuptake inhibitors are currently undergoing gait disturbances in PD (Karachi et al., 2010).
clinical trials for the treatment of depression and This study revealed that PD patients with gait
anxiety in PD (Fox etal.,2008). and balance abnormalities had a lower number
Reduced levels of noradrenaline have been of cholinergic neurons in their PPN than PD
observed in patients with PD compared to normal patients without these signs or unaffected con-
subjects (Barbic etal., 2007). Loss of noradrena- trols. Cholinergic PPN neurons are also shown
line, which occurs at early stages of PD, has been to undergo degeneration in patients with PD
reported to worsen disease progression, either by (Rinne etal.,2008).
Brain Homeostasis and Parkinsons Disease 491

Autonomic nervous system dysfunction (e.g., Approximately 80% of people diagnosed with PD
sleep disruption, sexual dysfunction, etc.) is experience pain (Beiske etal., 2009). Serotonergic
common in PD, affecting up to 80% of patients pathways apart from adrenergic pathways have
(Zesiewicz etal., 2003). It has been suggested that been implicated in modulating pain. It has been
cholinergic dysfunction might be responsible shown that treatment of PD patients with seroto-
for, or contribute to, autonomic dysfunctions in nin and adrenaline reuptake inhibitors resulted
PD (Vernino etal., 2009). It is being speculated in varying degrees of pain relief for 65% of
that inhibition of or diminished cholinergic patients with PD (Djaldetti etal.,2007).
neurotransmission in clinical and preclinical Thus stimulation of the serotonergic system
paradigms of PD results in attention dysfunc- in PD appears to result in decreases in several
tion, cognitive impairments, and executive dys- nonmotor symptoms, including anxiety, depres-
function. Conversely, decreased stimulation sion, and possibly pain. Dampening the trans-
or enhanced cholinergic neurotransmission is mission of serotonin leads to deficits in cognition,
associated with lessening of several nonmotor executive function, and possibly fatigue, thereby
symptoms, including anxiety, apathy, attention making serotonergic therapy as a treatment for
dysfunction, orthostatic hypotension, cognitive PD highly complicated.
impairment, and sleep disorders, which are more
autonomous in function. Norepinephrine-Mediated
Neurotransmission
Serotonergic Neurotransmission Along with acetylcholine receptors, norepineph-
The basal ganglia, including the globus pallidus rine has been postulated to be involved in pos-
and SN, receive numerous innervations from tural instability and gait abnormalities observed
the serotonergic (5-HT) neurons in the brain- in PD (Grimbergen etal., 2009). These manifes-
stem raphe nuclei. 5-HT neurons innervate both tations are unresponsive to L-DOPA therapy.
DAergic neuronal cell bodies of the SN and the However, treatment with a DA and norepineph-
region of their terminal projections in the stria- rine reuptake inhibitor improved freezing of gait
tum. The anatomical interaction of the 5-HT in patients with advanced PD who also received
system with DAergic components of the basal deep brain stimulation (Moreau et al., 2012).
ganglia facilitates functional modulation of DA These data suggest that modulation of norepi-
neurotransmission by serotonin in the normal, nephrine signaling might have some beneficial
non-parkinsonian brain. This mainly occurs via effects on certain symptomsofPD.
5-HT1A and 5-HT1B receptors, which facilitate DA
release, while stimulation of the 5-HT2C receptor Adenosine-Mediated
inhibits DA release (Alex and Pehek,2007). Neurotransmission
Levels of serotonin, its metabolite Adenosine is a ubiquitous endogenous purine
5-hydroxyindoleacetic acid, serotonin trans- nucleoside that acts as a synaptic modulator, and
porter, and tryptophan hydroxylase are reduced its effects are transmitted by at least four receptor
in striatal tissue from PD patients compared to subtypes:A1, A2A, A2B, and A3 receptors. The A1,
controls (Kish etal., 2008). Low levels of seroto- A2B, and A3 adenosine receptors are widely dis-
nin have been linked to alterations in mood, such tributed throughout the brain. In contrast, A2ARs
as depression and changes in sleep architecture are localized within the basal ganglia, with the
in PD patients, with a reduction of serotonin highest concentration in the caudate-putamen
levels of up to 50% in cortex and basal ganglia (Rosin etal., 1998; Schiffmann etal., 1991). A2ARs
(Scatton et al., 1983). Selective SSRIs, which are able to interact antagonistically with postsyn-
inhibit presynaptic reuptake of serotonin via the aptic dopamine D2 receptors and are also thought
serotonin transporter, have been used in several to interact with presynaptic glutamatergic recep-
studies to improve depression symptoms in PD tors. Apart from this, A2ARs also colocalize with
patients (Richard et al., 2012). However, not all dendritic spines of enkephalin-rich striatopallidal
patients respond to SSRI treatment, suggesting GABAergic neurons of the indirect pathway. Thus
that PD-related depression is not fully attribut- A2ARs are functionally relevant to the operation
able to perturbations in serotonin signaling. of the indirect pathway of the basal ganglia sys-
As mentioned in the discussion on adren- tem (Morelli etal., 2007; Schiffmann etal.,2007).
ergic neurotransmission, one of the other com- Patients with PD have altered adenosine
mon symptoms associated with PD is pain. A 2AR expression in the basal ganglia, suggesting
492 Part IV:Homeostatic Therapies

a pathogenic role for these receptors. In a post- via the modulation of A2AR might reduce PD
mortem study of patients with PD, a 2.95-fold symptoms and possibly exert neuroprotective
increase was observed in A 2AR density in the effects (Prediger,2010).
putamen compared with healthy subjects (Varani Altogether, alterations in adenosine-mediated
et al., 2010). Another autopsy study demon- neurotransmission impact upon DAergic
strated that patients with PD and dyskinesia who neuronal loss, cognition, pain, and sleep dis-
were treated with levodopa had increased lev- turbances. However, the effect of adenosine
els of A2AR and corresponding messenger RNA modulation has not yet been determined in PD
compared with levodopa-treated patients with- models.
out dyskinesia (Calon etal., 2004), suggesting a
tightly liked relation between A2ARs, dyskinesia, I N F L A M M AT O R Y
and PD symptoms. RESPONSEINPD
In animal models of PD, adenosine Inflammation in the central nervous system is a
A2AR-specific antagonists have been shown to prominent and common feature of neurodegener-
consistently reverse motor deficits such as ative diseases, including PD. Neurodegeneration
tremors and rigidity and/or enhance the effect of substantia nigral neurons leads to a plethora
of DA-based pharmaceuticals (Hauser and of inflammatory responses induced by soluble
Schwarzschild, 2005). This effect may result from factors secreted from injured neurons, which is
action on the indirect pathway while allowing also termed neuroinflammation (Tansey and
DAergic action by the D1-mediated direct path- Goldberg, 2010). However, whether neuroinflam-
way. Moreover, A 2AR antagonists are also neuro- mation is a causal/trigger factor or a secondary
protective in animal models of PD (Morelli etal., consequence in PD remains unknown. It appears
2007). Thus A 2AR antagonists can be considered there is complex interplay between neuroinflam-
prodopaminergic agents and could potentially mation and other proposed pathogenic mecha-
reduce the effects associated with DA depletion nisms of PD such as mitochondrial dysfunction
in PD. Despite all the data available showing the and oxidative stress (Witte etal.,2010).
protective effects of A2AR antagonists, the exact Inflammation, which is generated by an
mechanism responsible for the neuroprotective activated immune system, is a strictly regulated
potential of adenosine antagonists is not clear, self-defensive mechanism against pathogenic
although modulation of brain glutamate and stimuli or injury that results in the protection
aspartate release are one potential hypotheses. of the host organism by clearance of pathogenic
As described previously, disorders of sleep stimuli or debris to promote the healing pro-
and wakefulness are extremely common in cess (Khandelwal etal., 2011; Stone etal., 2009).
PD, with an estimated prevalence of 98%. Both innate and adaptive immune responses
Adenosine has long been accepted as a media- have been implicated in the pathophysiology of
tor of sleep, although the contributions of the PD. While innate immunity does not require the
adenosine receptor subtypes are still a matter presence of a specific antigen to develop, adap-
of intense debate. There is increasing evidence tive immunity occurs when specific antigens
that adenosine A 2AR play a critical role in the are presented and recognized by lymphocytes.
sleep-promoting effects of adenosine and most Immunogenic factors released by injured dopa-
likely mediates the sleep-inhibiting effects of minergic neurons have the potential to trig-
the nonselective adenosine receptor antagonist ger a detrimental innate and adaptive immune
caffeine. Accordingly, it has been suggested that response, thereby amplifying the pathological
insomnia be considered as a possible adverse side process. Neuroinflammatory responses in PD
effect of A2ARs antagonists used in the treatment consist of phenotypic and morphological activa-
of PD (Ferre etal.,2007). tion of microglia cells, astrogliosis, and lymphatic
Epidemiological and preclinical data suggest infiltration of T-cells (Brochard et al., 2009;
that caffeine may confer neuroprotection against Hirsch and Hunot, 2009). It also leads to release
the underlying DAergic neuron degeneration and of proinflammatory cytokines, chemokines, ele-
may influence the onset and progression of PD. It ments of the complement cascade, and increased
has been demonstrated that among the various expression of several enzymes that are involved
pharmacological effects attributed to caffeine, in the excess production of reactive oxygen and
this agent exerts A2AR antagonistic properties. nitrogen species (Lull and Block, 2010; Tansey
Thus it is possible that caffeine and its derivatives and Goldberg,2010).
Brain Homeostasis and Parkinsons Disease 493

E V I D E N C E O F I N F L A M M AT I O N as a potential mechanism in neurodegenera-


INPD:POSTMORTEM STUDIES tive disorders. Thus increased time of inflam-
Initial evidence of the involvement of inflam- mation flares the amount of proinflammatory
mation in the progression of PD stems from molecules and subsequently the neurons are
a postmortem study over 20 years ago, which eventually affected by neurodegenerative envi-
demonstrated the presence of activated micro- ronment (Czeh et al., 2011; Smith et al., 2012).
glia in the SNpc of a PD patient (McGeer etal., The role of microglia in neurodegenerative dis-
1988). This initial finding was later confirmed eases is unequivocal. However, the mechanism
by other postmortem studies (Desai Bradaric that underlies the switch from neuroprotective to
et al., 2012; Imamura et al., 2003; Mirza et al., autoaggressive effector microglia, which causes
2000). Enzymes associated with inflamma- neurodegeneration, is as yet unknown (Hanisch
tion, such as inducible nitric oxide synthase and and Kettenmann, 2007; Khandelwal etal.,2011).
cyclooxygenase-2, have also been identified post- Astrocytes are the most abundant cells in
mortem in PD brains (Hunot etal., 1996; Knott the brain, heavily outnumbering neurons. They
et al., 2000). Several cytokine levels, including are dispersed throughout the central nervous
tumor necrosis factor- (TNF-), TNF- recep- system, where one astrocyte occupies its own
tor, interleukin (IL)-1, IL-6, interferon (IFN)-, territorial region without any overlap with the
and the inflammation-related transcription fac- territory of another. Astrocytes perform regula-
tor NFk, were elevated in the SN of PD patients tory and supportive roles, such as biochemical
(Boka etal., 1994; Mogi etal., 1994a; Mogi etal., and nutritional support for neurons, extracel-
1994b; Mogi etal., 2007). Inflammatory markers lular ion balance, neuronal cellular activity and
analyzed in the cerebrospinal fluid of PD patients blood flow, and repair of scarring of brain and
also showed elevated levels of TNF-, IL-1, and spinal cord tissue (Sofroniew and Vinters, 2010).
IL-6 (Blum-Degen etal., 1995; Mogi etal., 1994a; Apart from this, astrocytes also have immune
Mogi etal., 1994b; Zhang etal.,2008). functions that limit the spreading of inflamma-
tory cells and infectious agents from the dam-
CELLS AND aged area to healthy parenchyma by secreting
N E U R O I N F L A M M AT O R Y pro- and anti-inflammatory molecules (Hamby
PAT H WAY S I N V O LV E D I N P D and Sofroniew,2010).
A glial reaction involving astrocytes and micro- Glial reaction in pathological situations of the
glial cells and lymphocytic infiltration has been central nervous system such as PD can play either
described in several animal models of PD includ- a beneficial or a detrimental role. The former
ing MPTP, 6-hydroxydopamine, rotenone, and properties are thought to be possibly mediated by
genetic mouse models. Microglia are active sen- the production of a variety of trophic factors, the
sors of the body that mediate innate immune uptake of any excess of glutamate, or the removal
responses in the brain via antigen-presenting of cell debris; the latter can result from the pro-
and effector functions such as phagocytosis. They duction of a host of cytotoxic molecules rang-
also possess various beneficial functions, such ing from ROS and nitrogen oxygen species to
as neurotrophic factor release, removal of toxic cytokines. The astrocytic reaction is a well-known
substances, neuronal repair, synaptic remod- neuropathological characteristic of the SN in PD.
eling, synaptic pruning, and guidance to neu- Glial fibrilliary acidic protein-specific staining in
ral stem cells during synaptogenesis. Microglial PD patients showed that astrocytes are heteroge-
activation can be stimulated by aggregation of neously distributed within the mesencephalon
mutant protein such as -synuclein and other in healthy individuals (Damier etal., 1993). The
cellular mediators that are released by degener- density of astrocytes is low in the SNpc, which
ating neurons (Lull and Block, 2010). Following is severely affected in PD, whereas the density is
activation in the early phases, microglia migrate high in the ventral tegmental area and the cat-
through the site of damage and secretes pro- and echolaminergic cell group A8, areas that are less
anti-inflammatory cytokines. Microglial activa- affected in PD. Therefore, vulnerable neurons in
tion can also be acute or chronic depending on patients with PD might have few surrounding
the type and duration of the external stimuli or astroglial cells, which detoxify oxygen free radi-
activating factor. Whereas short-term activation cals by glutathione peroxidase in healthy indi-
of microglia is generally believed to be neuropro- viduals, and this limited astroglial environment
tective, chronic activation has been implicated might be a susceptibility factor for the disorder.
494 Part IV:Homeostatic Therapies

A30% increase in the density of astroglial cells need to be established. Neuroinflammation


in the SN of patients at postmortem was detected might be a simple consequence of neuronal
by quantitative analysis (Damier et al., 1993). changes or degeneration. Alternatively, neuro-
Looking at this evidence, it might be safe to pos- inflammatory processes might be a main cause
tulate that in a disease like PD, in which not all of the disease. Whatever the origin of the neu-
neurons die at the same time, the very first cells roinflammatory processes in PD, therapeutic
to succumb to the pathological process activate intervention aimed at prevention or downregu-
the neighboring glial cells. Once activated, glial lation of these immune-associated mechanisms
cells, and especially microglia, can engage in could be of great use to stop disease progression
the production of toxic molecules that can pro- or even halt the pathological process. Different
mote the demise of surrounding compromised commonly used analgesics and antipyretics such
neurons. as nonsteroidal anti-inflammatory drugs (e.g.,
Lymphocytes might also participate in the ibuprofen) and cyclooxygenase inhibitors can
inflammatory reaction in the brains of patients have protective effects on the symptomatology
with PD. Different studies have reported higher of PD (Rees et al., 2011; Reksidler et al., 2007;
densities of cytotoxic T lymphocytes (CD8+ and Teismann and Ferger, 2001; Teismann et al.,
CD4+) in the SN of PD patients as compared to 2003a; Teismann etal., 2003b).
healthy controls (Brochard etal., 2009; McGeer Strategies aimed at suppressing the activa-
etal., 1988). These cells were in close contact with tion of glial cells and their inflammatory prop-
blood vessels (suggesting migration from the erties by use of various drugs is another way
bloodstream) and near to melanised dopamin- medications are being used to study their effects
ergic neurons (suggesting an interaction between on PD. Administration of drugs with a broad
the lymphocytes and the dopaminergic neurons). spectrum of action on inflammation would
The CD8+ and CD4+ T cells were not detected in be more likely to protect dopaminergic neu-
the red nucleus, an area not affected in PD, sug- rons efficiently than more selectively targeted
gesting that this infiltration is highly selective drugs. Some of these drugs include agonists of
for the lesioned brain areas (Hirsch and Hunot, PPAR, a receptor that regulates fatty acid stor-
2009). These data indicate that some peripheral age and glucose metabolism (Dehmer et al.,
cells can enter the brain parenchyma during the 2004; Quinn et al., 2008; Swanson et al., 2011;
neurodegenerative process; therefore, changes Whitehead, 2011), adenosine receptor antago-
in blood-brain barrier function might occur in nists (Bibbiani et al., 2003; Hauser et al., 2011;
the brains of patients with PD. The presence of Hodgson etal., 2010; Kanda etal., 1998; LeWitt
neuroinflammatory processes in portmortem etal., 2008; Pourcher etal., 2012), 7 nicotinic
tissues has also been confirmed on a molecu- acetylcholine receptor agonists (Acker et al.,
lar basis. Increased concentrations of TNF-, 2008; Mazurov et al., 2005), as well as immu-
2-microglobulin, epidermal growth factor, notherapy (Laurie et al., 2007; Reynolds et al.,
transforming growth factor (TGF), TGF1, 2010; Reynolds etal.,2009).
and interleukins 1, 6, and 2 has been found in
the striatum and SN of patients with PD, indicat- NEUROTROPHIC
ing that there are neuroinflammatory processes FA C T O R S I N P D
in the affected brain regions of patients with PD. Neurotrophic factors are neuroprotective
However, these studies do not help to determine secreted peptides that promote development,
whether such changes are involved in the patho- influence neuronal survival and axonal growth,
logical process or are merely a consequence of and modulate neuronal functions during devel-
neuronal degeneration. opment. Neurons that fail to obtain a sufficient
quantity of the necessary neurotrophic factors
THER APEUTIC die by a process called programmed cell death.
INTERVENTIONS In response to injury, many trophic factors and
F O R N E U R O I N F L A M M AT I O N their receptors have been shown to increase in
An increasing amount of evidence, such as concentration, suggesting an endogenous regen-
that discussed previously as well as continuing erative response by these molecules (Hughes
research, has shown that neuroinflammation is etal., 1999). Furthermore, these factors serve as
involved in pathogenesis of PD. However, the ori- neuroprotectant molecules against cytotoxic cell
gin and role of these neuroinflammatory changes damage.
Brain Homeostasis and Parkinsons Disease 495

It has been proposed that the loss of endoge- Despite its high efficacy in rodents and non-
nous target-derived trophic support for selective human primate models, neurotrophic factor
neuronal populations may lead to the neuronal therapy has not provided expected beneficial
degeneration characteristic of PD and other effects in PD patients. The lack of symptomatic
neurodegenerative diseases, but direct support relief seen in these patients may be due to the
for this hypothesis is currently lacking (Connor inadequate penetration of neurotrophic factors
and Dragunow, 1998). Initial in vitro studies for from the cerebrospinal fluid into the striatum
determining the effects of different neurotrophic upon intracerebroventricular delivery, suggest-
factors on dopaminergic neuronal populations ing that the factors did not diffuse sufficiently to
included assays of: brain-derived neurotrophic reach the degenerating nigrostriatal fibers and
factor (BDNF; Hyman et al., 1991; Hyman neurons (Kordower, 2003). Later studies involv-
etal., 1994)and glial-derived neurotrophic fac- ing intraputamental injections in PD patients
tor (GDNF; Engele etal., 1996; Lin etal., 1993). (Patel et al., 2005), GDNF delivery via intrapu-
In MPTP- and 6-hydroxydopamine-lesioned tamental pumps in monkeys (Maswood et al.,
animal models with PD, BDNF (Frim et al., 2002), transplantation of GDNF-secreting cells
1994) and GDNF (Gash et al., 1996; Tomac in rodents (Akerud etal., 2001; Cunningham and
et al., 1995) have been shown to promote the Su, 2002), and in-vivo gene therapy using viral
survival of neurons and ameliorate PD symp- vectors (Bilang-Bleuel et al., 1997; Zheng et al.,
toms. Supranigral implants of fibroblasts engi- 2005)have all shown beneficial effects of modu-
neered to secrete human BDNF are shown to lating GDNF levels on PD symptoms.
protect dopaminergic neurons from MPTP
toxicity (Frim et al., 1994). These neuroprotec- CONCLUDING REMARKS
tive effects of BDNF may be mediated by its AND FUTURE DIRECTIONS
antioxidant-promoting capacity. BDNF also PD is predominantly a motor disorder; how-
stimulates DA activity and turnover and is ever, other variable indicators (such as nonmo-
hypothesized to play a role in the compensatory tor symptoms, depression, sleep disturbances,
actions of surviving DA neurons in early-stage cognitive dysfunction, etc.) cannot be ignored
PD (Blochl and Sirrenberg, 1996; Murer et al., in its pathophysiology. Cumulatively, all of these
2001). Although BDNF has been found to pro- symptoms are enervating and can have a major
tect DA neurons in cell culture and animal mod- impact on the quality of life of patients. Since
els of PD, its large molecular size may prevent there is currently no cure for PD, current thera-
its delivery to DA neurons. It has been suggested peutic strategies are focused on (i) delaying the
that alternative strategies for BDNF delivery progression of the disease, (ii) providing symp-
may be more effective, such as using endogenous tomatic relief from the symptoms, (iii) causing as
factors to induce BDNF expression or transplan- few adverse effects as possible via different medi-
tation of BDNF-releasingcells. cations and therapeutics, and (iv) developing new
GDNF supports the survival of several differ- therapeutics to treat PD. No single agent cur-
ent neuronal populations in both the central and rently treats both the motor and the nonmotor
the peripheral nervous system. GDNFs potential manifestations of the disorder. Indeed, effective
therapeutic value for PD was first recognized in treatment of the nonmotor symptoms remains a
1993 when it was purified and shown to promote major unmet needinPD.
the growth and survival of midbrain embryonic Genes implicated in Mendelian forms of PD as
dopaminergic neurons (Lin et al., 1993). Later well as various risk factors associated with them
studies showed that GDNF encouraged neuronal have provided new insights into the pathogen-
fiber outgrowth and improved motor function esis of the disease. However, known PD-causing
when delivered into the cerebral ventricles or genes account for only a small fraction of mono-
directly into the striatum or SN in both rodent genic forms. Robust high-density single nucleo-
and primate models of PD (Bjorklund et al., tide polymorphism genotyping technologies
2000; Chiocco et al., 2007; Peterson and Nutt, and data analysis programs, combined with the
2008; Siegel and Chauhan, 2000). Similar to analysis of copy-number variations and large
rodent studies, the positive effects of GDNF have pathogenic genomic rearrangements, will help
also been demonstrated in nonhuman primate in identifying novel loci associated with PD. It is
PD models (Gash etal., 1995; Gash etal., 1996; also imperative to more closely study the cumu-
Zhang etal.,1997). lative effect of different geneenvironment and/
496 Part IV:Homeostatic Therapies

or genegene interactions to evaluate the overall PD. It should also be kept in mind that rodent and
picture for the various factors contributing to the nonhuman primate models can only partially
PD phenotype. recapitulate the pathological changes that occur
It is now well accepted that abnormal protein in patients with PD. The expression patterns of
degradation and accumulation is a critical fac- neurotransmitter receptors and transporters in
tor mediating the degeneration of dopaminergic normal and disease-like states, their conforma-
neurons in PD. As the oxidative environment in tion and subunit composition, and the interact-
neurons is also believed to occupy a central role in ing partners of receptors, as well as the binding
neurodegeneration, of particular relevance to PD affinity between a specific drug target and a test
is the increased detection of dopamine quinones compound are only some of the parameters that
under conditions of oxidative stress (Bisaglia may vary significantly between species. These
etal., 2007). These quinones are a highly impor- factors present a challenge for the translation
tant species since they are known to interact with of therapeutic strategies from disease models to
proteosomal and mitochondrial machineries to patients and highlight the importance of validat-
impair their functions (Berman and Hastings, ing preclinical data in human tissue or subjects
1999). Thus, despite the active debate over the whenever feasible.
sole cause of PD, it is likely that genegene and A major scientific breakthrough will occur if
geneenvironment interactions combined with we one day we develop agents that alleviate both
oxidative and proteolytic stress, as well as mito- the motor and the nonmotor symptoms of PD
chondrial dysfunction, are all intricately linked through interaction with several of the affected
in a pathogenic feedforward cycle responsible for homeostatic neurotransmission systems in the
the progression of PD. This gives rise to a multi- brain. Due to the plethora of dynamic interac-
factorial situation where all the factors are con- tions that occur between different neurotrans-
volutedly linked, making the development of a mitter systems in PD, it is likely that the future
single therapeutic agent for treatment of PD even of PD treatment will depend on type (and tim-
more complex. ing) of symptoms displayed by each patient, and
Deficits in dopaminergic neurotransmit- these factors will help us provide more effective
ters are largely responsible for the characteris- treatment of both the motor and the nonmotor
tic motor symptoms observed in patients with symptoms.
PD. However, numerous nonmotor disorders With a mounting body of evidence suggest-
are prevalent in many patients, even those with ing that neuroinflammation, and microglial
mild or early-stage disease. Study of the neu- activation in particular, is harmful to DA neu-
ropathology of these nonmotor symptoms has rons of the SNpc, it is logical to assume that an
shown that modulation of cholinergic, seroton- anti-inflammatory regimen may protect against
ergic, adenosinergic, glutamatergic, GABAergic, PD-related DA neuron degeneration. However,
and noradrenergic neurotransmitter systems are anti-inflammatory drugs may not provide magi-
involved in the etiology of these symptoms in cal answers for treatment of PD. Although pre-
addition to also having a role in maintenance of clinical trials of anti-inflammatory drugs have
smooth motor function via interactions with the largely been successful, the time of administra-
dopaminergic system. The development of PD tion is a caveat that cannot be ignored. Indeed,
symptoms arises from a comprehensive disrup- most preclinical trials administer drugs prior
tion of normal signal transmission throughout to or concomitantly with neurotoxin insult,
the basal ganglia because of modulation of each an option that, for now, given the absence of
of these types of neurotransmitters. presymptomatic biomarkers, is not available to
Our most significant hindrance in under- PD patients. With the unfortunate fact that PD
standing the complexity of these neurotrans- symptoms do not arise until a majority of DA
mitter systems arises from the fact that our neurons have already degenerated, treatment
knowledge regarding the effect of various param- prior to disease onset creates a clinical treatment
eters on the expression levels of receptors and barrier, an important aspect that needs to be con-
transporters in the human brain is very scarce. sidered when developing future drug therapies
Greater cellular, subcellular, and molecular forPD.
resolution of the expression profiles, linked to Various neurotrophic factors are being stud-
disease-specific changes, would further facilitate ied for therapeutic intervention of DA neuron
the identification of therapeutic approaches for loss in PD in both animal models and clinical
Brain Homeostasis and Parkinsons Disease 497

trials. To date, the most promising of these appear Malliani, A., and Furlan, R. (2007). Early abnor-
to augment the cell loss associated with PD. malities of vascular and cardiac autonomic con-
Additionally, findings from neurotrophic factor trol in Parkinsons disease without orthostatic
studies suggest that a combination of therapeutic hypotension. Hypertension 49, 120126.
approaches using different delivery methods or Barone, P. (2010). Neurotransmission in Parkinsons
endogenous induction may offer the most effec- disease:Beyond dopamine. European Journal of
tive therapy against the devastating neurodegen- Neurology 17, 364376.
erative effectsofPD. Beckman, K.B., and Ames, B.N. (1998). The free
radical theory of aging matures. Physiological
References Reviews 78, 547581.
Acker, B.A., Jacobsen, E.J., Rogers, B.N., Wishka, D.G., Beiske, A.G., Loge, J.H., Ronningen, A., and
Reitz, S.C., Piotrowski, D.W., Myers, J.K., Wolfe, Svensson, E. (2009). Pain in Parkinsons dis-
M.L., Groppi, V.E., Thornburgh, B.A., etal. (2008). ease: Prevalence and characteristics. Pain 141,
Discovery of N-[(3R,5R)-1-azabicyclo[3.2.1] 173177.
oct-3-yl]furo[2,3-c]pyridine-5-carboxamide as Berman, S.B., and Hastings, T.G. (1999). Dopamine
an agonist of the alpha7 nicotinic acetylcholine oxidation alters mitochondrial respiration and
receptor:In vitro and in vivo activity. Bioorganic & induces permeability transition in brain mito-
Medicinal Chemistry Letters 18, 36113615. chondria: Implications for Parkinsons disease.
Ahn, T.B., Kim, S.Y., Kim, J.Y., Park, S.S., Lee, D.S., Min, Journal of Neurochemistry 73, 11271137.
H.J., Kim, Y.K., Kim, S.E., Kim, J.M., Kim, H.J., Bernheimer, H., Birkmayer, W., Hornykiewicz, O.,
et al. (2008). Alpha-synuclein gene duplica- Jellinger, K., and Seitelberger, F. (1973). Brain
tion is present in sporadic Parkinson disease. dopamine and the syndromes of Parkinson
Neurology 70,4349. and Huntington. Clinical, morphological and
Akerud, P., Canals, J.M., Snyder, E.Y., and Arenas, E. neurochemical correlations. Journal of the
(2001). Neuroprotection through delivery of glial Neurological Sciences 20, 415455.
cell line-derived neurotrophic factor by neural Bertoncini, C.W., Fernandez, C.O., Griesinger, C.,
stem cells in a mouse model of Parkinsons dis- Jovin, T.M., and Zweckstetter, M. (2005).
ease. The Journal of Neuroscience 21, 81088118. Familial mutants of alpha-synuclein with
Alam, Z.I., Jenner, A., Daniel, S.E., Lees, A.J., increased neurotoxicity have a destabilized con-
Cairns, N., Marsden, C.D., Jenner, P., and formation. The Journal of Biological Chemistry
Halliwell, B. (1997). Oxidative DNA damage in 280, 3064930652.
the Parkinsonian brain: An apparent selective Betarbet, R., Sherer, T.B., MacKenzie, G.,
increase in 8-hydroxyguanine levels in sub- Garcia-Osuna, M., Panov, A.V., and Greenamyre,
stantia nigra. Journal of Neurochemistry 69, J.T. (2000). Chronic systemic pesticide expo-
11961203. sure reproduces features of Parkinsons disease.
Alex, K.D., and Pehek, E.A. (2007). Pharmacologic Nature Neuroscience 3, 13011306.
mechanisms of serotonergic regulation of dopa- Bibbiani, F., Oh, J.D., Petzer, J.P., Castagnoli, N.,
mine neurotransmission. Pharmacology & Jr., Chen, J.F., Schwarzschild, M.A., and Chase,
Therapeutics 113, 296320. T.N. (2003). A2A antagonist prevents dopamine
Athanassiadou, A., Voutsinas, G., Psiouri, L., Leroy, E., agonist-induced motor complications in animal
Polymeropoulos, M.H., Ilias, A., Maniatis, G.M., models of Parkinsons disease. Exp erimental
and Papapetropoulos, T. (1999). Genetic analy- Neurology 184, 285294.
sis of families with Parkinson disease that carry Bilang-Bleuel, A., Revah, F., Colin, P., Locquet, I.,
the Ala53Thr mutation in the gene encoding Robert, J.J., Mallet, J., and Horellou, P. (1997).
alpha-synuclein. American Journal of Human Intrastriatal injection of an adenoviral vector
Genetics 65, 555558. expressing glial-cell-line-derived neurotrophic
Auluck, P.K., Chan, H.Y., Trojanowski, J.Q., Lee, V.M., factor prevents dopaminergic neuron degenera-
and Bonini, N.M. (2002). Chaperone suppres- tion and behavioral impairment in a rat model of
sion of alpha-synuclein toxicity in a Drosophila Parkinson disease. Proceedings of the National
model for Parkinsons disease. Science 295, Academy of Sciences of the United States of
865868. America 94, 88188823.
Balch, W.E., Morimoto, R.I., Dillin, A., and Kelly, J.W. Bisaglia, M., Mammi, S., and Bubacco, L. (2007).
(2008). Adapting proteostasis for disease inter- Kinetic and structural analysis of the early oxi-
vention. Science 319, 916919. dation products of dopamine: Analysis of the
Barbic, F., Perego, F., Canesi, M., Gianni, M., interactions with alpha-synuclein. The Journal
Biagiotti, S., Costantino, G., Pezzoli, G., Porta, A., of Biological Chemistry 282, 1559715605.
498 Part IV:Homeostatic Therapies

Bjorklund, A., Kirik, D., Rosenblad, C., Brochard, V., Combadiere, B., Prigent, A., Laouar, Y.,
Georgievska, B., Lundberg, C., and Mandel, Perrin, A., Beray-Berthat, V., Bonduelle, O.,
R.J. (2000). Towards a neuroprotective gene Alvarez-Fischer, D., Callebert, J., Launay, J.M.,
therapy for Parkinsons disease:Use of adeno- et al. (2009). Infiltration of CD4+ lymphocytes
virus, AAV and lentivirus vectors for gene into the brain contributes to neurodegenera-
transfer of GDNF to the nigrostriatal system tion in a mouse model of Parkinson disease. The
in the rat Parkinson model. Brain research 886, Journal of Clinical Investigation 119, 182192.
8298. Brueggemann, N., Odin, P., Gruenewald, A., Tadic,
Blochl, A., and Sirrenberg, C. (1996). Neurotrophins V., Hagenah, J., Seidel, G., Lohmann, K., Klein,
stimulate the release of dopamine from rat mes- C., and Djarmati, A. (2008). Re:Alpha-synuclein
encephalic neurons via Trk and p75Lntr recep- gene duplication is present in sporadic Parkinson
tors. The Journal of Biological Chemistry 271, disease. Neurology 71, 1294; author reply1294.
2110021107. Bussell, R. Jr., and Eliezer, D. (2001). Residual
Blum-Degen, D., Muller, T., Kuhn, W., Gerlach, structure and dynamics in Parkinsons
M., Przuntek, H., and Riederer, P. (1995). disease-associated mutants of alpha-synuclein.
Interleukin-1 beta and interleukin-6 are elevated The Journal of Biological Chemistry 276,
in the cerebrospinal fluid of Alzheimers and de 4599646003.
novo Parkinsons disease patients. Neuroscience Calon, F., Dridi, M., Hornykiewicz, O., Bedard, P.J.,
letters 202,1720. Rajput, A.H., and Di Paolo, T. (2004). Increased
Boka, G., Anglade, P., Wallach, D., Javoy-Agid, F., adenosine A2A receptors in the brain of
Agid, Y., and Hirsch, E.C. (1994). Parkinsons disease patients with dyskinesias.
Immunocytochemical analysis of tumor necro- Brain 127, 10751084.
sis factor and its receptors in Parkinsons disease. Calon, F., Rajput, A.H., Hornykiewicz, O., Bedard, P.J.,
Neuroscience Letters 172, 151154. and Di Paolo, T. (2003). Levodopa-induced
Bonifati, V. (2014). Genetics of Parkinsons motor complications are associated with altera-
diseasestate of the art, 2013. Parkinsonism & tions of glutamate receptors in Parkinsons dis-
Related Disorders 20 Suppl 1, S23S28. ease. Neurobiol Dis 14, 404416.
Bonifati, V., Rizzu, P., van Baren, M.J., Schaap, O., Cardoso, S.M., Pereira, C., and Oliveira, R. (1999).
Breedveld, G.J., Krieger, E., Dekker, M.C., Mitochondrial function is differentially affected
Squitieri, F., Ibanez, P., Joosse, M., etal. (2003). upon oxidative stress. Free Radical Biology &
Mutations in the DJ-1 gene associated with Medicine 26,313.
autosomal recessive early-onset parkinsonism. Cecarini, V., Gee, J., Fioretti, E., Amici, M., Angeletti,
Science 299, 256259. M., Eleuteri, A.M., and Keller, J.N. (2007). Protein
Bonifati, V., Rohe, C.F., Breedveld, G.J., Fabrizio, E., De oxidation and cellular homeostasis:Emphasis on
Mari, M., Tassorelli, C., Tavella, A., Marconi, R., metabolism. Biochimica et Biophysica Acta 1773,
Nicholl, D.J., Chien, H.F., etal. (2005). Early-onset 93104.
parkinsonism associated with PINK1 muta- Chartier-Harlin, M.C., Dachsel, J.C., Vilarino-Guell,
tions: Frequency, genotypes, and phenotypes. C., Lincoln, S.J., Lepretre, F., Hulihan, M.M.,
Neurology 65,8795. Kachergus, J., Milnerwood, A.J., Tapia, L.,
Bostantjopoulou, S., Katsarou, Z., Papadimitriou, A., Song, M.S., et al. (2011). Translation initiator
Veletza, V., Hatzigeorgiou, G., and Lees, A. EIF4G1 mutations in familial Parkinson dis-
(2001). Clinical features of parkinsonian patients ease. American Journal of Human Genetics 89,
with the alpha-synuclein (G209A) mutation. 398406.
Movement Disorders 16, 10071013. Chartier-Harlin, M.C., Kachergus, J., Roumier, C.,
Bower, J.H., Maraganore, D.M., McDonnell, S.K., Mouroux, V., Douay, X., Lincoln, S., Levecque, C.,
and Rocca, W.A. (2000). Influence of strict, inter- Larvor, L., Andrieux, J., Hulihan, M., et al.
mediate, and broad diagnostic criteria on the (2004). Alpha-synuclein locus duplication as a
ageand sex-specific incidence of Parkinsons cause of familial Parkinsons disease. Lancet 364,
disease. Movement Disorders 15, 819825. 11671169.
Braak, H., Del Tredici, K., Rub, U., de Vos, R.A., Chen, L., and Feany, M.B. (2005). Alpha-synuclein
Jansen Steur, E.N., and Braak, E. (2003). phosphorylation controls neurotoxicity and
Staging of brain pathology related to sporadic inclusion formation in a Drosophila model
Parkinsons disease. Neurobiology of Aging 24, of Parkinson disease. Nature Neuroscience 8,
197211. 657663.
Brice, A. (2005). Genetics of Parkinsons disease: Chiocco, M.J., Harvey, B.K., Wang, Y., and Hoffer, B.J.
LRRK2 on the rise. Brain 128, 27602762. (2007). Neurotrophic factors for the treatment
Brain Homeostasis and Parkinsons Disease 499

of Parkinsons disease. Parkinsonism & Related Dauer, W., and Przedborski, S. (2003). Parkinsons
Disorders 13 Suppl 3, S321328. disease: Mechanisms and models. Neuron 39,
Choi, J.M., Woo, M.S., Ma, H.I., Kang, S.Y., Sung, Y.H., 889909.
Yong, S.W., Chung, S.J., Kim, J.S., Shin, H.W., Day, M., Wang, Z., Ding, J., An, X., Ingham, C.A.,
Lyoo, C.H., etal. (2008). Analysis of PARK genes Shering, A.F., Wokosin, D., Ilijic, E., Sun, Z.,
in a Korean cohort of early-onset Parkinson dis- Sampson, A.R., et al. (2006). Selective elimina-
ease. Neurogenetics 9, 263269. tion of glutamatergic synapses on striatopallidal
Ciechanover, A., and Brundin, P. (2003). The ubiq- neurons in Parkinson disease models. Nature
uitin proteasome system in neurodegenerative Neuroscience 9, 251259.
diseases:Sometimes the chicken, sometimes the Dehmer, T., Heneka, M.T., Sastre, M., Dichgans, J.,
egg. Neuron 40, 427446. and Schulz, J.B. (2004). Protection by piogli-
Ciechanover, A., Orian, A., and Schwartz, A.L. (2000). tazone in the MPTP model of Parkinsons dis-
Ubiquitin-mediated proteolysis:Biological regu- ease correlates with I kappa B alpha induction
lation via destruction. BioEssays 22, 442451. and block of NF kappa B and iNOS activation. J
Cohen, G. (1984). Oxy-radical toxicity in catechol- Journal of Neurochemistry 88, 494501.
amine neurons. Neurotoxicology 5,7782. Desai Bradaric, B., Patel, A., Schneider, J.A., Carvey,
Colosimo, C., and Craus, A. (2003). Noradrenergic P.M., and Hendey, B. (2012). Evidence for angio-
drugs for levodopa-induced dyskinesia. Clinical genesis in Parkinsons disease, incidental Lewy
Neuropharmacology 26, 299305. body disease, and progressive supranuclear
Colosimo, C., Fabbrini, G., and Berardelli, A. (2006). palsy. J Neural Transm 119,5971.
Drug insight: New drugs in development for Dexter, D.T., Carter, C.J., Wells, F.R., Javoy-Agid, F.,
Parkinsons disease. Nature Clinical Practice Agid, Y., Lees, A., Jenner, P., and Marsden, C.D.
Neurology 2, 600610. (1989). Basal lipid peroxidation in substantia
Conn, P.J., Battaglia, G., Marino, M.J., and Nicoletti, F. nigra is increased in Parkinsons disease. Journal
(2005). Metabotropic glutamate receptors in of Neurochemistry 52, 381389.
the basal ganglia motor circuit. Nature Reviews Djaldetti, R., Yust-Katz, S., Kolianov, V., Melamed, E.,
Neuroscience 6, 787798. and Dabby, R. (2007). The effect of duloxetine on
Connor, B., and Dragunow, M. (1998). The role of primary pain symptoms in Parkinson disease.
neuronal growth factors in neurodegenerative Clinical Neuropharmacology 30, 201205.
disorders of the human brain. Brain Research Driver, J.A., Logroscino, G., Gaziano, J.M., and
Brain Research Reviews 27,139. Kurth, T. (2009). Incidence and remaining life-
Cummings, C.J., Reinstein, E., Sun, Y., Antalffy, B., time risk of Parkinson disease in advanced age.
Jiang, Y., Ciechanover, A., Orr, H.T., Beaudet, A.L., Neurology 72, 432438.
and Zoghbi, H.Y. (1999). Mutation of the Ekstrand, M.I., Terzioglu, M., Galter, D., Zhu, S.,
E6-AP ubiquitin ligase reduces nuclear Hofstetter, C., Lindqvist, E., Thams, S.,
inclusion frequency while accelerating Bergstrand, A., Hansson, F.S., Trifunovic, A.,
polyglutamine-induced pathology in SCA1 mice. et al. (2007). Progressive parkinsonism in mice
Neuron 24, 879892. with respiratory-chain-deficient dopamine neu-
Cummings, C.J., Sun, Y., Opal, P., Antalffy, B., rons. Proceedings of the National Academy of
Mestril, R., Orr, H.T., Dillmann, W.H., and Sciences of the United States of America 104,
Zoghbi, H.Y. (2001). Over-expression of induc- 13251330.
ible HSP70 chaperone suppresses neuropathol- Engele, J., Rieck, H., Choi-Lundberg, D., and Bohn, M.C.
ogy and improves motor function in SCA1 mice. (1996). Evidence for a novel neurotrophic factor
Hum an Molecular Genetics 10, 15111518. for dopaminergic neurons secreted from mesen-
Cunningham, L.A., and Su, C. (2002). Astrocyte cephalic glial cell lines. Journal of Neuroscience
delivery of glial cell line-derived neurotrophic Research 43, 576586.
factor in a mouse model of Parkinsons disease. Farrer, M., Kachergus, J., Forno, L., Lincoln, S.,
Experimental Neurology 174, 230242. Wang, D.S., Hulihan, M., Maraganore, D.,
Czeh, M., Gressens, P., and Kaindl, A.M. (2011). Gwinn-Hardy, K., Wszolek, Z., Dickson, D., etal.
The yin and yang of microglia. Dev elopmental (2004). Comparison of kindreds with parkinson-
Neuroscience 33, 199209. ism and alpha-synuclein genomic multiplica-
Damier, P., Hirsch, E.C., Zhang, P., Agid, Y., and tions. Ann als of Neurology 55, 174179.
Javoy-Agid, F. (1993). Glutathione peroxidase, Farrer, M.J. (2006). Genetics of Parkinson dis-
glial cells and Parkinsons disease. Neuroscience ease: Paradigm shifts and future prospects.
52,16. Nature Reviews Genetics 7, 306318.
500 Part IV:Homeostatic Therapies

Fearnley, J.M., and Lees, A.J. (1991). Ageing and pedigree due to SNCA duplication and triplica-
Parkinsons disease: Substantia nigra regional tion. Neurology 68, 916922.
selectivity. Brain 114 (Pt 5), 22832301. Fujiwara, H., Hasegawa, M., Dohmae, N.,
Fernagut, P.O., and Chesselet, M.F. (2004). Kawashima, A., Masliah, E., Goldberg, M.S.,
Alpha-synuclein and transgenic mouse models. Shen, J., Takio, K., and Iwatsubo, T. (2002).
Neurobiology of Disease 17, 123130. Alpha-synuclein is phosphorylated in synucle-
Ferre, S., Diamond, I., Goldberg, S.R., Yao, L., inopathy lesions. Nature Cell Biology 4, 160164.
Hourani, S.M., Huang, Z.L., Urade, Y., and Galvan, A., and Wichmann, T. (2008).
Kitchen, I. (2007). Adenosine A2A receptors in Pathophysiology of parkinsonism. Clin ical
ventral striatum, hypothalamus and nocicep- Neurophysiology 119, 14591474.
tive circuitry implications for drug addiction, Gash, D.M., Rutland, K., Hudson, N.L., Sullivan,
sleep and pain. Prog ress in Neurobiology 83, P.G., Bing, G., Cass, W.A., Pandya, J.D., Liu,
332347. M., Choi, D.Y., Hunter, R.L., et al. (2008).
Fink, K.B., and Gothert, M. (2007). 5-HT recep- Trichloroethylene: Parkinsonism and com-
tor regulation of neurotransmitter release. plex 1 mitochondrial neurotoxicity. Annals of
Pharmacological Reviews 59, 360417. Neurology 63, 184192.
Firestone, J.A., Smith-Weller, T., Franklin, G., Swanson, Gash, D.M., Zhang, Z., Cass, W.A., Ovadia, A.,
P., Longstreth, W.T., Jr., and Checkoway, H. Simmerman, L., Martin, D., Russell, D., Collins,
(2005). Pesticides and risk of Parkinson disease: F., Hoffer, B.J., and Gerhardt, G.A. (1995).
A population-based case-control study. Archives Morphological and functional effects of intra-
of Neurology 62,9195. nigrally administered GDNF in normal rhesus
Floor, E., and Wetzel, M.G. (1998). Increased pro- monkeys. The Journal of Comparative Neurology
tein oxidation in human substantia nigra pars 363, 345358.
compacta in comparison with basal ganglia and Gash, D.M., Zhang, Z., Ovadia, A., Cass, W.A., Yi, A.,
prefrontal cortex measured with an improved Simmerman, L., Russell, D., Martin, D., Lapchak,
dinitrophenylhydrazine assay. Journal of P.A., Collins, F., etal. (1996). Functional recovery
Neurochemistry 70, 268275. in parkinsonian monkeys treated with GDNF.
Fornai, F., di Poggio, A.B., Pellegrini, A., Ruggieri, S., Nature 380, 252255.
and Paparelli, A. (2007). Noradrenaline in Gerfen, C.R., and Surmeier, D.J. (2011). Modulation
Parkinsons disease: From disease progression of striatal projection systems by dopamine.
to current therapeutics. Current Medicinal Annual Review of Neuroscience 34, 441466.
Chemistry 14, 23302334. Giasson, B.I., Duda, J.E., Murray, I.V., Chen, Q.,
Forno, L.S. (1996). Neuropathology of Parkinsons Souza, J.M., Hurtig, H.I., Ischiropoulos, H.,
disease. Journal of Neuropathology and Trojanowski, J.Q., and Lee, V.M. (2000).
Experimental Neurology 55, 259272. Oxidative damage linked to neurodegeneration
Fox, S.H., Brotchie, J.M., and Lang, A.E. (2008). by selective alpha-synuclein nitration in synucle-
Non-dopaminergic treatments in development inopathy lesions. Science 290, 985989.
for Parkinsons disease. Lancet neurology 7, Giasson, B.I., Murray, I.V., Trojanowski, J.Q., and Lee,
927938. V.M. (2001). A hydrophobic stretch of 12 amino
Frim, D.M., Uhler, T.A., Galpern, W.R., Beal, M.F., acid residues in the middle of alpha-synuclein is
Breakefield, X.O., and Isacson, O. (1994). essential for filament assembly. The Journal of
Implanted fibroblasts genetically engineered to Biological Chemistry 276, 23802386.
produce brain-derived neurotrophic factor pre- Goedert, M. (2001a). Alpha-synuclein and neu-
vent 1-methyl-4-phenylpyridinium toxicity to rodegenerative diseases. Nature Reviews
dopaminergic neurons in the rat. Proceedings of Neuroscience 2, 492501.
the National Academy of Sciences of the United Goedert, M. (2001b). The significance of tau and
States of America 91, 51045108. alpha-synuclein inclusions in neurodegenera-
Fuchs, J., Mueller, J.C., Lichtner, P., Schulte, C., Munz, tive diseases. Current Opinion in Genetics &
M., Berg, D., Wullner, U., Illig, T., Sharma, M., Development 11, 343351.
and Gasser, T. (2009). The transcription factor Grimbergen, Y.A., Langston, J.W., Roos, R.A.,
PITX3 is associated with sporadic Parkinsons and Bloem, B.R. (2009). Postural instability in
disease. Neurobiology of Aging 30, 731738. Parkinsons disease: The adrenergic hypoth-
Fuchs, J., Nilsson, C., Kachergus, J., Munz, M., esis and the locus coeruleus. Expert Review of
Larsson, E.M., Schule, B., Langston, J.W., Neurotherapeutics 9, 279290.
Middleton, F.A., Ross, O.A., Hulihan, M., etal. Halliday, G.M., Macdonald, V., and Henderson, J.M.
(2007). Phenotypic variation in a large Swedish (2005). A comparison of degeneration in motor
Brain Homeostasis and Parkinsons Disease 501

thalamus and cortex between progressive supra- Hunot, S., Boissiere, F., Faucheux, B., Brugg, B.,
nuclear palsy and Parkinsons disease. Brain 128, Mouatt-Prigent, A., Agid, Y., and Hirsch, E.C.
22722280. (1996). Nitric oxide synthase and neuronal vul-
Hamby, M.E., and Sofroniew, M.V. (2010). Reactive nerability in Parkinsons disease. Neuroscience
astrocytes as therapeutic targets for CNS disor- 72, 355363.
ders. Neurotherapeutics 7, 494506. Hyman, C., Hofer, M., Barde, Y.A., Juhasz, M.,
Hanisch, U.K., and Kettenmann, H. (2007). Yancopoulos, G.D., Squinto, S.P., and Lindsay, R.M.
Microglia: Active sensor and versatile effector (1991). BDNF is a neurotrophic factor for dopa-
cells in the normal and pathologic brain. Nature minergic neurons of the substantia nigra. Nature
Neuroscience 10, 13871394. 350, 230232.
Hauser, R.A., Cantillon, M., Pourcher, E., Micheli, F., Hyman, C., Juhasz, M., Jackson, C., Wright, P., Ip,
Mok, V., Onofrj, M., Huyck, S., and Wolski, K. N.Y., and Lindsay, R.M. (1994). Overlapping and
(2011). Preladenant in patients with Parkinsons distinct actions of the neurotrophins BDNF,
disease and motor fluctuations: A phase NT-3, and NT-4/5 on cultured dopaminergic and
2, double-blind, randomised trial. Lancet GABAergic neurons of the ventral mesencepha-
Neurology 10, 221229. lon. The Journal of Neuroscience 14, 335347.
Hauser, R.A., and Schwarzschild, M.A. (2005). Ibanez, P., Bonnet, A.M., Debarges, B., Lohmann, E.,
Adenosine A2A receptor antagonists for Tison, F., Pollak, P., Agid, Y., Durr, A., and
Parkinsons disease: Rationale, therapeutic Brice, A. (2004). Causal relation between
potential and clinical experience. Drugs & Aging alpha-synuclein gene duplication and familial
22, 471482. Parkinsons disease. Lancet 364, 11691171.
Healy, D.G., Abou-Sleiman, P.M., Gibson, J.M., Ibanez, P., Lesage, S., Janin, S., Lohmann, E., Durif,
Ross, O.A., Jain, S., Gandhi, S., Gosal, D., F., Destee, A., Bonnet, A.M., Brefel-Courbon, C.,
Muqit, M.M., Wood, N.W., and Lynch, T. (2004). Heath, S., Zelenika, D., etal. (2009). Alpha-synuclein
PINK1 (PARK6) associated Parkinson disease in gene rearrangements in dominantly inherited par-
Ireland. Neurology 63, 14861488. kinsonism: Frequency, phenotype, and mecha-
Heo, J.Y., Park, J.H., Kim, S.J., Seo, K.S., Han, J.S., nisms. Archives of Neurology 66, 102108.
Lee, S.H., Kim, J.M., Park, J.I., Park, S.K., Lim, Ii, K., Ito, H., Tanaka, K., and Hirano, A. (1997).
K., etal. (2012). DJ-1 null dopaminergic neuronal Immunocytochemical co-localization of the
cells exhibit defects in mitochondrial function proteasome in ubiquitinated structures in neu-
and structure: Involvement of mitochondrial rodegenerative diseases and the elderly. Journal
complex Iassembly. PLoS One 7, e32629. of Neuropathology and Experimental Neurology
Hernan, M.A., Takkouche, B., Caamano-Isorna, F., 56, 125131.
and Gestal-Otero, J.J. (2002). A meta-analysis of Ikeuchi, T., Kakita, A., Shiga, A., Kasuga, K., Kaneko, H.,
coffee drinking, cigarette smoking, and the risk Tan, C.F., Idezuka, J., Wakabayashi, K., Onodera,
of Parkinsons disease. Annals of Neurology 52, O., Iwatsubo, T., etal. (2008). Patients homozy-
276284. gous and heterozygous for SNCA duplication
Hershko, A., and Ciechanover, A. (1998). The ubiq- in a family with parkinsonism and dementia.
uitin system. Annual Review of Biochemistry 67, Archives of Neurology 65, 514519.
425479. Imamura, K., Hishikawa, N., Sawada, M., Nagatsu, T.,
Hirsch, E.C., and Hunot, S. (2009). Neuroinflammation Yoshida, M., and Hashizume, Y. (2003).
in Parkinsons disease: A target for neuroprotec- Distribution of major histocompatibility com-
tion? Lancet Neurology 8, 382397. plex class II-positive microglia and cytokine
Hodgson, R.A., Bedard, P.J., Varty, G.B., Kazdoba, T.M., profile of Parkinsons disease brains. Acta
Di Paolo, T., Grzelak, M.E., Pond, A.J., Hadjtahar, A., Neuropathologica 106, 518526.
Belanger, N., Gregoire, L., etal. (2010). Preladenant, Jankovic, J. (2005). Searching for a relationship
a selective A(2A) receptor antagonist, is active between manganese and welding and Parkinsons
in primate models of movement disorders. disease. Neurology 64, 20212028.
Experimental Neurology 225, 384390. Jankovic, J. (2008). Parkinsons disease: Clinical
Hughes, P.E., Alexi, T., Walton, M., Williams, C.E., features and diagnosis. Journal of Neurology,
Dragunow, M., Clark, R.G., and Gluckman, P.D. Neurosurgery, and Psychiatry 79, 368376.
(1999). Activity and injury-dependent expression Jenner, P. (2003). Oxidative stress in Parkinsons dis-
of inducible transcription factors, growth factors ease. Annals of Neurology 53 Suppl 3, S26S36;
and apoptosis-related genes within the central discussion S36S28.
nervous system. Progress in Neurobiology 57, Kanda, T., Tashiro, T., Kuwana, Y., and Jenner, P.
421450. (1998). Adenosine A2A receptors modify motor
502 Part IV:Homeostatic Therapies

function in MPTP-treated common marmosets. Schols, L., and Riess, O. (1998). Ala30Pro muta-
Neuroreport 9, 28572860. tion in the gene encoding alpha-synuclein in
Karachi, C., Grabli, D., Bernard, F.A., Tande, D., Parkinsons disease. Nature genetics 18, 106108.
Wattiez, N., Belaid, H., Bardinet, E., Prigent, A., Kumar, K.R., Weissbach, A., Heldmann, M., Kasten,
Nothacker, H.P., Hunot, S., et al. (2010). M., Tunc, S., Sue, C.M., Svetel, M., Kostic, V.S.,
Cholinergic mesencephalic neurons are involved Segura-Aguilar, J., Ramirez, A., et al. (2012).
in gait and postural disorders in Parkinson dis- Frequency of the D620N mutation in VPS35 in
ease. The Journal of Clinical Investigation 120, Parkinson disease. Archives of neurology 69,
27452754. 13601364.
Khandelwal, P.J., Herman, A.M., and Moussa, C.E. Lange, K.W., Kornhuber, J., and Riederer, P. (1997).
(2011). Inflammation in the early stages of Dopamine/glutamate interactions in Parkinsons
neurodegenerative pathology. Journal of disease. Neuroscience and Biobehavioral
Neuroimmunology 238,111. Reviews 21, 393400.
Khateeb, S., Flusser, H., Ofir, R., Shelef, I., Narkis, G., Langston, J.W., Ballard, P., Tetrud, J.W., and Irwin,
Vardi, G., Shorer, Z., Levy, R., Galil, A., Elbedour, K., I. (1983). Chronic Parkinsonism in humans due
etal. (2006). PLA2G6 mutation underlies infan- to a product of meperidine-analog synthesis.
tile neuroaxonal dystrophy. American Journal of Science 219, 979980.
Human Genetics 79, 942948. Laurie, C., Reynolds, A., Coskun, O., Bowman, E.,
Ki, C.S., Stavrou, E.F., Davanos, N., Lee, W.Y., Chung, Gendelman, H.E., and Mosley, R.L. (2007).
E.J., Kim, J.Y., and Athanassiadou, A. (2007). CD4+ T cells from Copolymer-1 immunized
The Ala53Thr mutation in the alpha-synuclein mice protect dopaminergic neurons in the
gene in a Korean family with Parkinson disease. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
Clinical Genetics 71, 471473. model of Parkinsons disease. Journal of
Kish, S.J., Tong, J., Hornykiewicz, O., Rajput, A., Neuroimmunology 183,6068.
Chang, L.J., Guttman, M., and Furukawa, Y. Lee, H.J., Khoshaghideh, F., Patel, S., and Lee, S.J.
(2008). Preferential loss of serotonin markers in (2004). Clearance of alpha-synuclein oligo-
caudate versus putamen in Parkinsons disease. meric intermediates via the lysosomal degrada-
Brain 131, 120131. tion pathway. The Journal of Neuroscience 24,
Kitada, T., Asakawa, S., Hattori, N., Matsumine, 18881896.
H., Yamamura, Y., Minoshima, S., Yokochi, M., Lees, A.J., Hardy, J., and Revesz, T. (2009). Parkinsons
Mizuno, Y., and Shimizu, N. (1998). Mutations in disease. Lancet 373, 20552066.
the parkin gene cause autosomal recessive juve- Lennox, G., Lowe, J., Morrell, K., Landon, M., and
nile parkinsonism. Nature 392, 605608. Mayer, R.J. (1989). Anti-ubiquitin immunocy-
Klein, C., Djarmati, A., Hedrich, K., Schafer, N., tochemistry is more sensitive than conventional
Scaglione, C., Marchese, R., Kock, N., Schule, techniques in the detection of diffuse Lewy body
B., Hiller, A., Lohnau, T., et al. (2005). PINK1, disease. Journal of Neurology, Neurosurgery,
Parkin, and DJ-1 mutations in Italian patients and Psychiatry 52,6771.
with early-onset parkinsonism. European Leroy, E., Anastasopoulos, D., Konitsiotis, S.,
Journal of Human Genetics 13, 10861093. Lavedan, C., and Polymeropoulos, M.H. (1998a).
Klein, C., and Westenberger, A. (2012). Genetics Deletions in the Parkin gene and genetic het-
of Parkinsons disease. Cold Spring Harbor erogeneity in a Greek family with early onset
Perspectives in Medicine 2, a008888. Parkinsons disease. Human Genetics 103,
Knott, C., Stern, G., and Wilkin, G.P. (2000). 424427.
Inflammatory regulators in Parkinsons dis- Leroy, E., Boyer, R., Auburger, G., Leube, B., Ulm,
ease:iNOS, lipocortin-1, and cyclooxygenases-1 G., Mezey, E., Harta, G., Brownstein, M.J.,
and -2. Molecular and Cellular Neurosciences Jonnalagada, S., Chernova, T., etal. (1998b). The
16, 724739. ubiquitin pathway in Parkinsons disease. Nature
Kopito, R.R. (2000). Aggresomes, inclusion bodies 395, 451452.
and protein aggregation. Trends in Cell Biology Lesage, S., Durr, A., Tazir, M., Lohmann, E.,
10, 524530. Leutenegger, A.L., Janin, S., Pollak, P., and
Kordower, J.H. (2003). In vivo gene delivery of Brice, A. (2006). LRRK2 G2019S as a cause of
glial cell line-derived neurotrophic factor for Parkinsons disease in North African Arabs. The
Parkinsons disease. Annals of Neurology 53 New England Journal of Medicine 354, 422423.
Suppl 3, S120S132; discussion S132S124. LeWitt, P.A., Guttman, M., Tetrud, J.W., Tuite,
Kruger, R., Kuhn, W., Muller, T., Woitalla, D., P.J., Mori, A., Chaikin, P., and Sussman, N.M.
Graeber, M., Kosel, S., Przuntek, H., Epplen, J.T., (2008). Adenosine A2A receptor antagonist
Brain Homeostasis and Parkinsons Disease 503

istradefylline (KW-6002) reduces off time in Massano, J., and Bhatia, K.P. (2012). Clinical approach
Parkinsons disease: A double-blind, random- to Parkinsons disease: Features, diagnosis, and
ized, multicenter clinical trial (6002-US-005). principles of management. Cold Spring Harbor
Annals of Neurology 63, 295302. Perspectives in Medicine 2, a008870.
Li, Y., Tomiyama, H., Sato, K., Hatano, Y., Yoshino, Maswood, N., Grondin, R., Zhang, Z., Stanford, J.A.,
H., Atsumi, M., Kitaguchi, M., Sasaki, S., Surgener, S.P., Gash, D.M., and Gerhardt, G.A.
Kawaguchi, S., Miyajima, H., et al. (2005). (2002). Effects of chronic intraputamenal infu-
Clinicogenetic study of PINK1 mutations in sion of glial cell line-derived neurotrophic factor
autosomal recessive early-onset parkinsonism. (GDNF) in aged Rhesus monkeys. Neurobiology
Neurology 64, 19551957. of Aging 23, 881889.
Lin, L.F., Doherty, D.H., Lile, J.D., Bektesh, S., and Mazurov, A., Klucik, J., Miao, L., Phillips, T.Y.,
Collins, F. (1993). GDNF:Aglial cell line-derived Seamans, A., Schmitt, J.D., Hauser, T.A.,
neurotrophic factor for midbrain dopaminergic Johnson, R.T., Jr., and Miller, C. (2005).
neurons. Science 260, 11301132. 2-(Arylmethyl)-3-substituted quinuclidines
Lohmann, E., Periquet, M., Bonifati, V., Wood, as selective alpha 7 nicotinic receptor ligands.
N.W., De Michele, G., Bonnet, A.M., Fraix, V., Bioorganic & Medicinal Chemistry Letters 15,
Broussolle, E., Horstink, M.W., Vidailhet, M., 20732077.
et al. (2003). How much phenotypic variation McGeer, P.L., Itagaki, S., Boyes, B.E., and McGeer,
can be attributed to parkin genotype? Annals of E.G. (1988). Reactive microglia are positive for
Neurology 54, 176185. HLA-DR in the substantia nigra of Parkinsons
Lotharius, J., and Brundin, P. (2002). Pathogenesis and Alzheimers disease brains. Neurology 38,
of Parkinsons disease: Dopamine, vesicles and 12851291.
alpha-synuclein. Nature Reviews Neuroscience McNeill, T.H., Brown, S.A., Rafols, J.A., and
3, 932942. Shoulson, I. (1988). Atrophy of medium spiny
Lowe, J., McDermott, H., Landon, M., Mayer, Istriatal dendrites in advanced Parkinsons dis-
R.J., and Wilkinson, K.D. (1990). Ubiquitin ease. Brain Research 455, 148152.
carboxyl-terminal hydrolase (PGP 9.5) is selec- Mirza, B., Hadberg, H., Thomsen, P., and Moos, T.
tively present in ubiquitinated inclusion bodies (2000). The absence of reactive astrocytosis is
characteristic of human neurodegenerative dis- indicative of a unique inflammatory process in
eases. The Journal of Pathology 161, 153160. Parkinsons disease. Neuroscience 95, 425432.
Lull, M.E., and Block, M.L. (2010). Microglial Mogi, M., Harada, M., Kondo, T., Riederer, P.,
activation and chronic neurodegeneration. Inagaki, H., Minami, M., and Nagatsu, T.
Neurotherapeutics 7, 354365. (1994a). Interleukin-1 beta, interleukin-6, epi-
Lwin, A., Orvisky, E., Goker-Alpan, O., dermal growth factor and transforming growth
LaMarca, M.E., and Sidransky, E. (2004). factor-alpha are elevated in the brain from par-
Glucocerebrosidase mutations in subjects kinsonian patients. Neuroscience Letters 180,
with parkinsonism. Molecular Genetics and 147150.
Metabolism 81,7073. Mogi, M., Harada, M., Riederer, P., Narabayashi, H.,
Mandemakers, W., Morais, V.A., and De Strooper, Fujita, K., and Nagatsu, T. (1994b). Tumor necro-
B. (2007). A cell biological perspective on mito- sis factor-alpha (TNF-alpha) increases both in
chondrial dysfunction in Parkinson disease and the brain and in the cerebrospinal fluid from
other neurodegenerative diseases. Journal of Cell parkinsonian patients. Neuroscience Letters 165,
Science 120, 17071716. 208210.
Maraganore, D.M., de Andrade, M., Lesnick, T.G., Mogi, M., Kondo, T., Mizuno, Y., and Nagatsu, T.
Strain, K.J., Farrer, M.J., Rocca, W.A., Pant, (2007). p53 protein, interferon-gamma, and
P.V., Frazer, K.A., Cox, D.R., and Ballinger, D.G. NF-kappaB levels are elevated in the parkinso-
(2005). High-resolution whole-genome asso- nian brain. Neuroscience Letters 414,9497.
ciation study of Parkinson disease. American Moreau, C., Delval, A., Defebvre, L., Dujardin, K.,
Journal of Human Genetics 77, 685693. Duhamel, A., Petyt, G., Vuillaume, I., Corvol,
Marsden, C.D. (1982). The mysterious motor func- J.C., Brefel-Courbon, C., Ory-Magne, F., et al.
tion of the basal ganglia:The Robert Wartenberg (2012). Methylphenidate for gait hypokinesia
Lecture. Neurology 32, 514539. and freezing in patients with Parkinsons disease
Marsden, C.D. (1983). Neuromelanin and Parkinsons undergoing subthalamic stimulation: A multi-
disease. Journal of Neural Transmission centre, parallel, randomised, placebo-controlled
Supplementum 19, 121141. trial. Lancet Neurology 11, 589596.
504 Part IV:Homeostatic Therapies

Morelli, M., Di Paolo, T., Wardas, J., Calon, F., Xiao, D., Paisan-Ruiz, C., Jain, S., Evans, E.W., Gilks, W.P.,
and Schwarzschild, M.A. (2007). Role of ade- Simon, J., van der Brug, M., Lopez de Munain,
nosine A2A receptors in parkinsonian motor A., Aparicio, S., Gil, A.M., Khan, N., etal. (2004).
impairment and l-DOPA-induced motor compli- Cloning of the gene containing mutations
cations. Progress in Neurobiology 83, 293309. that cause PARK8-linked Parkinsons disease.
Morgan, N.V., Westaway, S.K., Morton, J.E., Gregory, A., Neuron 44, 595600.
Gissen, P., Sonek, S., Cangul, H., Coryell, J., Paisan-Ruiz, C., Li, A., Schneider, S.A., Holton, J.L.,
Canham, N., Nardocci, N., etal. (2006). PLA2G6, Johnson, R., Kidd, D., Chataway, J., Bhatia, K.P.,
encoding a phospholipase A2, is mutated in neu- Lees, A.J., Hardy, J., et al. (2012). Widespread
rodegenerative disorders with high brain iron. Lewy body and tau accumulation in childhood
Nature Genetics 38, 752754. and adult onset dystonia-parkinsonism cases
Murer, M.G., Yan, Q., and Raisman-Vozari, R. (2001). with PLA2G6 mutations. Neurobiology of Aging
Brain-derived neurotrophic factor in the control 33, 814823.
human brain, and in Alzheimers disease and Pan, T., Kondo, S., Le, W., and Jankovic, J. (2008).
Parkinsons disease. Progress in Neurobiology The role of autophagy-lysosome pathway in neu-
63, 71124. rodegeneration associated with Parkinsons dis-
Nagatsu, T. (1997). Isoquinoline neurotoxins in the ease. Brain 131, 19691978.
brain and Parkinsons disease. Neuroscience Pankratz, N., Pauciulo, M.W., Elsaesser, V.E., Marek,
Research 29, 99111. D.K., Halter, C.A., Wojcieszek, J., Rudolph, A.,
Nishioka, K., Hayashi, S., Farrer, M.J., Singleton, Shults, C.W., Foroud, T., and Nichols, W.C.
A.B., Yoshino, H., Imai, H., Kitami, T., Sato, K., (2006). Mutations in DJ-1 are rare in familial
Kuroda, R., Tomiyama, H., etal. (2006). Clinical Parkinson disease. Neuroscience Letters 408,
heterogeneity of alpha-synuclein gene duplica- 209213.
tion in Parkinsons disease. Annals of Neurology Parker, W.D., Jr., and Swerdlow, R.H. (1998).
59, 298309. Mitochondrial dysfunction in idiopathic
Nonaka, T., Iwatsubo, T., and Hasegawa, M. (2005). Parkinson disease. American Journal of Human
Ubiquitination of alpha-synuclein. Biochemistry Genetics 62, 758762.
44, 361368. Patel, N.K., Bunnage, M., Plaha, P., Svendsen,
ONeill, M.J., and Dix, S. (2007). AMPA receptor C.N., Heywood, P., and Gill, S.S. (2005).
potentiators as cognitive enhancers. IDrugs 10, Intraputamenal infusion of glial cell line-derived
185192. neurotrophic factor in PD:Atwo-year outcome
ONeill, M.J., and Witkin, J.M. (2007). AMPA recep- study. Annals of Neurology 57, 298302.
tor potentiators:Application for depression and Perry, E.K., Curtis, M., Dick, D.J., Candy, J.M., Atack,
Parkinsons disease. Current Drug Targets 8, J.R., Bloxham, C.A., Blessed, G., Fairbairn,
603620. A., Tomlinson, B.E., and Perry, R.H. (1985).
Olanow, C.W., and Schapira, A.H. (2013). Therapeutic Cholinergic correlates of cognitive impair-
prospects for Parkinson disease. Annals of ment in Parkinsons disease:Comparisons with
Neurology 74, 337347. Alzheimers disease. Journal of Neurology,
Ouattara, B., Gregoire, L., Morissette, M., Gasparini, Neurosurgery, and Psychiatry 48, 413421.
F., Vranesic, I., Bilbe, G., Johns, D.R., Rajput, Pertovaara, A. (2006). Noradrenergic pain modula-
A., Hornykiewicz, O., Rajput, A.H., et al. tion. Progress in Neurobiology 80,5383.
(2011). Metabotropic glutamate receptor type Peterson, A.L., and Nutt, J.G. (2008). Treatment
5 in levodopa-induced motor complications. of Parkinsons disease with trophic factors.
Neurobiology of Aging 32, 12861295. Neurotherapeutics 5, 270280.
Ozelius, L.J., Senthil, G., Saunders-Pullman, R., Piccini, P., Burn, D.J., Ceravolo, R., Maraganore, D.,
Ohmann, E., Deligtisch, A., Tagliati, M., Hunt, and Brooks, D.J. (1999). The role of inheritance
A.L., Klein, C., Henick, B., Hailpern, S.M., etal. in sporadic Parkinsons disease:Evidence from a
(2006). LRRK2 G2019S as a cause of Parkinsons longitudinal study of dopaminergic function in
disease in Ashkenazi Jews. The New England twins. Annals of Neurology 45, 577582.
Journal of Medicine 354, 424425. Pollack, A.E. (2001). Anatomy, physiology, and
Paisan-Ruiz, C., Bhatia, K.P., Li, A., Hernandez, pharmacology of the basal ganglia. Neurologic
D., Davis, M., Wood, N.W., Hardy, J., Houlden, Clinics 19, 523534,v.
H., Singleton, A., and Schneider, S.A. (2009). Polymeropoulos, M.H., Higgins, J.J., Golbe, L.I.,
Characterization of PLA2G6 as a locus for Johnson, W.G., Ide, S.E., Di Iorio, G., Sanges, G.,
dystonia-parkinsonism. Annals of Neurology Stenroos, E.S., Pho, L.T., Schaffer, A.A., et al.
65,1923. (1996). Mapping of a gene for Parkinsons
Brain Homeostasis and Parkinsons Disease 505

disease to chromosome 4q21-q23. Science 274, in MPTP-lesioned rats. European Journal of


11971199. Pharmacology 560, 163175.
Polymeropoulos, M.H., Lavedan, C., Leroy, E., Ide, Remy, P., Doder, M., Lees, A., Turjanski, N., and
S.E., Dehejia, A., Dutra, A., Pike, B., Root, H., Brooks, D. (2005). Depression in Parkinsons
Rubenstein, J., Boyer, R., etal. (1997). Mutation disease: Loss of dopamine and noradrenaline
in the alpha-synuclein gene identified in fami- innervation in the limbic system. Brain 128,
lies with Parkinsons disease. Science 276, 13141322.
20452047. Reynolds, A.D., Stone, D.K., Hutter, J.A., Benner,
Pourcher, E., Fernandez, H.H., Stacy, M., Mori, E.J., Mosley, R.L., and Gendelman, H.E. (2010).
A., Ballerini, R., and Chaikin, P. (2012). Regulatory T cells attenuate Th17 cell-mediated
Istradefylline for Parkinsons disease patients nigrostriatal dopaminergic neurodegeneration
experiencing motor fluctuations: Results of in a model of Parkinsons disease. Journal of
the KW-6002-US-018 study. Parkinsonism & Immunology 184, 22612271.
Related Disorders 18, 178184. Reynolds, A.D., Stone, D.K., Mosley, R.L., and
Powers, E.T., Morimoto, R.I., Dillin, A., Kelly, J.W., Gendelman, H.E. (2009). Proteomic studies of
and Balch, W.E. (2009). Biological and chemical nitrated alpha-synuclein microglia regulation
approaches to diseases of proteostasis deficiency. by CD4+CD25+ T cells. Journal of Proteome
Annual Review of Biochemistry 78, 959991. Research 8, 34973511.
Prediger, R.D. (2010). Effects of caffeine in Richard, I.H. (2005). Anxiety disorders in Parkinsons
Parkinsons disease: From neuroprotection to disease. Advances in Neurology 96,4255.
the management of motor and non-motor symp- Richard, I.H., McDermott, M.P., Kurlan, R.,
toms. Journal of Alzheimers Disease 20 Suppl 1, Lyness, J.M., Como, P.G., Pearson, N., Factor,
S205S220. S.A., Juncos, J., Serrano Ramos, C., Brodsky,
Priyadarshi, A., Khuder, S.A., Schaub, E.A., and M., et al. (2012). A randomized, double-blind,
Priyadarshi, S.S. (2001). Environmental risk fac- placebo-controlled trial of antidepressants in
tors and Parkinsons disease: A meta-analysis. Parkinson disease. Neurology 78, 12291236.
Environmental Research 86, 122127. Rinne, J.O., Ma, S.Y., Lee, M.S., Collan, Y., and
Puschmann, A., Ross, O.A., Vilarino-Guell, C., Roytta, M. (2008). Loss of cholinergic neu-
Lincoln, S.J., Kachergus, J.M., Cobb, S.A., rons in the pedunculopontine nucleus in
Lindquist, S.G., Nielsen, J.E., Wszolek, Z.K., Parkinsons disease is related to disabil-
Farrer, M., etal. (2009). A Swedish family with de ity of the patients. Parkinsonism & Related
novo alpha-synuclein A53T mutation: Evidence Disorders 14, 553557.
for early cortical dysfunction. Parkinsonism & Rodriguez, M.C., Obeso, J.A., and Olanow, C.W.
Related Disorders 15, 627632. (1998). Subthalamic nucleus-mediated excitotox-
Quinn, L.P., Crook, B., Hows, M.E., Vidgeon-Hart, icity in Parkinsons disease:Atarget for neuro-
M., Chapman, H., Upton, N., Medhurst, A.D., protection. Annals of Neurology 44, S175S188.
and Virley, D.J. (2008). The PPARgamma ago- Rodriguez-Oroz, M.C., Jahanshahi, M., Krack, P.,
nist pioglitazone is effective in the MPTP mouse Litvan, I., Macias, R., Bezard, E., and Obeso,
model of Parkinsons disease through inhibi- J.A. (2009). Initial clinical manifestations of
tion of monoamine oxidase B. British Journal of Parkinsons disease: Features and pathophysi-
Pharmacology 154, 226233. ological mechanisms. Lancet Neurology 8,
Rajput, A.H., Sitte, H.H., Rajput, A., Fenton, M.E., 11281139.
Pifl, C., and Hornykiewicz, O. (2008). Globus Rogaeva, E., Johnson, J., Lang, A.E., Gulick, C.,
pallidus dopamine and Parkinson motor sub- Gwinn-Hardy, K., Kawarai, T., Sato, C., Morgan, A.,
types: Clinical and brain biochemical correla- Werner, J., Nussbaum, R., etal. (2004). Analysis
tion. Neurology 70, 14031410. of the PINK1 gene in a large cohort of cases with
Rees, K., Stowe, R., Patel, S., Ives, N., Breen, K., Clarke, Parkinson disease. Archives of Neurology 61,
C.E., and Ben-Shlomo, Y. (2011). Non-steroidal 18981904.
anti-inflammatory drugs as disease-modifying Rosin, D.L., Robeva, A., Woodard, R.L., Guyenet,
agents for Parkinsons disease: Evidence from P.G., and Linden, J. (1998). Immunohistochemical
observational studies. Cochrane Database of localization of adenosine A2A receptors in
Systematic Reviews, CD008454. the rat central nervous system. The Journal of
Reksidler, A.B., Lima, M.M., Zanata, S.M., Machado, Comparative Neurology 401, 163186.
H.B., da Cunha, C., Andreatini, R., Tufik, S., Ross, C.A., and Poirier, M.A. (2004). Protein aggre-
and Vital, M.A. (2007). The COX-2 inhibi- gation and neurodegenerative disease. Nature
tor parecoxib produces neuroprotective effects Medicine 10 Suppl, S10S17.
506 Part IV:Homeostatic Therapies

Sandy, M.S., Armstrong, M., Tanner, C.M., Daly, Simon, D.K., Lin, M.T., and Pascual-Leone, A. (2002).
A.K., Di Monte, D.A., Langston, J.W., and Idle, J.R. Nature versus nurture and incompletely
(1996). CYP2D6 allelic frequencies in penetrant mutations. Journal of Neurology,
young-onset Parkinsons disease. Neurology 47, Neurosurgery, and Psychiatry 72, 686689.
225230. Singleton, A.B., Farrer, M., Johnson, J., Singleton, A.,
Sandy, M.S., Langston, J.W., Smith, M.T., and Di Hague, S., Kachergus, J., Hulihan, M.,
Monte, D.A. (1993). PCR analysis of platelet Peuralinna, T., Dutra, A., Nussbaum, R., et al.
mtDNA:Lack of specific changes in Parkinsons (2003). alpha-Synuclein locus triplication causes
disease. Movement disorders 8,7482. Parkinsons disease. Science 302,841.
Scatton, B., Javoy-Agid, F., Rouquier, L., Dubois, B., Smith, J.A., Das, A., Ray, S.K., and Banik, N.L. (2012).
and Agid, Y. (1983). Reduction of cortical dopa- Role of pro-inflammatory cytokines released
mine, noradrenaline, serotonin and their metab- from microglia in neurodegenerative diseases.
olites in Parkinsons disease. Brain Research 275, Brain Research BULLETIN 87,1020.
321328. Sofroniew, M.V., and Vinters, H.V. (2010).
Schapira, A.H., Cooper, J.M., Dexter, D., Clark, J.B., Astrocytes:Biology and pathology. Acta neuro-
Jenner, P., and Marsden, C.D. (1990). pathologica 119,735.
Mitochondrial complex I deficiency in Spillantini, M.G., Crowther, R.A., Jakes, R., Cairns, N.J.,
Parkinsons disease. Journal of Neurochemistry Lantos, P.L., and Goedert, M. (1998). Filamentous
54, 823827. alpha-synuclein inclusions link multiple system
Schiffmann, S.N., Fisone, G., Moresco, R., Cunha, R.A., atrophy with Parkinsons disease and dementia
and Ferre, S. (2007). Adenosine A2A recep- with Lewy bodies. Neuroscience Letters 251,
tors and basal ganglia physiology. Progress in 205208.
Neurobiology 83, 277292. Spira, P.J., Sharpe, D.M., Halliday, G., Cavanagh, J.,
Schiffmann, S.N., Jacobs, O., and Vanderhaeghen, J.J. and Nicholson, G.A. (2001). Clinical and patho-
(1991). Striatal restricted adenosine A2 receptor logical features of a Parkinsonian syndrome in a
(RDC8) is expressed by enkephalin but not by family with an Ala53Thr alpha-synuclein muta-
substance P neurons: An in situ hybridization tion. Annals of Neurology 49, 313319.
histochemistry study. Journal of Neurochemistry Stephens, B., Mueller, A.J., Shering, A.F., Hood, S.H.,
57, 10621067. Taggart, P., Arbuthnott, G.W., Bell, J.E., Kilford, L.,
Schlossmacher, M.G., Frosch, M.P., Gai, W.P., Kingsbury, A.E., Daniel, S.E., et al. (2005).
Medina, M., Sharma, N., Forno, L., Ochiishi, T., Evidence of a breakdown of corticostriatal con-
Shimura, H., Sharon, R., Hattori, N., etal. (2002). nections in Parkinsons disease. Neuroscience
Parkin localizes to the Lewy bodies of Parkinson 132, 741754.
disease and dementia with Lewy bodies. The Stone, D.K., Reynolds, A.D., Mosley, R.L., and
American Journal of Pathology 160, 16551667. Gendelman, H.E. (2009). Innate and adaptive
Sian, J., Dexter, D.T., Lees, A.J., Daniel, S., Agid, Y., immunity for the pathobiology of Parkinsons
Javoy-Agid, F., Jenner, P., and Marsden, C.D. disease. Antioxidants & Redox Signaling 11,
(1994). Alterations in glutathione levels in 21512166.
Parkinsons disease and other neurodegenera- Swanson, C.R., Joers, V., Bondarenko, V., Brunner, K.,
tive disorders affecting basal ganglia. Annals of Simmons, H.A., Ziegler, T.E., Kemnitz, J.W.,
Neurology 36, 348355. Johnson, J.A., and Emborg, M.E. (2011). The
Sidhu, A., Wersinger, C., Moussa, C.E., and Vernier, PPAR-gamma agonist pioglitazone modu-
P. (2004). The role of alpha-synuclein in both lates inflammation and induces neuropro-
neuroprotection and neurodegeneration. Annals tection in parkinsonian monkeys. Journal of
of the New York Academy of Sciences 1035, Neuroinflammation8,91.
250270. Tanner, C.M. (1992). Epidemiology of Parkinsons
Sidransky, E., Nalls, M.A., Aasly, J.O., Aharon-Peretz, J., disease. Neurologic Clinics 10, 317329.
Annesi, G., Barbosa, E.R., Bar-Shira, A., Berg, D., Tanner, C.M., Ottman, R., Goldman, S.M., Ellenberg, J.,
Bras, J., Brice, A., et al. (2009). Multicenter Chan, P., Mayeux, R., and Langston, J.W. (1999).
analysis of glucocerebrosidase mutations in Parkinson disease in twins: An etiologic study.
Parkinsons disease. The New England Journal of JAMA 281, 341346.
Medicine 361, 16511661. Tansey, M.G., and Goldberg, M.S. (2010).
Siegel, G.J., and Chauhan, N.B. (2000). Neurotrophic Neuroinflammation in Parkinsons disease: Its
factors in Alzheimers and Parkinsons disease role in neuronal death and implications for ther-
brain. Brain Research Brain Research Reviews apeutic intervention. Neurobiology of Disease
33, 199227. 37, 510518.
Brain Homeostasis and Parkinsons Disease 507

Teismann, P., and Ferger, B. (2001). Inhibition of the Disease. Journal of Clinical Neuroscience 12,
cyclooxygenase isoenzymes COX-1 and COX-2 739743.
provide neuroprotection in the MPTP-mouse Valente, E.M., Abou-Sleiman, P.M., Caputo, V.,
model of Parkinsons disease. Synapse 39, Muqit, M.M., Harvey, K., Gispert, S., Ali, Z., Del
167174. Turco, D., Bentivoglio, A.R., Healy, D.G., et al.
Teismann, P., Tieu, K., Choi, D.K., Wu, D.C., Naini, A., (2004a). Hereditary early-onset Parkinsons dis-
Hunot, S., Vila, M., Jackson-Lewis, V., and ease caused by mutations in PINK1. Science 304,
Przedborski, S. (2003a). Cyclooxygenase-2 is 11581160.
instrumental in Parkinsons disease neurodegen- Valente, E.M., Salvi, S., Ialongo, T., Marongiu, R.,
eration. Proceedings of the National Academy of Elia, A.E., Caputo, V., Romito, L., Albanese, A.,
Sciences of the United States of America 100, Dallapiccola, B., and Bentivoglio, A.R. (2004b).
54735478. PINK1 mutations are associated with sporadic
Teismann, P., Vila, M., Choi, D.K., Tieu, K., Wu, D.C., early-onset parkinsonism. Annals of Neurology
Jackson-Lewis, V., and Przedborski, S. (2003b). 56, 336341.
COX-2 and neurodegeneration in Parkinsons Varani, K., Vincenzi, F., Tosi, A., Gessi, S., Casetta, I.,
disease. Annals of the New York Academy of Granieri, G., Fazio, P., Leung, E., MacLennan, S.,
Sciences 991, 272277. Granieri, E., etal. (2010). A2A adenosine recep-
Thomas, B., and Beal, M.F. (2007). Parkinsons dis- tor overexpression and functionality, as well as
ease. Human Molecular Genetics 16 Spec No. 2, TNF-alpha levels, correlate with motor symp-
R183R194. toms in Parkinsons disease. FASEB Journal 24,
Tolosa, E., Wenning, G., and Poewe, W. (2006). 587598.
The diagnosis of Parkinsons disease. Lancet Vernino, S., Hopkins, S., and Wang, Z. (2009).
Neurology 5,7586. Autonomic ganglia, acetylcholine receptor
Tomac, A., Lindqvist, E., Lin, L.F., Ogren, S.O., antibodies, and autoimmune ganglionopathy.
Young, D., Hoffer, B.J., and Olson, L. (1995). Autonomic Neuroscience: Basic & Clinical
Protection and repair of the nigrostriatal dopa- 146,37.
minergic system by GDNF in vivo. Nature 373, Vilarino-Guell, C., Wider, C., Ross, O.A.,
335339. Dachsel, J.C., Kachergus, J.M., Lincoln, S.J.,
Troiano, A.R., Cazeneuve, C., Le Ber, I., Bonnet, A.M., Soto-Ortolaza, A.I., Cobb, S.A., Wilhoite, G.J.,
Lesage, S., and Brice, A. (2008). Re: Bacon, J.A., et al. (2011). VPS35 mutations in
Alpha-synuclein gene duplication is present in Parkinson disease. American Journal of Human
sporadic Parkinson disease. Neurology 71, 1295; Genetics 89, 162167.
author reply1295. Voges, D., Zwickl, P., and Baumeister, W. (1999). The
Tsang, A.H., and Chung, K.K. (2009). Oxidative 26S proteasome:Amolecular machine designed
and nitrosative stress in Parkinsons disease. for controlled proteolysis. Annual Review of
Biochimica et Biophysica Acta 1792, 643650. Biochemistry 68, 10151068.
Tucci, A., Charlesworth, G., Sheerin, U.M., Plagnol, V., Warrick, J.M., Chan, H.Y., Gray-Board, G.L., Chai, Y.,
Wood, N.W., and Hardy, J. (2012). Study of the Paulson, H.L., and Bonini, N.M. (1999).
genetic variability in a Parkinsons Disease Suppression of polyglutamine-mediated neu-
gene:EIF4G1. Neuroscience Letters 518,1922. rodegeneration in Drosophila by the molecu-
Turrens, J.F. (2003). Mitochondrial formation of reac- lar chaperone HSP70. Nature Genetics 23,
tive oxygen species. The Journal of Physiology 425428.
552, 335344. Whitehead, J.P. (2011). Diabetes: New conduc-
Uchiyama, T., Ikeuchi, T., Ouchi, Y., Sakamoto, M., tors for the peroxisome proliferator-activated
Kasuga, K., Shiga, A., Suzuki, M., Ito, M., Atsumi, receptor gamma (PPARgamma) orchestra. The
T., Shimizu, T., etal. (2008). Prominent psychiat- International Journal of Biochemistry & Cell
ric symptoms and glucose hypometabolism in a Biology 43, 10711074.
family with a SNCA duplication. Neurology 71, Williams-Gray, C.H., Foltynie, T., Lewis, S.J., and
12891291. Barker, R.A. (2006). Cognitive deficits and psy-
Uhl, G.R., Walther, D., Mash, D., Faucheux, B., and chosis in Parkinsons disease:Areview of patho-
Javoy-Agid, F. (1994). Dopamine transporter physiology and therapeutic options. CNS Drugs
messenger RNA in Parkinsons disease and 20, 477505.
control substantia nigra neurons. Annals of Wirdefeldt, K., Gatz, M., Schalling, M., and Pedersen,
Neurology 35, 494498. N.L. (2004). No evidence for heritability of
Vajda, F.J., and Solinas, C. (2005). Current approaches Parkinson disease in Swedish twins. Neurology
to management of depression in Parkinsons 63, 305311.
508 Part IV:Homeostatic Therapies

Witte, M.E., Geurts, J.J., de Vries, H.E., van der Valk, and Parkinson diseases. American Journal of
P., and van Horssen, J. (2010). Mitochondrial dys- Clinical Pathology 129, 526529.
function:Apotential link between neuroinflam- Zhang, Z., Miyoshi, Y., Lapchak, P.A., Collins, F.,
mation and neurodegeneration? Mitochondrion Hilt, D., Lebel, C., Kryscio, R., and Gash, D.M.
10, 411418. (1997). Dose response to intraventricular glial
Yamada, M., Iwatsubo, T., Mizuno, Y., and Mochizuki, cell line-derived neurotrophic factor adminis-
H. (2004). Overexpression of alpha-synuclein in tration in parkinsonian monkeys. The Journal of
rat substantia nigra results in loss of dopaminer- Pharmacology and Experimental Therapeutics
gic neurons, phosphorylation of alpha-synuclein 282, 13961401.
and activation of caspase-9: Resemblance to Zheng, J.S., Tang, L.L., Zheng, S.S., Zhan, R.Y.,
pathogenetic changes in Parkinsons disease. Zhou, Y.Q., Goudreau, J., Kaufman, D., and
Journal of Neurochemistry 91, 451461. Chen, A.F. (2005). Delayed gene therapy of glial
Yoritaka, A., Hattori, N., Uchida, K., Tanaka, cell line-derived neurotrophic factor is effica-
M., Stadtman, E.R., and Mizuno, Y. cious in a rat model of Parkinsons disease.
(1996). Immunohistochemical detection of Brain Research Molecular Brain Research 134,
4-hydroxynonenal protein adducts in Parkinson 155161.
disease. Proceedings of the National Academy Zhou, F.M., Wilson, C., and Dani, J.A. (2003).
of Sciences of the United States of America 93, Muscarinic and nicotinic cholinergic mecha-
26962701. nisms in the mesostriatal dopamine systems. The
Zarranz, J.J., Alegre, J., Gomez-Esteban, J.C., Lezcano, Neuroscientist 9,2336.
E., Ros, R., Ampuero, I., Vidal, L., Hoenicka, Zimprich, A., Benet-Pages, A., Struhal, W., Graf,
J., Rodriguez, O., Atares, B., et al. (2004). The E., Eck, S.H., Offman, M.N., Haubenberger, D.,
new mutation, E46K, of alpha-synuclein causes Spielberger, S., Schulte, E.C., Lichtner, P., et al.
Parkinson and Lewy body dementia. Annals of (2011). A mutation in VPS35, encoding a sub-
Neurology 55, 164173. unit of the retromer complex, causes late-onset
Zesiewicz, T.A., Baker, M.J., Wahba, M., and Parkinson disease. American Journal of Human
Hauser, R.A. (2003). Autonomic Nervous System Genetics 89, 168175.
Dysfunction in Parkinsons Disease. Current Zimprich, A., Biskup, S., Leitner, P., Lichtner, P.,
Treatment Options in Neurology 5, 149160. Farrer, M., Lincoln, S., Kachergus, J., Hulihan,
Zhang, J., Sokal, I., Peskind, E.R., Quinn, J.F., M., Uitti, R.J., Calne, D.B., etal. (2004). Mutations
Jankovic, J., Kenney, C., Chung, K.A., Millard, in LRRK2 cause autosomal-dominant parkin-
S.P., Nutt, J.G., and Montine, T.J. (2008). CSF sonism with pleomorphic pathology. Neuron 44,
multianalyte profile distinguishes Alzheimer 601607.
27
Brain Homeostasis and Addiction
L E T I S H A R . W YAT T

ADDICTION OVERVIEW common to have an array of reactions to hallu-


Drug addiction, or substance dependence, is a cinogenic drugs such as ecstasy and lysergic acid
national epidemic. In the United States, costs diethylamide (LSD), including rapid shifts in
related to illicit drug use reaches nearly $200 bil- emotion, enhanced tactile sensitivity, and impul-
lion annually (National Drug Intelligence Center, sivity. Finally, inhalants typically cause stimula-
2011). As reported by the US National Drug tion and the loss of inhibition (National Institute
Intelligence Center, these direct and indirect costs on Drug Abuse,2012).
cover crime, health, and a decline in workforce
productivity and ultimately affect individuals MESOCORTICOLIMBIC
from many cultural, racial, and socioeconomic R E WA R D C I R C U I T R Y
backgrounds. By diagnostic standards, addiction INADDICTION
is characterized as the transition from casual Several anatomical structures have been identi-
to compulsive drug use whereby an individual fied as neural substrates of drugs of abuse, includ-
spends more of his or her time in drug-related ing the ventral tegmental area (VTA), prefrontal
versus non-drug-related activities (Zernig etal., cortex (PFC), nucleus accumbens (NAc), amyg-
2007). Within the past three decades, clinicians dala, and hippocampus. These areas of the brain
have modified the definition of substance use in have distinct features and interconnected nuclei
the Diagnostic and Statistical Manual of Mental where cellular modulation by drugs may elicit
Disorders, distinguishing between those that obvious or subtle changes in behavior. Studies in
abuse and those who are dependent (American this area have shown that the VTA and NAc are
Psychiatric Association, 2000). Many physi- likely mediators of addictive behaviors. The VTA,
cians and behavioral researchers in the field a midbrain structure distinct from the substantia
agree about the stages of the drug addiction nigra, is one of the most commonly studied brain
cycle: acquisition, tolerance, dependence, with- areas with respect to drug reward. The VTA is
drawal (abstinence), and relapse (Le Moal and largely comprised of dopamine (DA)-releasing
Koob, 2007). These distinct stages are thought to cells. As theorized, the normal physiological
evolve from initial sensitivity and then tolerance function of these dopaminergic neurons include
acquired during drug use (Vetulani,2001). (a) the release of DA as a function of pleasure
All drugs fall into one of six classifica- or liking, (b) a mechanism to aid in associa-
tions: (a) narcotics, (b) stimulants, (c) depres- tive learning by encoding predictions for future
sants, (d) cannabinoids, (e) hallucinogens, and rewards that are necessary for survival through
(f) inhalants (Feltenstein and See, 2008). Each firing rates and the release of DA, and (c)incen-
class comes with its own behavioral characteri- tive salience or wanting (Ikemoto, 2007). Drugs
zation, for example narcotics such as heroin are of abuse are capable of corrupting the system
classically known to produce feelings of eupho- by stimulating the release or slowing down the
ria (National Drug Intelligence Center, 2011). reuptake of DA in the VTA, thereby eliciting a
Stimulants such as nicotine and cocaine, on the high or extreme pleasure response that becomes
other hand, are known for eliciting feelings of reinforcing for drug use (Ikemoto, 2007). The
exhilaration (National Institute on Drug Abuse, NAc is designated as having a specific role in
2012). For sensations of relaxation, drug users directly mediating behavioral output by control-
typically turn to depressants and cannabinoids ling information flow to the thalamus, striatum,
(e.g., ethanol and marijuana, respectively). It is and PFC via the ventral pallidum (Pierce and
510 Part IV:Homeostatic Therapies

Kumaresan, 2006). Some investigations differen- behavior, no one model can encompass all aspects
tiate the role of the accumbens shell versus the of the disease. Nonetheless, proper selection of
core for the rewarding effect of drugs, with the the animal model to be used in preclinical testing
shell as the most involved in limbic processes and is essential to ensure validity of the interpreta-
the core the most involved in motor functions tion of data with respect to the human condition.
(Ikemoto, 2007; Pierce and Kumaresan,2006). Based on the hypotheses of the investigator and
Historically, research has centered on the the drug to be studied, procedures utilized for
involvement of the mesocorticolimbic reward modeling addiction in animals willassess
pathways, which specifically describes afferent
connections between the VTA and regions such 1. Acute drug effect and sensitivity via
as the NAc, amygdala, hippocampus, and medial experimenter administration
PFC (Pierce and Kumaresan, 2006). This path- 2. Acquisition (intoxication/binging),
way is involved in the emotional, motivational, often through self-administration using
and contextual influence on behaviors (Pierce intracranial self-stimulation or operant
and Kumaresan, 2006). Drugs of abuse can affect chambers
both pathways, thereby altering behavior and 3. Motivation using operant chambers to
creating a vicious cycle that results in addic- illuminate the reinforcing effects of the
tion. Some studies argue that local drug admin- drug and investigate how hard the animal
istration into the NAc does not increase DA but will work for thedrug
rather may enhance the upstream firing rate of 4. Acute withdrawal following abrupt
dopaminergic neurons in the VTA (Pierce and sobriety after several sessions of drug
Kumaresan,2006). exposure by monitoring for shaking,
The mesocorticolimbic areas are enriched ptosis, chewing, and heightened anxiety.
with receptors for DA, -amino butyric acid Alternatively, withdrawal can be studied
(GABA), glutamate, serotonin, and endoge- using the conditioned place avoidance
nous opioids (Volkow et al., 2012), all of which test, which is achieved by providing
have been identified as key regulators of drug the drug in a defined environment over
responses and addictive behavior. Notably, DA, several exposures and then allowing the
GABA, and glutamate have been linked to the animal to experience drug withdrawal in
actions of depressants, stimulants, and narcot- the same environment. The animal will
ics. Central opioid receptors (i.e., mu, kappa, and display conditioned place avoidance and
delta)typically those found in the NAcare thus will spend less time in the environ-
responsible for behavioral effects of opioid drugs ment associated with drug withdrawal
such as morphine or heroin (Lutz and Kieffer, (Heidbreder,2011).
2013). Although mechanistically the differ- 5. Reinstatement/craving and relapse, using
ent classes of drugs have associations with dif- a drug/cue reinstatement procedure
ferent receptors in the central nervous system whereby animals are drug exposed over
(CNS), they share some common neurobiological many sessions to a conditioned stimu-
underpinnings. For instance, as mentioned ear- lus, then are given an extinction period
lier, all drugs of abuse alter the firing of neuronal during which the drug and conditioned
dopaminergic cells and elevate basal DA levels. stimulus are absent until drug-seeking
Moreover, genetic analyses have also shown that behavior is gone, followed by re-exposure
individual differences in gene expression and to either the drug or a conditioned stimu-
function of various neurotransmitters correlate lus; behavior is then compared to initial
with vulnerability to drug self-administration drug responses (Heidbreder,2011).
(for a review, see Goldman etal.,2005).
The subjective effect of drugs is comparatively
DRUG ADDICTION MODELING more challenging to test in animals without their
INANIMALS verbal feedback. Thus behavioral tests have been
Animal modeling of drug addiction is a not an designed to assess the rewarding effects of drugs.
easy task, though it is critical for the understand- The conditioned place preference (CPP) para-
ing of the molecular basis of the disease and the digm measures the amount of time spent in a
development of new therapies. Due to the com- specific compartment where an animal has been
plexity of the progressive stages in addictive trained to associate the effects of a particular
Brain Homeostasis and Addiction 511

test substance. When the amount of time spent is critical for mechanisms underlying addic-
in the conditioned compartment of the appa- tive behavior, and altered homeostasis can have
ratus increases, then it can be inferred that the serious consequences regarding substance use.
test substance was rewarding (Becker etal., 2012; Several systems exist and work together to main-
Heidbreder, 2011). Tests for drug discrimina- tain the ideal brain environment for neuronal
tion also measure reward. For this, animals are functions. The release of excitatory or inhibi-
trained to associate the feelings provided from an tory neurotransmitters plays an integral role
injected drug or non-drug (typically saline) with in maintaining the localized synaptic milieu.
food delivered through a lever press. Subsequent Neuromodulation by molecules that are neither
test substances administered to the animal will directly excitatory nor inhibitory, but rather alter
cause the animal to press either the drug-food the response of groups of neurons over a long dis-
lever or the saline-food lever, indicating the sub- tance, is also important in brain homeostasis (Ito
jective effect of the test substance compared to and Schuman, 2008). Additionally, inflammatory
the drug (Heidbreder,2011). immune responses, endocrine functions, energy
From a psychological perspective, drug balance, and neurotrophic factors contribute to
addiction is a disorder of compulsivity and/or maintaining brain homeostasis. Currently, there
impulsivity. Not to be confused with impulsivity is some debate as to whether inherent altered
(or the tendency toward rapid, unplanned reac- brain homeostasis proceeds drug addiction or,
tions to internal or external stimuli), compulsiv- alternatively, drug exposure begets dysfunctional
ity is defined as the performance of repetitive homeostatic responses. Using recent neurophar-
behaviors to reduce or prevent anxiety or dis- macological and behavioral studies, here we dis-
tress (Grant and Potenza, 2006). Both are related cuss the various systems and their involvement
to the craving and intoxication/binge stages of in the compulsive intake and addictive patterns
addiction (Koob and Le Moal, 2008). To model of the different classes ofdrugs.
these endophenotypes in animals, the delayed
discounting and the 5-choice serial reaction Neuromodulators
time tasks are often utilized. These tests are able Dopamine Homeostasis
to evaluate impulsive choice and determine any We begin with DA homeostatic control of
impairments in inhibitory control in animals addictive behavior, as the majority of addiction
and can be used as a measure of risk for increased research emphasizes a key role for DA in these
drug-seeking and binge-like patterns of addic- processes. However, because DA has been so
tion (Heidbreder,2011). extensively covered in addiction research (see
This chapter provides a review of homeostatic Blum etal., 2012), a brief overview of the involve-
mechanisms involved in drug addiction and ment of DA in drug reward and addiction is
offers current examples in the literature where introduced in this section. DA has a long history
investigators have used the aforementioned ani- of being considered a molecular target, since all
mal models to describe the involvement of vari- drugs of abuse enhance brain levels of this mol-
ous homeostatic systems in drug addiction. The ecule. DA is a neuromodulator responsible for
mechanisms underlying the progression from both motor and cognitive functions. Produced by
substance use to abuse and dependence are dis- neurons in the midbrain, namely the substantia
cussed in light of brain homeostasis. The chap- nigra and VTA, firing patterns of DA-releasing
ter concludes with a brief description of current neurons and increases in extracellular DA in the
pharmacological strategies and also nonpharma- NAc is thought to be a way in which the brain
cological approaches involving the modulation encodes natural reward (Ikemoto, 2007). In both
of brain homeostasis as an alternative treatment humans and rodents, DA receptors (D1-like and
strategy for addiction. D2-like) and the DA transporter are responsible
for regulating DA homeostasis, and each is sus-
O V E R V I E W O F H O M E O S TAT I C ceptible to expression changes following drug
CONTROL SYSTEMS exposure (Becker etal.,2012).
R E L E VA N T T O A D D I C T I O N
As outlined in the introductory portion of this Adenosine Homoeostasis
book, brain homeostatic mechanisms are impor- Adenosine is a potent homeostatic regulator of
tant for normal and optimal function of human network function that activates A1, A2A, A 2B, and
physiology. Regulation of brain homeostasis A3 adenosine receptors, which are expressed
512 Part IV:Homeostatic Therapies

throughout the brain (Diogenes etal., 2014). Intra- break addictive behavior. Interestingly, in 2004
and extracellular adenosine levels are rapidly and 2009 the antiepileptic drugs Gabapentin
regulated by the astrocyte-based enzyme adeno- and Vigabatrin were highlighted for their poten-
sine kinase, responsible for adenosine metabo- tial to treat addiction (Brodie et al., 2009; Raby
lism to adenosine 5-monophosphate (Boison, and Coomaraswamy, 2004). Although both
2013). In both humans and animals, A1 receptor drugs are known to increase the level of circu-
activation has been linked to alterations in glu- lating GABA neurotransmitter, the underlying
tamate and DA activity in the brain. Adenosine anti-addictive mechanism has not been fully
agonists reduce striatal DA levels and inhibit cor- identified. Nonetheless, evidence from the litera-
tical glutamate function (Dohrman etal., 1997). ture support the idea that adenosine homeostasis
Glutamate, DA, and other neurotransmitters plays a critical role in the regulation of addictive
modulated by adenosine are known mediators behavior.
of addictive behaviors (Lingford-Hughes et al.,
2010; Sebastiao and Ribeiro,2009). Serotonin Homoeostasis
In support of a glial basis of addiction, rodent Serotonin (5-hydroxytryptamine; 5-HT) func-
studies demonstrate an upregulation of reactive tions as both a neurotransmitter and a neuro-
astrocytes following drug administration, with- modulator with important regulatory roles in the
drawal, and abstinence (Baydas and Tuzcu, 2005; CNS for mood, sleep, and some cognitive pro-
Fattore et al., 2002; Kelso et al., 2011). Several cesses such as learning and memory, as well as
lines of evidence support the notion that astro- intestinal motility (Berger etal., 2009; Gonzalez
gliosis increases adenosine kinase, resulting in et al., 2013). 5-HT is produced by serotonergic
adenosine hypofunction (Boison, 2011; Li et al., neurons originating in the raphe nucleus and
2008). Notably, addictive behavior is a recog- released via vesicles into the synaptic cleft, target-
nized comorbidity in epilepsy, a condition char- ing most areas of the brain. The action of seroto-
acterized by pathological hypofunction of the nin is dependent on 5-HT receptors, and the level
endogenous anticonvulsant adenosine (Boison, of this molecule is modulated by reuptake via the
2011). In addition, seizures are a common side monoamine transporter (5-HTT). 5-HT recep-
effect for some recreational drugs and a symptom tors modulate both excitatory and inhibitory neu-
of drug withdrawal. rotransmission and may alter the release of other
Recent studies have more directly addressed neurotransmitters (e.g., GABA, glutamate, and
a role for adenosine in mechanisms underly- DA) and various hormones (Nichols and Nichols,
ing drug addiction and/or responses to drugs of 2008). This system has been exploited by clini-
abuse. Wu etal. (2013) assessed changes in brain cians for its antidepressant effects since the dis-
adenosine following the administration of mor- covery of selective serotonin reuptake inhibitors
phine. The findings from this study indicate that (SSRIs). Also, because inhibition of serotonergic
adenosine levels are significantly reduced during activity enhances mood and reduces consumma-
opiate dependency and withdrawal in C57BL/6J tory behaviors, researchers have focused on the
mice compared to 129Sv1 mice, which do not role of serotonin in psychostimulant, depressant,
develop opiate physical dependence to morphine and hallucinogenicdrugs.
under the same conditions. Additionally, opiate The 5-HTT has received the most atten-
withdrawal behavior was modified after genetic tion in investigations related to drugs of abuse.
alteration of adenosine kinase expression in However, recent studies on the G-protein cou-
mice (Wu et al., 2013). Based on their findings, pled receptors 5-HT1B, -HT2A, -HT2C, and the
and in conjunction with previous studies in the ligand-gated ion channel 5-HT3, have also impli-
literature, the authors suggest that adenosines cated these receptor subtypes in mediating some
involvement in opiate addiction and withdrawal of the behavioral effects of drugs like ecstasy
is in part linked to its modulation of glutamate (or 3,4-methylenedioxymethamphetamine;
release and the action of other excitatory neuro- MDMA), alcohol, cocaine, and methampheta-
transmitters in the brainstem (Wu etal.,2013). mine (for a review, see Higgins and Fletcher,
Such reports suggest that disruption of aden- 2003). Studies by Bengel etal. (1998) have shown
osine homeostasis, possibly induced by glial acti- that administration of ecstasy and ampheta-
vation, might be implicated in the development mine causes an increase in serotonin levels in
of addictive behavior and that therapeutic adeno- mice. Additionally, by making use of 5-HTT
sine augmentation might be a novel strategy to homozygous and heterozygous knockout mice,
Brain Homeostasis and Addiction 513

the authors provided support that acute behav- (Parrott, 2002). This is not surprising considering
ioral responses such as increased locomotor the involvement of serotonin in biological pro-
activity are likely mediated through the 5-HTT, cesses, including impulsivity, anxiety, depression,
an MDMA-induced behavioral effect that was and schizophrenia (Parrott, 2002). The working
eliminated in the 5-HTT null animals. These hypothesis is that serotonin overexcitability leads
data implicate a regulatory role for serotonin in to a greater release of serotonin from neurons
the behavioral response to ecstasy that may serve in the raphe nucleus projecting to mesocorti-
as a new therapeutic target. colimbic areas that mediate impulsive behavior
SSRIs such as fluoxetine elevate the level of and decision-making (Dalley and Roiser, 2012).
serotonin in the brain and are capable of modify- While several studies provide evidence that sero-
ing behavioral reactions to cocaine and alcohol. tonin and certain receptor subtypes are capable
For instance, Burmeister and colleagues (2003) of mediating genetic risk for addiction (Herman
report that fluoxetine can dose-dependently and Balogh, 2012), increases in impulsivity are
attenuate cocaine reinstatement after extin- proposed to be particularly problematic for drug
guishing cocaine-seeking behavior in rats. More users due to impulsive behavior during relapse
recently, Sawyer et al. (2012) evaluated these after drug withdrawal. However, self-reports of
findings in rhesus macaques after clinical tri- impulsivity from drug users have inconclusively
als using SSRIs failed to consistently reduce linked this cognitive deficit to addiction, where
cocaine abuse. The authors found that after 4 some studies report no differences in impulsivity
weeks of daily cocaine self-administration fol- scores between ecstasy users and nave controls
lowed by a 2-week extinction period, 10 mg/kg and others suggest that ecstasy use induces neu-
fluoxetine attenuated cocaine reinstatement. This rocognitive deficits in impulsivity (Rogers etal.,
was also the first study to demonstrate a reduc- 2009; Schilt etal., 2010). Notably, these types of
tion in drug-induced DA overflow by fluoxetine, cognitive hypotheses are difficult to test and are
an effect that Sawyer and colleagues attribute to limited by not knowing the inherent responses in
desensitization of 5-HT2A receptors. Similarly, a participants and whether higher basal impulsiv-
low although behaviorally relevant dose of eth- ity is a risk factor for drug use or, rather, if ecstasy
anol (2.5 g/kg) injected directly into the CA3 use either directly or indirectly enhances impul-
region of the hippocampus in mice produces a sivity (Halpern etal.,2011).
significant reduction in local serotonin clearance
(Daws etal., 2006). These authors also found that Endocannabinoid Homeostasis
inactivation of 5-HTT, as investigated via 5-HTT Endocannabinoids are a family of lipids capa-
knockout mice, results in a potentiation of the ble of modulating neuronal synapses (Pertwee,
sedative/hypnotic effects of ethanol. Similar 2006). Such endogenous cannabinoids as ara-
findings were not achieved when administering chidonoylethanolamide (or anandamide) and
fluoxetine to C57BL/6J mice, demonstrating that 2-arachidonoylglycerol (2-AG) interact with
the genetic loss of 5-HTT provides a more robust the central G-protein coupled CB1 cannabinoid
alteration in ethanol-induced behavior than non- receptors to mediate several neurobiological pro-
selective pharmacological inactivation of 5-HTT cesses (Pacher et al., 2006). The CB1 receptor is
(Daws etal.,2006). thought to largely influence chemical messengers
Although the psychological effects of such as GABA and glutamate, thereby affect-
serotonin-altering drugs are relatively short, ing learning and memory (Wang and Lupica,
the spike in serotonergic activity is thought to 2014). Notably, these receptors have been recog-
be neurotoxic, and these acute neurobiological nized to be sensitive to the exogenous cannabi-
changes can be very detrimental, as evidenced noid 9-tetrahydrocannabinol (9-THC) and
in long-term ecstasy users who reportedly suffer are involved with the drug effects of marijuana
from cognitive and memory impairments and (Clapper etal.,2009).
psychiatric problems (Parrott, 2002). Much of As with several other abused drugs, THC is
the problem regarding alterations in serotonin capable of enhancing DA neurotransmission in
homeostasis, particularly excess, has to do with the mesolimbic reward pathway. Intense inter-
an increase in impulsivity. Users of drugs that est has been placed on investigating the mecha-
enhance serotonergic function are often found nism for these actions. With regard to marijuana,
to have comorbid psychiatric disorders such as THC-induced activation of CB1 receptors elicits
increased general anxiety and impulsiveness GABA inhibition in the NAc, allowing for an
514 Part IV:Homeostatic Therapies

increase in circulating DA levels and eventu- withdrawal, they represent a reasonable new
ally leading to drug tolerance and addiction target for the treatment of opiate dependence
(Clapper etal., 2009; Rashidy-Pour etal., 2013). (Ramesh etal.,2011).
Rashidy-Pour et al. tested various synthetic
cannabinoids in rats and found that intracra- Neurotransmitters and Receptors
nial injection of CB1 receptor agonists into the Glutamate Homoeostasis
VTA could potentiate the rewarding properties Glutamate is the main excitatory neurotransmit-
of morphine in a CPP test. Interestingly, these ter in the CNS. Ionotropic glutamate receptors,
agonists independently produced rewarding located throughout the brain at synapses, bind
effects in the animals. It has been well docu- glutamate and allow for the influx of calcium
mented that endocannabinoids released from (Dingledine et al., 1999). The balance between
the postsynaptic neuron activate CB1 receptors release and elimination of glutamate between
located presynaptically and so are thought to act ionotropic receptors (-amino-3-hydroxy-5
in a retrograde fashion (Pertwee, 2006). In this -methyl-4-isoxazolepropionic acid [AMPA] and
way modulation of the endocannabinoid sys- N-methyl-D-aspartic acid [NMDA]), metabo-
tem typically participates synergistically with tropic glutamate receptors (mGluR), and glial
drugs of abuse to cause an increased reward cells is essential for the regulation of extracel-
response and reinforcement. For example, sev- lular glutamate. Glutamate neurotransmission is
eral studies report elevated endocannabinoids of importance in addiction, largely with regard
following administration of alcohol, nicotine, to synaptic plasticity associated with drug rein-
and morphine. Collectively, these studies show forcement and reinstatement processes (Koob
reductions in drug self-administration, reward, and Le Moal, 2008). Pre- and postsynaptic glu-
and withdrawal in CB1 receptor knockout mice tamate receptors located in the NAc are involved
and following administration of CB1 receptor in communication to the PFC, an area that is
antagonists (see Pacher etal., 2006, for a detailed heavily involved in generating learned behaviors
review). and behavioral adaptations to the environment
Studies that investigate the changes in anan- (Kalivas, 2009). Such corticostriatal connections
damide and 2-AG homeostasis in the various are altered in the presence of abused drugs. In the
stages of drug addiction target enzymatic deg- literature, studies involving glutamate predomi-
radation of these proteins via fatty acid amid nately investigate narcotic (opiates, e.g., heroin
hydrolysis (FAAH) and monoacylglycerol lipase and morphine) and stimulant (cocaine)drugs.
(MAGL), respectively (Maldonado et al., 2013). Reinforcement studies have demonstrated
Consensus from a review of the literature sug- that alterations in glutamate tone are part of the
gests that in FAAH-deficient mice, the endo- process of the escalation of drug-taking. While
cannabinoid system is differentially involved in cocaine facilitates glutamate release, morphine
consumption, reward, and withdrawal of etha- reduces the amount of extracellular glutamate
nol and nicotine. Pharmacological inhibition of in some mesocorticolimbic areas of the brain
FAAH activity by URB597 shows a more com- (i.e., VTA, NAc, dorsal striatum). Analyses of
plex effect, whereby cue-induced reinstatement AMPA and NMDA receptor expression in rats
of nicotine seeking was reduced in rats but not after cocaine self-administration revealed upreg-
self-administration (Forget etal., 2009). Because ulation of AMPA receptors in the VTA as an
of the critical central role of 2-AG in energy effect of drug reinforcement (Choi et al., 2011).
homeostasis, adverse effects are associated with Mechanistically, one theory involves changes
genetic manipulation of MAGL (Jung et al., at dendritic spines. Morphological changes in
2012), however, blockade with an MAGL inhibi- spine diameter and density via expression of
tor (JZL184) in morphine-dependent mice was proteins for spine formation as well as expres-
able to reduce precipitated withdrawal induced sion of AMPA receptors and synaptic glutamate
by naloxone (Ramesh etal., 2011). Selective inhi- transporters lead to increased excitability of
bition of MAGL over 2-AG completely alleviated the brain regions involved, triggering cravings
all measured signs of morphine withdrawal in and eventually drug tolerance (Choi etal., 2011;
these mice (Ramesh et al., 2011). Moreover, the Kalivas, 2009). Glutamate neurotransmission
inhibitors did not substitute for THC or other has also been shown to modulate reinstatement
cannabinoid receptor agonists, and, while pro- after drug withdrawal. Circulating glutamate lev-
viding great face validity as a model for morphine els and AMPA receptor expression is reportedly
Brain Homeostasis and Addiction 515

elevated in the PFC and NAc during cue-induced in nature, and accordingly GABA has a major
drug-seeking following cocaine, heroin, and role in modulating DA neurotransmission in this
methamphetamine self-administration, increas- part of the brain. Some drugs specifically target
ing the reactivity of this area thereby inten- the GABAergic neurons in the VTA and cause a
sifying cravings and susceptibility to relapse reduction in their activity. Consequently, GABA
(Conrad et al., 2008; LaLumiere and Kalivas, inhibition of DA release is attenuated, thereby
2008; Parsegian and See, 2014). Recent stud- enhancing the rewarding properties of such drugs
ies have also pointed to an adenosine and DA (Addolorato etal.,2012).
receptor component for regulating the func- GABA A receptor physiology is quite complex.
tion and expression of AMPA receptors for drug The various subunits (i.e., , , , and ) come
reinstatement. Specifically, Hobson et al. (2013) together in many different combinations to form
suggest that stimulating adenosine A1 receptors a pentameric receptor (Uusi-Oukari and Korpi,
reduces AMPA receptor function via an antago- 2010). Each subunit, with its subtypes, has been
nistic interaction with D1 receptors. The authors implicated in drug addiction in various ways,
results support the idea of the importance of and at this time a specific molecular target, even
AMPA receptor signaling in the NAc for cocaine within this family of receptors, has not be iden-
reinstatement. tified. However, as evidenced by the number of
Glutamate homeostasis is also highly regu- publications in databases of scientific journals,
lated by transporters and exchangers on neurons it is clear that the GABAA 1 subunit receives
and nearby glial cells that modulate its release the most attention in addiction research, likely
and elimination. The excitatory amino acid trans- due to the ubiquitous expression of this subtype.
porters (EAAT1-5) are responsible for glutamate Most studies of the GABA A receptor have investi-
transport in many different brain regions, includ- gated alcohol addiction, primarily due to a more
ing the striatum, hippocampus, and cerebellum. comprehensive understanding of GABA receptor
Astrocytic EAAT2, also known as GLT-1 (glu- physiology as well as behavioral similarities in
tamate transporter-1), accounts for the major- the sedative effects of ethanol and CNS depres-
ity of glutamate transporter activity in the CNS sive effects after GABA receptor activation.
(Robinson, 1998). Studies have shown that GLT-1 The role of GABA A receptors is complex and
mRNA is decreased in morphine-dependent rats differs depending on the stage of addiction and
and that activation of the glutamate transporter the specific brain region that is targeted by the
reduces morphine tolerance and dependence drug. For instance, the expression of accumbal
(Wu etal., 2008). Yan and colleagues (2013a) also GABA A receptors has been demonstrated to be
investigated the mechanism for this and hypoth- important for ethanol intake and reinforcement
esized that aquaporin-4 (AQP4), which coexists in rodents where greater expression results in
with GLT-1, is deficient in rodents, leading to more intake (Nie et al., 2011). In addition, this
reduced opiate dependence. Interestingly, GLT-1 receptor subtype is downregulated with chronic
expression and glutamate reuptake was reduced ethanol exposure and also has been shown to be
in wild-type mice treated with morphine, an endocytosed in the hippocampus during with-
effect that was not observed in AQP4-deficient drawal (Sarviharju etal., 2006; Shen etal., 2011).
mice (Yan et al., 2013a). The authors conclude Similarly, benzodiazepines elicit disinhibition of
that regulation of glutamate homeostasis through GABA to increase the amount of circulating DA.
transporter (i.e., GLT-1 and AQP4) expression Several groups have proposed this mechanism, as
may be a novel strategy for treating addiction. benzodiazepine drugs are allosteric modulators
of GABAA receptors and have been extensively
GABA Homoeostasis studied because of their high addictive liability
GABA homeostasis has been studied extensively in (e.g., alprazolam or Xanax and diazepam or
the context of sedative drugs such as barbiturates, Valium). Benzodiazepines cause sleepiness and
benzodiazepines, and alcohol. Both the ionotropic have been used to treat anxiety, convulsions,
GABAA and metabotropic GABAB receptors and sleep disorders. Tolerance and dependence
respond to GABA neurotransmitter and are capa- quickly develops, and this is likely due in part to
ble of eliciting inhibitory responses from neurons the drugs strong interaction with GABAA recep-
by either preventing firing of action potentials or tors (Tan etal.,2010).
neurotransmitter release, respectively. Neurons in Drug responses and addictive behavior medi-
the VTA are primarily DAergic and GABAergic ated by GABAB receptors seemingly function
516 Part IV:Homeostatic Therapies

a little differently. Findings in the literature postsynaptically, neuronal forms are found both
point toward a potential for GABAB receptors to presynaptically where they can influence the
reduce drug self-administration. The use of the release of multiple neurotransmitters and post-
GABAB receptor agonist baclofen has shed some synaptically (for classical neurotransmission;
light on the potential mechanism for its sup- Hurst etal., 2013). In the case of nicotinic recep-
pression of drug-related behaviors. Presumably, tors, the high permeability to calcium allows for
GABAB receptors located on DAergic cell bod- great influence over intracellular signaling cas-
ies in the VTA cause an inhibition of DA release cades and thus gene activation and neurotrans-
to the NAc (Filip and Frankowska, 2008). Using mitter release (Hurst etal.,2013).
rats and an intracranial self-stimulation para- The cholinergic system has been shown to
digm, Slattery etal. (2005) showed that baclofen be targeted by several different drugs of abuse.
dose-dependently elevated the intracranial The majority of the literature suggests that the
self-stimulation reward threshold for cocaine. role of the cholinergic system is dependent on
The authors suggest that activating GABAB recep- the receptors involved within the drug response.
tors may represent a strategy for reducing the Accordingly, disruption in acetylcholine homeo-
rewarding effects of cocaine. However, a recent stasis largely affects the level of reinforcement,
ethanol self-administration study in baboons self-administration, and behaviors related to
showed that baclofen could reduce ethanol intake withdrawal and relapse. For example, genetic
but also consumption of vehicle, in this case the deletion of the muscarinic M5 receptor attenu-
orange-flavored drink Tang, suggesting that ates cocaine self- and operant administration in
the GABAB receptor agonist may just generally rats, and ethanol intake is significantly reduced
increase consummatory behavior (Duke et al., in mice after repeated administration of partial
2014). Tests of baclofen on alcohol-dependent nAch receptor agonists varenicline and cytisine
individuals are underway, and so far small clini- (Sotomayor-Zarate et al., 2013; Thomsen et al.,
cal trials have shown that the drug does not 2005). The authors report that the mechanism
produce any untoward side effects and can mod- behind these findings relates to muscarinic ace-
erately reduce alcohol intake (Leggio etal., 2010; tylcholine receptor-dependent increase in DA
Leggio et al., 2013). Currently a phase 2 clinical release in the VTA, a process that is lost with
trial is ongoing to evaluate the safety and efficacy deletion of the M5 receptor (Sotomayor-Zarate
of baclofen in abstinent individuals (clinicaltri- etal., 2013; Thomsen etal., 2005). Alternatively,
als.gov, NCT01604330). However, more tests muscarinic M4 receptor knockout mice have
are needed to determine the efficacy of GABAB increased cocaine operant self-administration
agonists for other drugs of abuse and stages of (Schmidt etal., 2011). In this case, Schmidt etal.
addiction. (2011) suggests that the deletion of M4 acetyl-
choline receptors, which colocalize with D1 DA
Cholinergic Homoeostasis receptors on GABAergic interneurons within the
In the CNS, acetylcholine is a principle inhibi- midbrain, may cause an indirect activation of DA
tory neurotransmitter with functions also as receptors.
a neuromodulator (Itier and Bertrand, 2001). With regard to drug behavioral effects, alter-
Acetylcholine is synthesized primarily in cholin- ations to acetylcholine homeostasis may affect
ergic neurons of the basal forebrain from choline drug responses that lead to addiction as well as
and acetyl-CoA. The ligand-gated nicotinic and withdrawal and relapse. Ethanol is of particular
G-protein coupled muscarinic (M1M5) acetyl- interest as individuals often co-abuse alcohol and
choline receptors are responsible for the action cigarettes or nicotine (Dawson etal., 2013). In a
of this neurotransmitter (Hurst et al., 2013). study where mice lacking the 2 subunit of nAch
Nicotinic acetylcholine (nAch) receptors are receptors underwent a battery of ethanol-related
broadly classified as muscle type and neuronal behavioral tests and self-administration, the
type and share structural homology with with authors found that mice lacking this particular
others in the cys-loop receptor family (i.e., GABA subunit had reduced time to recovery from etha-
and glycine receptors). Seventeen subtypes make nol hypnosis, enhanced anxiety, and no change in
pentameric ligand-gated ion channels, resulting ethanol intake (Dawson etal., 2013). In another
in much variation in how the receptors can come study, the rewarding effect of morphine (meas-
together and thus allowing very diverse actions. ured via the CPP paradigm) was reduced in mus-
While muscle nicotinic receptors always function carinic M5 knockout mice (Basile et al., 2002).
Brain Homeostasis and Addiction 517

In rats, pretreatment with the nAch receptor and amplification of cell surface Toll-like recep-
antagonist mecamylamine caused a significant tor proteins (Crews et al., 2011). Beyond this
reduction in the delayed discounting impulsivity stage, activated microglia secrete chemokines
test when paired with cocaine (Xie etal., 2012). leading to protease release (Crews et al., 2011).
The authors state that the data indicate that nAch These regulatory functions serve to return the
receptor populations, likely in central regions brain to a normal homeostatic environment.
such as the frontal cortex and NAc, are responsi- Of interest to the topic of addiction, these cells
ble for some impulsive decision-making and that have been shown to be sensitive to and activated
these processes in turn may influence relapse in by substance abuse. For example, a recent study
individuals addicted to cocaine (Xie etal., 2012). tested immune-suppressing drugs ibudilast and
Overall, these associations between drug behav- minocycline for their ability to reduce metham-
iors and the discrete molecular targets often phetamine self-administration in rats (Snider
identified by investigators from human genetic etal., 2013). The rationale for these experiments
studies, serve as a platform for further study and were based on previous work demonstrating that
provide promising targets for the development of drug use causes microglia-induced neuroinflam-
novel treatments for various stages of addiction. mation and that methamphetamine specifically
stimulates the production of proinflammatory
Inflammatory Immune Responses cytokines. Trained rats were systemically treated
Many investigators have taken several approaches with ibudilast and minocycline, resulting in
to study the role of immune inflammatory a significant reduction in methamphetamine
responses in addiction. These include looking consumption (Snider etal., 2013). Although the
at changes in immune genes of drug addicts, reduction of methamphetamine-induced glial
delivery of such genes to the brain of rodents activation was not measured in these animals,
to assess alterations in behavior using animal the findings suggest that modulating microglial
models of addiction, and investigating the effect activity may be a new avenue for treating drug
of pharmaceutical drug therapies on immunity addiction.
and inflammation (Crews et al., 2011). Recent
studies have shown that immune gene expres- Cytokines
sion is altered in the presence of drug stimuli. Cytokines are considered by some to be hor-
The transcription factor NF-kB (nuclear factor mones of the immune system that are responsible
kappa-light-chain-enhancer of activated B cells) for modulating responses to inflammation and
has been studied in this context due to its role injury (Dinarello, 2000; Kronfol and Remick,
in activating the production of immune signal- 2000). Cytokines do not readily cross the
ing molecules such as chemokines and cytokines blood-brain barrier (BBB); however, most of these
(Crews et al., 2011). Highly expressed in mono- small molecules can be produced and secreted in
cytes and microglia, NF-kB is necessary for the brain by astrocytes and microglia targeting
proper immune development, and dysregulation their respective receptors to modulate brain activ-
of this transcription factor can induce inflamma- ity (Kronfol and Remick, 2000). Classification of
tory responses (Crews and Vetreno, 2011; Garg cytokines is dependent on their biological activ-
and Aggarwal,2002). ity, and they are typically grouped into three
categories:proinflammatory, anti-inflammatory,
Microglia and hematopoietic (referring to the ability to
Microglial cell signaling is essential to the brains alter the responses of blood cells; Kronfol and
immune system. Microglia make up about 12% of Remick, 2000). The proinflammatory cytokines,
brain cells and have a very specific role in regulat- interleukins (IL)-1 and -6, and tumor necrosis
ing inflammatory immune responses to infection factor-alpha (TNF-), have been implicated in
and injury. Microglia recognize threats, travel to addiction with studies showing that several of the
the site of injury, and in some instances act as ILs can increase sensitivity to amphetamines and
macrophages to phagocytose foreign materials. modulate the DA system (Kronfol and Remick,
These scavenger cells are activated for several rea- 2000). Methamphetamine increases IL-1 and
sons, including altered extracellular potassium TNF- expression in the hypothalamus and NAc,
levels and in the presence of proinflammatory respectively (Yamada and Nabeshima, 2004). The
cytokines (Czirr and Wyss-Coray, 2012). In such authors propose that TNF- acts as a neuropro-
cases, their activated status calls for signaling tectant against drug addiction and neurotoxicity
518 Part IV:Homeostatic Therapies

by enhancing vesicular reuptake of DA (Yamada groundbreaking findings suggesting that antago-


and Nabeshima, 2004). A similar finding was nism or ablation of the histaminergic system can
reported by Tien et al. (2011), demonstrating promote the rewarding effects of drugs (Brabant
that neonatal exposure to lipopolysaccharide etal.,2010).
(LPS; an endotoxin responsible for the inflam- Administration of histaminergic drugs
matory effects of infection by Gram-negative causes changes in responses to amphetamines,
bacteria) causes increased behavioral sensitiza- opioids, and ethanol. The behavioral effects
tion and reinstatement for methamphetamine. after administration of various abused drugs are
Lipopolysaccharide exposure has been formerly thought to be differentially mediated by the three
linked to persistent increases in inflammatory histamine receptors. Dimenhydrinate, an H1
cytokines IL-1 and IL-6 and dysregulation of receptor antagonist was shown to be a rewarding
the DAergic system in the brains of rats (Tien drug for rats tested in the CPP paradigm (Halpert
etal., 2011; Tien etal., 2013). The findings from et al., 2003). Stimulation of H3 receptors has an
this study were interpreted to mean that inflam- opposite effect on drug behavior. For instance,
mation early in life may affect the susceptibility methamphetamine-induced increases in his-
to drug addiction later inlife. tamine are significantly decreased by pretreat-
ment with histamine H3 agonists. Consequently,
Histamine the reduction of hypothalamic histamine levels
Some endocrine functions, particularly the resulted in an increase in stereotypical behav-
histaminergic system of the hypothalamus, iors induced by the administration of metham-
have been demonstrated to be able to modulate phetamine (Kitanaka et al., 2011). Additionally,
immune inflammatory responses. Histamine in investigating the effects of methamphetamine
does not cross the BBB but instead is produced administration on histamine release in mice,
locally and may also be found in the gut via Morisset et al. (2002) observed an increase in
production in white blood cells (Brabant et al., histamine neuron activity in synaptosomes
2010; Smolinska etal., 2014). Typically the hypo- of mouse cerebral cortex and also increased
thalamus has endocrine and metabolic functions tele-methylhistamine (a histamine metabolite)
and, through the pituitary, links the nervous levels in several brain regions. Studies by these
system with several endocrine systems (e.g., laboratories and others attribute such effects to
hypothalamic-pituitary-adrenal axis [HPA] and the direct or indirect modulation of midbrain DA
hypothalamic-pituitary-gonadal). The normal levels by histaminergic drugs. The caveat to some
physiological function of histamine is to elicit of this work is the fact that histaminergic drugs
local immune responses and to act as a neuro- are not entirely specific to the three histamine
transmitter for the regulation of sleep patterns receptors and are capable of modulating other
(Smolinska etal., 2014). Because this molecule is neurotransmitter systems. Thus no clinical trials
largely considered a proinflammatory signaling are yet underway and future studies are needed
molecule, it has been studied in the context of var- to establish the neural mechanisms underlying
ious neurological disorders such as schizophre- alterations in drug reward by changes in hista-
nia and multiple sclerosis (Panula and Nuutinen, mine homeostasis.
2013). Behavioral neuroscientists have honed in
on histamine and its potential role in drug addic- Endocrine Function
tion due to influences on the DAergic system and Sex Steroids
brain reward (Brabant et al., 2010) and, more Although the majority of basic laboratory and
recently, the abuse of over-the-counter antihis- preclinical research utilize male subjects, those
tamine drugs such as Benadryl (Forest, 2008). that have studied addiction neural circuitry,
Notably, in several species histamine innerva- behavior, and underlying mechanisms in male
tion and the presence of histamine receptors and female animals often report sex differences.
(i.e., H1, H2, and H3) has been found to be high in Such differences are likely due, in part, to differ-
brain areas that are deemed important for drug ential patterns of brain organization and func-
addiction, including the NAc and VTA (Brabant tion between males and females. Considering
et al., 2010). Naturally these mechanisms could physiological differences that exist in circulating
translate to substance abuse and addiction. Early sex hormones, it is reasonable to expect sexu-
studies into the relevance of histamine recep- ally dimorphic responses to drugs in males and
tor presence in these brain regions have led to females. Even though sex differences cannot be
Brain Homeostasis and Addiction 519

exclusively explained by differences in circulat- return ethanol intake to levels of intact males.
ing levels of sex steroids, mounting evidence In a study of 10- to 12-year-old boys, regression
support the notion that sex hormones do indeed analysis of testosterone levels was able to pre-
influence many of the previously discussed dict illicit drug use and substance abuse prob-
homeostatic systems. Testosterone and estrogen lems during young adulthood (Reynolds et al.,
(i.e., estradiol) are well known for their func- 2007). Moreover, recent studies report reduced
tions in vertebrate development and reproduc- testosterone during withdrawal in opiate- and
tion (for a comprehensive review, see Viveros cocaine-dependent individuals (Wisniewski
et al., 2012). The mechanism of action for the etal., 2007; Yee etal., 2014). Undoubtedly, testos-
former involves direct activation of androgen terone activity in the brain contributes to drink-
receptors located throughout mammalian tissues ing behavior, some suggest as an influence over
and subsequent genomic influence via transloca- psychological factors; however, the mechanism(s)
tion to the nucleus, or a conversion to estradiol have yet to be elucidated.
and binding to estrogen receptors (Hiipakka In humans, the prevalence of addiction to
and Liao, 1998). Similarly, cytosolic estrogens alcohol and marijuana is high in males while,
bind to their respective receptors, which enter conversely, women have been found to have
the nucleus, bind to DNA, and alter gene expres- dependence problems with cocaine and psycho-
sion (McEwen, 2002). Of interest to this discus- stimulant drugs (Becker et al., 2012). Females
sion, and keeping with the theme of homeostatic also appear to be different than males in many
systems, this section focuses on the role that sex other drug-related measures, including earlier
steroids play in mediating activation of the brain age of first use as well as the rate of drug use esca-
in the presence of abused drugs, leading to the lation and quantity (Becker et al., 2012). Many
progression of addiction. Surprisingly, in this researchers have focused on the involvement of
regard, fewer studies have investigated the direct estrogen and its modulation during the female
influence of testosterone on addictive behaviors menstrual cycle. Estrogen receptors alpha (ER)
than estrogen (Carroll etal., 2004). Both testos- and beta (ER) reside in the cytoplasm of cells
terone and estrogen are derived from cholesterol throughout many tissues; however, notably, the
and produced in the testes of males and ovaries ER subtype is localized in the brain (Matthews
of females, respectively; however, testosterone and Gustafsson, 2003). Ovarian hormones appear
function is not exclusive to males nor is estro- to primarily be involved with drug acquisition
gen to females (Neil and Kulkarni, 2011). Despite and sensitization, though mounting evidence
being synthesized in areas outside of the brain points toward a role for such hormones in the
and their importance for reproductive functions, reinforcing effects of drugs. Studies of estradiol
these steroid hormones are permissible across the in rodents have shown that the administration of
BBB, and testosterone and estrogen receptors are estradiol to gonadectomized females facilitates
dispersed throughout thebrain. the acquisition of cocaine self-administration
Testosterone fluctuations in males are largely (Hu et al., 2004). This is supported by previ-
related to social interactions and reproductive ous evidence that cocaine use varies over the
status (Becker etal., 2012). As with hormone var- course of the estrous cycle (Hu et al., 2004).
iations during estrous in females, the pattern of Investigations of DA levels in the dialysate of ova-
testosterone release in males may influence drug riectomized females, castrated males, and intact
behaviors. Due to the difficulty and potential females and males revealed that after treatment
stressful confound of monitoring estrous status with estradiol, cocaine-induced DA release was
in addiction animal models, most studies utilize greater in the dorsolateral striatum (and not the
surgical alternatives such as gonadectomies and NAc) in females compared to males. In addition,
hormone replacement to study the effect of ster- locomotor sensitization after systemic cocaine
oid hormones on drug behaviors. Vetter-OHagen administration was higher in females, indicat-
and colleagues (2011) found that ethanol prefer- ing sex differences in behavioral sensitization
ence in gonadectomized male rats was higher (Cummings et al., 2014). A recent review high-
compared to gonadectomized males receiving lights the developing role of ovarian hormones
testosterone hormone replacement. In addi- in mediating drug reward within the mesolimbic
tion, although the amount of ethanol consumed area and HPA-axis, suggesting that processes that
did not differ between the groups, testosterone are altered in dueling brain systems contribute to
replacement in castrated males was sufficient to a net change in brain reward, which eventually
520 Part IV:Homeostatic Therapies

leads to attenuation in positive drug reinforce- cue-induced reinstatement in rats (Schmoutz


ment (e.g., tolerance; Bobzean etal., 2014). Once et al., 2014). Conversely, chronic morphine
this occurs, the drug user then consumes increas- administration reduces the level of neurosteroids
ing amounts of drug to re-experience the initial (e.g., progesterone and pregnenolone sulfate) in
rewarding sensation (Bobzean etal., 2014). Taken male rats (Yan and Hou, 2004). The authors report
together, the authors results suggest that estra- that the reduction of neurosteroid concentration
diol possibly mediates long-term neural changes was related to a morphine-induced inhibition of
related to the progression from voluntary to com- the HPA axis resulting in lower levels of precur-
pulsive use (Cummings etal.,2014). sors such as testosterone and corticosterone (Yan
and Hou, 2004). Using repeated administration
Neurosteroids of morphine and naloxone-induced withdrawal
Unlike sex steroids, neurosteroids are hormones to test mice for the development and expression
produced de novo, directly in the brain (Helms of morphine dependence and tolerance, Ren and
et al., 2012). Neurosteroids are derived from colleagues (2004) found that DHEAS prevented
local cholesterol or the metabolism of steroid tolerance to morphine-induced analgesia and a
precursors that are synthesized in the adrenals significant reduction in morphine dependence
and gonads (Mellon and Griffin, 2002) and are measured via withdrawal symptoms.
capable of modulating both excitatory and inhib- Surprisingly, a recent extensive study of
itory (i.e., NMDA receptor and GABA recep- drugs from all five classifications reported that
tor, respectively) activity and neurotransmitter pregnenolone reduced the firing of VTA DA
release (Helms etal., 2012). While their normal neurons, increased the level of DA in the NAc,
function is to provide rapid homeostatic modu- and blunted behavioral disturbances only to
lation of neurotransmission, pregnenolone sul- THC or Cannibis sativa (Vallee etal., 2014). It is
fate, dehydroepiandrosterone sulfate (DHEAS), clear from the literature that the effects of neu-
and allopregnanolone (among others) are a few rosteroid on addiction is complex and subject to
naturally occurring neurosteroids that have confounding factors, including species differ-
been recognized as regulatory molecules in drug ences, endogenous levels of circulating steroids,
addiction. The pharmacological action of preg- and experimental conditions (e.g., time of day;
nenolone in the brain involves enhancement of Helms etal., 2012). However, neurosteroids still
GABA A receptor activity (Besheer et al., 2010). warrant continued investigation as underlying
Allopregnanolone is also a GABAergic neuro- mechanisms may provide additional insight into
steroid with hypnotic and anxiolytic effects in the the diverse neuropathology of different abused
brain (Reddy, 2010). DHEAS, the sulfated metab- drugs and the advantage of allosteric modulation
olite produced from the conversion of DHEA to of neurotransmitter systems.
testosterone, antagonizes GABA A receptor activ-
ity and has been implicated in age-related bone Energy Balance and Metabolic
and skin pathologies as well as neuropsychiatric Disturbance
and mood disorders (Mo etal.,2006). Energy balance is one of the basic processes
Collectively, investigations on neurosteroids necessary to survival. The body is intricately
and abuse point toward the idea that neuroster- designed to maintain energy homeostasis, and
oids may be a viable avenue for preventing addic- disturbances in metabolism can have highly dele-
tion to some substances. Studies have shown terious effects throughout the peripheral nervous
that pregnenolone can reduce operant ethanol system and CNS. The main signaling molecules
intake in alcohol-preferring rats, a genetic model responsible for proper energy balance are peptide
of excessive alcohol intake (Besheer etal., 2010). hormones leptin, ghrelin, and insulin. Leptin and
The mechanism for this is still under investi- ghrelin are two appetite-regulating hormones.
gation, but the authors suggest that the key to Leptin is produced primarily in white adipose
pregnenolone-induced reduction of ethanol tissue and has also been found in the placenta,
intake is related to the alcohol-dependent pheno- stomach, and the pituitary (Ahima and Osei,
type and potentially the idea that this neuroster- 2004). The traditional role of leptin is to activate
oid can improve cognitive deficitsa common receptors in the hypothalamus, thereby promot-
comorbidity linked to alcohol addiction (Besheer ing appetite suppression by counteracting the
et al., 2010). With regard to relapse to cocaine, feeding stimulants neuropeptide Y (Ahima and
allopregnanolone has been shown to reduce Osei, 2004). Ghrelin acts in opposition to leptin
Brain Homeostasis and Addiction 521

in that it stimulates hunger (Klok et al., 2007). the analysis indicated that leptin increases dur-
Human P/D1 (or rodent X/A-like) cells in the ing abstinence is potentially related to a reduc-
stomach are central to the production and release tion in DAergic activity in the reward system
of ghrelin, which increases just before hunger and (Aguiar-Nemer et al., 2013). Finally, a study of
diminishes after eating (Kirchner et al., 2012). the effects of ghrelin on alcohol craving and use
The main function of insulin, coming from beta in heavy drinkers is being evaluated in a clini-
cells of the pancreas, is for carbohydrate and fat cal trial that is currently recruiting participants
metabolism. This is achieved by removing excess and is slated to complete by the fall of 2020.
glucose from the blood stream. Participants will be tested for alcohol craving
Feeding and other activities important for and use after receiving an infusion of ghrelin or
survival (e.g., mating) are considered to be nat- placebo, and investigators will use fMRI to assess
ural rewards. As such, when performing any function of brain regions associated with incen-
of these activities, the reward brain circuitry is tive salience and reward (clinicaltrials.gov, NCT
activated as a means to motivate toward these NCT01779024).
behaviors. This is the same reward circuitry that
is hijacked by many drugs of abuse, and similar Neurotrophic Factors
DA-mediated mechanisms are at play. Notably, a Neurotrophic factors are a family of signaling
deficiency in energy status increases the salience molecules with many integral roles in the brain,
and hedonic value of food and drinks. Both lep- including neuronal growth and maturation,
tin and insulin are known to reduce food- and maintenance of mature neurons, and synap-
drug-seeking in animals (Cummings etal., 2007). tic plasticity (Autry and Monteggia, 2012; Poon
Similarly, these energy homeostatic neuropep- et al., 2013). Within the family are three clas-
tides have influence over the mesocorticolimbic sifications (a) neurotrophins including neuro-
DA reward system (Fulton et al., 2000; Jerlhag trophin -3 and -4, nerve growth factor (NGF),
et al., 2007). Preclinical studies have explored and brain-derived neurotrophic factor (BDNF);
the potential for leptin to modulate drug-related (b) glial cell line-derived neurotrophic factor
behavior. Interestingly, leptin-deficient ob/ob family ligands (e.g., glial cell line-derived neu-
mice have been shown to have reductions in rotrophic factor [GDNF]); and (c) neuropoietic
their locomotor responses and sensitization to cytokines (Deister and Schmidt, 2006). Since the
amphetamine. These findings were substantiated discovery of these proteins more than several
in other models, including a reversal in behav- decades ago, they continue to be intensely stud-
iors after a leptin infusion as well as reductions ied in order to fully understand the diversity of
in DA released from the NAc in slice prepara- their biological roles (Bolanos and Nestler, 2004).
tions (Fulton et al., 2006). With regard to drug While a few studies have looked at the contri-
reinstatement, Shalev etal. (2001) demonstrated bution of NGF and neurotrophin-3 in addictive
that intracerebral ventricular infusion of leptin behavior, BDNF and GDNF have received the
in rats can attenuate heroin-seeking induced by most attention to date with regard to synaptic
food deprivation. The authors theorize that lep- plasticity and various neuropsychiatric disorders
tin is involved in modulating motivational states (Autry and Monteggia, 2012). Thus this section
linked to food restriction or deprivation (Shalev focuses specifically on the links between drug
etal., 2001). In contrast, ghrelin, when adminis- addiction and brain homeostasis involving these
tered to the VTA and ventricles of mice, causes two neurotrophic factors.
a significant increase in ethanol consumption.
Additionally, accumbal DA release and alcohol BDNF
CPP was abolished in ghrelin receptor knock- Within the past couple of decades, a potential
out mice as well as wild-type mice treated with link between neurotrophic factors and addic-
receptor antagonists (Jerlhag etal.,2009). tion has been made via chronic drug studies
Fewer studies can be found in the literature in animals while investigating the underlying
relating energy homeostasis and drug addiction mechanisms for observed plasticity in mesocor-
in humans. A meta-analysis by Aguiar-Nemer ticolimbic regions (Bolanos and Nestler, 2004).
and colleagues (2013) of publications on the topic Mounting evidence supports the notion that
revealed that leptin levels may be considered a BDNF is an important modulator of drug
biomarker for the risk of relapse in alcoholics reward. Protein and mRNA of this neurotrophic
and smokers. The 12 studies identified during factor is expressed in mesocorticolimbic areas,
522 Part IV:Homeostatic Therapies

and several groups have reported that cocaine, responses following chronic drug administration
ethanol, and amphetamine alter the levels of (Messer etal., 2000). Similarly, GDNF attenuates
BDNF in several brain regions key to reward pro- ethanol self-administration and relapse when
cesses, including the striatum, frontal cortex, and infused into the VTA but not the substantia nigra
the amygdala (Meredith etal., 2002; Raivio etal., of rats (Carnicella etal., 2008)and significantly
2012). Alcohol abuse appears to be more complex reduces methamphetamine self-administration
regarding BDNF. Logrip etal. (2009) found that and relapse when delivered to the striatum in
acute ethanol self-administration increases cor- mice (Yan etal., 2013b).
tical BDNF expression in male C57Bl6/J mice, Since neurotrophic factor signaling path-
whereas long-term exposure (6 weeks) to ethanol ways are so vast and critical to the mainte-
reduces BDNF in the cortex. The authors con- nance and survival of neurons, one idea for the
sider such findings to be representative of how way they facilitate various stages of addiction,
BDNF is involved in the escalation of drug use mainly reward and relapse, is based on findings
to addiction. In this study BDNF appeared to be that these proteins can alter neuronal plasticity.
protective early on but did not recover 2 weeks Although the mechanisms are still under inves-
after ethanol deprivation, indicating that this tigation, several groups report that increased
neurotrophic factor may be involved with the BDNF expression enhances dendritic formation
inflexibility of drug addiction (Grimm et al., (Bolanos and Nestler, 2004). As a consequence
2003; Logrip etal.,2009). of increased synaptic connections at dendrites,
To directly assess the role of BDNF signaling synaptic strength and neuronal signaling is also
on addictive behaviors, some groups have investi- altered (Bolanos and Nestler, 2004). Furthermore,
gated drug-seeking and relapse after BDNF infu- neurotrophic factors activate several complex
sion into reward areas. While a study by Berglind signaling pathways involving phospholipase C,
etal. (2007) reports that infusing BDNF into the phosphatidylinositol-3-kinase, and extracellu-
medial PFC suppresses cocaine-seeking, another lar signal-regulated protein kinase. In doing so,
group has shown that cocaine-seeking behavior and depending on which pathway is activated, a
is enhanced for up to 30 days after withdrawal rise in intracellular calcium or cellular structural
in rats receiving intra-VTA infusions of BDNF changes is initiated and thus long-term poten-
(Lu etal., 2004). Notably, there was no effect of tiation is increased, causing an elevation in drug
NGF infusion on cocaine-seeking or BDNF infu- sensitivity and reward (Bolanos and Nestler,
sion when delivered to the substantia nigra (Lu 2004). Surely these mechanisms come into play
et al., 2004). Although differential roles of vari- during chronic drug exposure, and so it is plausi-
ous brain regions are evident, these data support ble to attribute some aspects of addiction to plas-
the notion of BDNF-mediated development of ticity changes mediated by neurotrophic factors.
drug addiction.
A LT E R N AT I V E A D D I C T I O N
GDNF THERAPIES
GDNF is expressed throughout the CNS and at its Most currently approved treatments for drug
highest levels in the striatum, thalamus, cortex, addiction utilize pharmacotherapies that mod-
and hippocampus. GDNF activity in the adult ulate the various neurotransmitter systems or
brain is primarily located in the midbrain where act as replacement substances. Interestingly, in
its expression is kept low under normal condi- the United States the only treatments approved
tions and then supplied to regions of injury via by the Federal Drug Administration are use-
retrograde transport by the striatum (Carnicella ful for alcohol, nicotine, and opioid addiction.
and Ron, 2009). Interestingly, studies show that Those suffering with alcohol use disorders have
this neurotrophic factor has a mostly universal the most options. Aside from the moderate suc-
effect on drug self-administration, reward, and cess of self-help programs such as Alcoholics
relapse in that activating GDNF signaling attenu- Anonymous, which describes necessary actions
ates drug (i.e., psychostimulant, morphine, and and spirituality leading toward recovery (Detar,
ethanol) behaviors. This has been demonstrated 2011), pharmacological interventions are availa-
by intra-VTA infusions of GDNF in rats as well as ble, including the alcohol dehydrogenase blocker
infusion of anti-GDNF antibodies in mice, result- disulfiram (Antabuse), the opioid antagonizing
ing in significant reductions in cocaine reward drug naltrexone (Vivitrol ), and NMDA recep-
or place preference and locomotor sensitization tor modulator acamprosate (Campral ). Of late,
Brain Homeostasis and Addiction 523

clinical trials have tested the anti-alcohol efficacy be centered on impulsivity, which is a predic-
of nalmefene, an opioid antagonist with a longer tor of relapse in drug addiction. Schmaal et al.
half-life, better bioavailability, and reduced tox- (2012) was able to show that impulsivity scores in
icity compared to naltrexone (Hoofnagle, 2012). cocaine-addicted patients were predictive of the
Two large studies show that pharmacological amount of NAC-induced changes in glutamate.
treatment with nalmefene effectively reduces Although this study had limitations related to
alcohol intake in dependent individuals who relatively moderate sample sizes and confound-
are unable to reduce consumption on their own ing comorbid addictions in the patient popu-
(Gual et al., 2013; van den Brink et al., 2013). lation (e.g., alcohol and nicotine), others have
Opioid addiction is often treated with the syn- provided evidence that NAC may work therapeu-
thetic opioids methadone or buprenorphine. tically in patients with dependence on marijuana,
Similarly, nicotine-dependent individuals have pathological gambling, and nicotine (Schmaal
the option of pharmacological treatment with etal.,2012).
nicotine replacements or varenicline, which is Promising new therapies have come to frui-
a partial nicotinic acetylcholine receptor ago- tion for those with cannabis (or marijuana)
nist (Tomek et al., 2013). Although these treat- substance abuse and dependence problems.
ments are available, they have limited efficacy, These largely exploit the notion that those with
and addiction remains a pervasive problem, par- cannabis addiction are also more likely to have
tially due to patient noncompliance and also due concomitant mental health problems. Such
to a poor understanding and the complexity of pharmacotherapies as lithium and divalproex,
underlying mechanisms of action of these drugs. two mood-stabilizing drugs, have been shown
Accordingly, scientists are seeking alternative to improve cannabis withdrawal symptoms
strategies for blocking the progression to addic- (Danovitch and Gorelick, 2012). Nonetheless, the
tion and preventing relapse. continued development of alternative treatments
Researchers are beginning to investigate the for addiction is slow, costly, and largely limited
ability of biologic compounds to act as thera- by the complexity of drug interactions with dif-
pies for addiction to certain drugs. Several com- ferent parts of the brain. Accordingly, research-
pelling recent preclinical studies suggest that ers are turning to natural interventions that are
N-acetylcysteine (NAC) may be a novel thera- capable of modulating several brain homeostatic
peutic for cocaine addiction. NAC is a cysteine mechanisms.
prodrug that purportedly acts at mGluR2/3
receptors to facilitate the exchange of extracel- Exercise
lular cysteine for intracellular glutamate, thereby Some say that there are similarities between the
normalizing glutamate levels (Moran et al., act of using drugs and the act of exercise. This
2005). This process is important for cocaine commonality is thought to exist based on the
addicts since chronic use of this psychostimulant fact that both produce positive affective states.
drug results in reduced levels of glutamate out- Preclinical studies support the idea that exercise
side of the cells within the NAc and PFC regions may have many benefits for substance-dependent
(Moran et al., 2005). Taking this work further, people. Particularly with regard to cocaine and
Murray and colleagues (2012) tested the utility methamphetamine, exercise has been shown to
of NAC as an antidrug therapy in rats using an reduce self-administration at several stages of
operant self-administration paradigm, finding addition throughout the progression from ini-
that animals reduced cocaine-seeking in both tial drug use to abuse and dependence (Smith
early- and late-stage addiction without affecting and Lynch, 2011). Several drugs have been stud-
the amount of cocaine consumed. The authors ied for the effect of exercise on reducing intake
concluded that this work supports the potential and progression to addiction, with much of the
for NAC to act as a therapy to prevent relapse, work focused on drug withdrawal and reinstate-
but not reinforcement, for cocaine via regulation ment after abstinence. Devaud and colleagues
of glutamate homeostasis. Since then, NAC has (2012) report that rats with access to running
also been tested clinically in cocaine-dependent wheels that voluntarily exercised (measured
patients and has resulted in similar effects as as wheel turns) had a significant increase in
seen in the preclinical studies (Schmaal et al., pentylenetetrazol-induced seizure threshold after
2012). In humans, the link between NAC and removal from an ethanol liquid diet. In an oper-
regulation of glutamate homeostasis seems to ant responding paradigm, rats that had access to
524 Part IV:Homeostatic Therapies

a running wheel displayed a reduction in rein- normalized after exercise, a mechanism that may
statement after extinction and abstinence from similarly be related to exercise-induced attenua-
cocaine self-administration compared to seden- tion of drug-seeking behavior and relapse (Smith
tary rats (Smith etal., 2012). In recent years the and Lynch, 2011). Neurotrophins have also been
Solinas group has demonstrated in a number of implicated, as several studies support the idea
studies that environmental enrichment (EE; i.e., that exercise promotes neurogenesis in the hip-
access to ramps, running wheels, and toys) causes pocampus, a process that may be involved in the
a significant reduction in craving and the pro- prevention of drug- and cue-induced cravings
pensity for relapse. Cocaine-seeking in rats was and relapse (Smith and Lynch,2011).
reduced after 1-week exposure to EE compared Although the effect of exercise-mediated
to standard environment, and this effect was alterations in brain homeostasis on addictive
lost upon discontinuation of the enriched hous- behavior seems to be an interesting avenue, sev-
ing conditions (Chauvet etal., 2012). In addition, eral questions remain unanswered. Studies inves-
studies show that exercise attenuates the positive tigating how much exercise and when exercise
reinforcing effect in rodents when challenged needs to be performed in relation to drug use and
with other drugs of abuse such as heroin (Smith the progression to addiction is one. Furthermore,
and Pitts, 2012). The ability for exercise to slow of the different types of exercise, which type is
the escalation from drug use to abuse has been the most therapeutic? Another area for more
demonstrated to be more effective in adolescent research is whether sex hormones, which defi-
rodents than adult rodents (Zlebnik etal., 2012). nitely influence addictive behavior, have differ-
Moreover, humans undergoing treatment for ential effects of exercise in males and females.
substance-use disorders have been more success- And last, gaining a better understanding of the
ful with the incorporation of exercise (Weinstock underlying biological mechanisms involved and
etal.,2008). how this relates to alterations of brain homeosta-
Behavioral and psychological mechanisms sis after drug use will help inform clinicians on
involved in the ability of exercise to reduce drug the best strategies for exercise as an alternative or
intake are likely related to (a) reductions of the conjunctive therapy for addiction.
drug as a positive reinforcer when both the
drug and exercise are available at the same time; Mindfulness Training and Meditation
(b) a reduction in potential comorbid disorders Mindfulness training is a technique that theo-
(e.g., depression or anxiety), which are risk fac- retically involves shifting ones attention to the
tors of substance abuse and reduced by exercise immediate experience in a nonjudgmental and
in humans; and (c) a general improvement in accepting kind of way. This skill is at the core of
affective state since in most conditions exercise meditation, is often learned through repetition
increases feelings of well-being and self-esteem or practice, and has been shown to be particu-
in humans (Smith and Lynch, 2011). Nader etal. larly beneficial in individuals with concomi-
(2012) substantiated this theory in a study of the tant mental health and substance use disorders
influence of EE versus standard environment (Brewer etal., 2010). Meditation is rooted in reli-
on cocaine addiction in mice. In this study, the gious practices (i.e., Hindu-based transcendental
authors found that in young mice, removal of EE meditation or Buddhist meditation); however,
after exposure prevented the protective effect of although mindfulness relies on Buddhist ideas,
the enriched conditions and thus these animals when used therapeutically the spiritual associa-
reward for cocaine was higher and they were more tion is not made (Dakwar and Levin, 2009). Two
vulnerable to addiction. This effect was attrib- major aspects of mindfulness training involve
uted to emotional distress related to the loss of the development of improved attention and
EE measured via levels of corticotropin-releasing acceptance skill sets in those with addiction. This
factor (Nader etal., 2012). Related to the neuro- allows the individual to pay attention to present
biology behind exercise-induced reductions in internal and external stimuli and become accept-
drug-taking, many lines of evidence suggest the ing of these observations in a nonjudgmental
involvement of neurotransmitters also relevant way. Mindfulness-training techniques have been
to drug addiction. For example, studies show that applied as ways to manage stress, change behav-
exercise is capable of increasing DA in the rodent ior, aid cognition, and prevent relapse. Relapse
basal ganglia (Meeusen etal., 1997). In addition, prevention therapy is not mindfulness treatment
increased glutamate levels caused by ischemia are per se but rather uses mindfulness as a strategy to
Brain Homeostasis and Addiction 525

help manage cravings. For example, addicts are empirically tested using functional and struc-
trained to expect that cravings will occur but will tural neuroimaging. Findings from several stud-
eventually pass and to accept and learn to deal ies show that neuroplastic changes occur in the
with them (Dakwar and Levin,2009). fronto-limbic areas, and others after mindfulness
A growing number of clinical studies have mediation (for a review, see Hlzel et al., 2011),
investigated the effectiveness of mindful- and that these are likely to be beneficial for recov-
ness training for substance misuse and abuse. ery from addiction.
Currently, the literature provides evidence that a A major limitation is that much of the work
mindfulness approach, sometimes coupled with in the field of mindfulness training and addic-
more contemporary treatments, can be useful for tion utilizes small sample sizes. This raises the
preventing anxiety, depression, stress, cravings, question as to whether studies with nonsignifi-
and impulsive thinking (Bowen and Enkema, cant effects actually lack statistical power. In
2014). Such studies utilize various mindfulness addition, as mentioned earlier, investigators may
techniques and individuals with differing racial employ different styles of mindfulness training
backgrounds and addictions to a variety of drugs and often cast a wide net for study participants.
(i.e., alcohol, cocaine, methamphetamine, and Although more controlled studies are needed on
marijuana), and all report positive relationships the different types of mindfulness techniques
between psychological states after this therapy and their utility for treating substance abuse,
and craving and drug use during the subsequent mindfulness-based meditation has great poten-
months (de Dios et al., 2012; Witkiewitz and tial for restoring brain homeostasis in addicts,
Bowen, 2010). Furthermore, when polysubstance especially in light of success with various appli-
abusers were tested in measures of working mem- cations such as stress reduction and behavior
ory, behavioral inhibition, and decision-making, modification.
those who received mindfulness training per-
formed better than those who did not (Alfonso Acupuncture
etal., 2011). The authors believe that these tech- Acupuncture, a form of traditional Chinese med-
niques warrant continued examination to deter- icine, is a technique involving stimulation of var-
mine whether mindfulness training represents a ious points along the skin using needles, heat, or
novel therapy for reducing neuropsychological pressure (Ernst, 2006). This practice is thought to
deficits and relapse in drug addicts. restore the imbalance of flow (or qi) along merid-
The proposed theory for the efficacy of mind- ians in the body (Ernst, 2006). The prevalence
fulness training and meditation is related to a of the use of acupuncture to treat drug-related
potential role in attenuating cravings, facilitating disorders in the United States was climbing until
cue extinction, and reducing compulsive behav- 2002 when two large-scale clinical trials reported
iors (Dakwar and Levin, 2009). Neurobiological negative results (White, 2013). These two studies
information on this hypothesis is difficult to showed no difference between the acupuncture
attain since all studies are done on humans. and sham treatment in maintaining abstinence
Clinicians have been strategic in their experi- in individuals dependent on alcohol and cocaine
mental design to investigate mechanisms behind (White, 2013). Despite the lack of research evi-
the efficacy of mindfulness training. Subject dence to support the ability of acupuncture to
plasma analysis and also brain imaging can be reduce drug dependence, it is still used in clinical
applied pre-, post-, and/or during the interven- practice for this problem. White (2013) believes
tion. The release of the stress-induced hormone that this is due to the large variability in the many
cortisol is a common pathological trait in addicts parameters involved with acupuncture, including
that has been linked to craving and drug use bilateral versus unilateral application of needles
(Brewer etal., 2010). Thus it is logical to believe and the use of electrical stimulation. In addition,
that attention and acceptance of the present with acupuncture methods appear to be successful
a nonjudgmental attitude instead of rumination on discrete components of the drug-addiction
would elicit alterations in circulating cortisol in stages (i.e., craving and withdrawal; White,
those with substance abuse problems; however, 2013). Arecent study that examined tobacco ces-
this has not been explicitly studied. In addition, sation within the Chinese population at a treat-
neural mechanisms to support reported changes ment center found that individuals who elected
in memory, decision-making, and impulsivity to receive acupuncture in addition to nicotine
resultant from mindfulness training have been replacement therapy had higher cessation rates
526 Part IV:Homeostatic Therapies

after 6 months (Chang et al., 2013). Although doses of PF11 had a reduction in the level of
there were several limitations to the study, the morphine-induced reduction of extracellular
results suggest that acupuncture could be a bene- glutamate in the medial PFC (Hao etal., 2007).
ficial and attractive supplementary treatment for Taken together, PF11 may represent a novel ther-
addictions to a wide range of substances. Further apy for addiction, particularly to morphine and
evaluation is needed, but acupuncture appears to methamphetamine relapse.
be a safe, potential alternative therapy for drug Daidzin and daidzein, two extracts from the
addiction. root of the East Asian Puerariae plant (or Kudzu),
have been used to treat hypertension and reduce
Herbal Remedies fever and nausea (Lu et al., 2009). These herbal
In China, herbal remedies have long been a extracts reportedly reduce metabolism of liver
way to treat ailments. However, only within the monoamines and serotonin. Additionally, with
past couple of decades has this approach come regard to alcohol, daidzin is capable of inhib-
into acceptance elsewhere. Clinical trials have iting mitochondrial aldehyde dehydrogenase
recently brought to light the notion that herbal in the liver (Overstreet et al., 2003). Kudzu has
medicine can be an alternative strategy to tra- been studied for the ability to reduce free-choice,
ditional pharmacological treatments for addic- continuous-access ethanol intake in rodents.
tion. In addition, discoveries providing possible Although at this time few preclinical studies
mechanisms of action for various herbal com- exist, this natural medicine has been tested in
pounds support their utility and aid in the design human clinical trials where alcohol consump-
of new therapies. To date, several traditional tion in alcohol-dependent individuals was exam-
herbs have been tested for their ability to reduce ined following treatment with Kudzu root. The
behaviors associated with addiction, and some results have been mixed. In a recent trial, Lukas
have even gained popularity in Western culture. etal. (2005) reported that subjects who received
Plant extracts such as ginseng, Kudzu, kava, and and took the herbal remedy for a period of 7 con-
St. Johns Wort are pharmacologically active and secutive days drank significantly fewer alcoholic
have been described as beneficial for treating a beverages during laboratory testing compared
variety of ailments. to the number of beverages consumed prior to
One of the most popular and most stud- treatment. The authors provide evidence that
ied herbs is ginseng (Panax quinquefolium in Kudzu may work by enhancing the effects of the
America). The major compound responsible for first beverage, since pretest craving scores were
most action(s) of ginseng in the CNS is ginse- not significantly altered by Kudzu treatment.
nosides. More than 20 ginsensoides have been Because those treated with Kudzu drank smaller
identified and are thought to have an inhibitory volumes and took more sips of alcohol for subse-
and excitatory influence on neurotransmission, quent drinks, also reporting feeling more intoxi-
effecting learning and memory and nociception cated after the first drink, Kudzu appears to be a
(Attele et al., 1999). Because of the diversity of way to curb binge-drinking (Lukas etal., 2005).
ginsengs structural constituents and effects on Compared to other trials, the one by Lukas and
the brain, it has gained attention for its potential colleagues differs in that a natural setting (i.e.,
as an anti-addiction remedy. This herb report- the lab testing area resembled a small studio con-
edly reduces methamphetamine and morphine taining an entertainment center, kitchen, and
behaviors in rodents, an effect resultant from couches) was utilized, a more concentrated dose
the antagonist action of psuedoginsenoside-F11 of Kudzu was tested similar to preclinical testing,
(PF11) on opioid signaling and DA release (Wu and the amount of active ingredient and com-
et al., 2003). Ginseng is known to have both pliance was verified by urine analysis. Differing
excitatory and inhibitory actions in the CNS, but from the aforementioned study, a recently listed
in a recent study by Hao and colleagues (2007), clinical trial will test the efficacy of Kudzu in
mice that were pretreated with PF11 followed by a heavy drinkers predetermined to be alcohol
systemic injection of morphine showed no devel- dependent (clinicaltrials.gov, NCT01853293).
opment of behavioral sensitization in locomotor The interaction of Kudzu with the metabolism
responses, thereby suggesting an inhibition of of ethanol has been well documented. Acting
morphine-induced DAergic neurotransmission. as an inhibitor of acetaldehyde dehydrogenase,
Furthermore, in vivo microdialysis revealed that much like disulfiram, this natural medicine has
animals pretreated chronically or with higher the potential to be a strong anti-alcohol therapy.
Brain Homeostasis and Addiction 527

Additionally, Kudzu binds to benzodiazepine Aguiar-Nemer, A.S., Toffolo, M.C., da Silva, C.J.,
receptors on GABA receptors in brain regions Laranjeira, R., and Silva-Fonseca, V.A. (2013).
that are neural substrates of alcohol (e.g., cortex, Leptin influence in craving and relapse of alco-
cerebellum, and hippocampus; Lu etal.,2009). holics and smokers. Journal of Clinical Medicine
Lesser-studied plant extracts such as kava and Research 5, 164167.
St. Johns Wort have been clinically used for the Ahima, R.S., and Osei, S.Y. (2004). Leptin signaling.
treatment of psychological disorders such as anx- Physiology & Behavior 81, 223241.
iety and depression, respectively (Lu etal., 2009; Alfonso, J.P., Caracuel, A., Delgado-Pastor, L.C., and
Overstreet et al., 2003). Notably, lipid-soluble Verdejo-Garcia, A. (2011). Combined goal man-
kava lactones and St. Johns Wort modulate brain agement training and mindfulness meditation
neurotransmitter concentrations (e.g., DA, sero- improve executive functions and decision-making
tonin, and GABA), allowing for a role in promot- performance in abstinent polysubstance abusers.
ing abstinence and reducing cravings in addicts Drug and Alcohol Dependence 117,7881.
(Lu etal., 2009; Overstreet etal., 2003). Thorough American Psychiatric Association. (2000). Diagnostic
and Statistical Manual of Mental Disorders (4th
preclinical testing and human clinical trials are
ed., text rev.). Washington, DC:Author.
still needed to better understand efficacy, mech-
Attele, A.S., Wu, J.A., and Yuan, C.S. (1999). Ginseng
anisms, and the interaction of various drugs of
pharmacology: Multiple constituents and mul-
abuse with these herbal medicines. In addition,
tiple actions. Biochemical Pharmacology 58,
herbs such as Kudzu and kava are heavily regu-
16851693.
lated as they can be very toxic, particularly to the
Autry, A.E., and Monteggia, L.M. (2012).
liver. Some of these herbs have been utilized for Brain-derived neurotrophic factor and neuro-
centuries in other parts of the world; however, psychiatric disorders. Pharmacological Reviews
their safety and toxicity for the masses have not 64, 238258.
been thoroughly investigated and documented. Basile, A.S., Fedorova, I., Zapata, A., Liu, X.,
Shippenberg, T., Duttaroy, A., Yamada, M., and
CONCLUSIONS
Wess, J. (2002). Deletion of the M5 muscarinic
In summary, brain homeostasis is critical for acetylcholine receptor attenuates morphine
proper physiological function and also appears reinforcement and withdrawal but not morphine
to be very much involved in the progression of analgesia. Proceedings of the National Academy
drug use to addiction. Drug interactions with of Sciences of the United States of America 99,
many of the homeostatic systems eventually 1145211457.
lead to the modulation of DA-related neuronal Baydas, G., and Tuzcu, M. (2005). Protective effects
activity in the mesocorticolimbic regions as well of melatonin against ethanol-induced reactive
as influence excitatory and inhibitory neural gliosis in hippocampus and cortex of young and
transmission. Despite the need for much more aged rats. Experimental Neurology 194, 175181.
testing, the likelihood that brain homeosta- Becker, J.B., Perry, A.N., and Westenbroek, C. (2012).
sis manipulation of various stages of addiction Sex differences in the neural mechanisms medi-
seems plausible. Current investigations along the ating addiction:Anew synthesis and hypothesis.
lines of alternative treatment options (e.g., diet, Biology of Sex Differences3,14.
exercise, meditation, acupuncture, and herbal Bengel, D., Murphy, D.L., Andrews, A.M., Wichems,
medicines) for addiction are promising as ways C.H., Feltner, D., Heils, A., Mossner, R., Westphal,
to overcome some of the challenges related to the H., and Lesch, K.P. (1998). Altered brain sero-
time and funds required to develop new phar- tonin homeostasis and locomotor insensitiv-
macotherapies. Natural treatments as alternative ity to 3, 4-methylenedioxymethamphetamine
ways to restore normal brain homeostasis should (Ecstasy) in serotonin transporter-deficient
also prove beneficial in continuing to inform mice. Molecular Pharmacology 53, 649655.
researchers on the complex neurological disorder Berger, M., Gray, J.A., and Roth, B.L. (2009). The
of addiction. expanded biology of serotonin. Annual Review
of Medicine 60, 355366.
References Berglind, W.J., See, R.E., Fuchs, R.A., Ghee, S.M.,
Addolorato, G., Leggio, L., Hopf, F.W., Diana, M., Whitfield, T.W., Jr., Miller, S.W., and McGinty, J.F.
and Bonci, A. (2012). Novel therapeutic strategies (2007). A BDNF infusion into the medial pre-
for alcohol and drug addiction:Focus on GABA, frontal cortex suppresses cocaine seeking in
ion channels and transcranial magnetic stimula- rats. The European Journal of Neuroscience 26,
tion. Neuropsychopharmacology 37, 163177. 757766.
528 Part IV:Homeostatic Therapies

Besheer, J., Lindsay, T.G., OBuckley, T.K., Hodge, Carnicella, S., and Ron, D. (2009). GDNFa poten-
C.W., and Morrow, A.L. (2010). Pregnenolone tial target to treat addiction. Pharmacology &
and ganaxolone reduce operant ethanol Therapeutics 122,918.
self-administration in alcohol-preferring p rats. Carroll, M.E., Lynch, W.J., Roth, M.E., Morgan, A.D.,
Alcoholism, Clinical and Experimental Research and Cosgrove, K.P. (2004). Sex and estrogen
34, 20442052. influence drug abuse. Trends in Pharmacological
Blum, K., Chen, A.L., Giordano, J., Borsten, J., Sciences 25, 273279.
Chen, T.J., Hauser, M., Simpatico, T., Femino, J., Chang, E., Fung, L.C., Li, C.S., Lin, T.C., Tam, L.,
Braverman, E.R., and Barh, D. (2012). The addic- Tang, C., and Tong, E.K. (2013). Offering acu-
tive brain:All roads lead to dopamine. Journal of puncture as an adjunct for tobacco cessa-
Psychoactive Drugs 44, 134143. tion: A community clinic experience. Health
Bobzean, S.A., DeNobrega, A.K., and Perrotti, L.I. Promotion Practice 14, 80S87S.
(2014). Sex differences in the neurobiology Chauvet, C., Goldberg, S.R., Jaber, M., and Solinas, M.
of drug addiction. Experimental Neurology (2012). Effects of environmental enrich-
259,6474. ment on the incubation of cocaine craving.
Boison, D. (2011). Modulators of nucleoside metab- Neuropharmacology 63, 635641.
olism in the therapy of brain diseases. Current Choi, K.H., Edwards, S., Graham, D.L., Larson, E.B.,
topics in Medicinal Chemistry 11, 10681086. Whisler, K.N., Simmons, D., Friedman, A.K.,
Boison, D. (2013). Role of adenosine in status epilep- Walsh, J.J., Rahman, Z., Monteggia, L.M., etal.
ticus:Apotential new target? Epilepsia 54 Suppl (2011). Reinforcement-related regulation of
6,2022. AMPA glutamate receptor subunits in the ventral
Bolanos, C.A., and Nestler, E.J. (2004). Neurotrophic tegmental area enhances motivation for cocaine.
mechanisms in drug addiction. Neuromolecular The Journal of Neuroscience 31, 79277937.
Medicine 5,6983. Clapper, J.R., Mangieri, R.A., and Piomelli, D.
Bowen, S., and Enkema, M.C. (2014). Relationship (2009). The endocannabinoid system as a tar-
between dispositional mindfulness and sub- get for the treatment of cannabis dependence.
stance use: Findings from a clinical sample. Neuropharmacology 56 Suppl 1, 235243.
Addictive Behaviors 39, 532537. Conrad, K.L., Tseng, K.Y., Uejima, J.L., Reimers, J.M.,
Brabant, C., Alleva, L., Quertemont, E., and Tirelli, Heng, L.J., Shaham, Y., Marinelli, M., and
E. (2010). Involvement of the brain histaminergic Wolf, M.E. (2008). Formation of accumbens
system in addiction and addiction-related behav- GluR2-lacking AMPA receptors mediates incu-
iors:Acomprehensive review with emphasis on bation of cocaine craving. Nature 454, 118121.
the potential therapeutic use of histaminergic Crews, F.T., and Vetreno, R.P. (2011). Addiction, ado-
compounds in drug dependence. Progress in lescence, and innate immune gene induction.
Neurobiology 92, 421441. Frontiers in Psychiatry2,19.
Brewer, J.A., Bowen, S., Smith, J.T., Marlatt, G.A., Crews, F.T., Zou, J., and Qin, L. (2011). Induction of
and Potenza, M.N. (2010). Mindfulness-based innate immune genes in brain create the neu-
treatments for co-occurring depression and sub- robiology of addiction. Brain, Behavior, and
stance use disorders:What can we learn from the Immunity 25 Suppl 1, S4-S12.
brain? Addiction 105, 16981706. Cummings, D.E., Naleid, A.M., and Figlewicz
Brodie, J.D., Case, B.G., Figueroa, E., Dewey, S.L., Lattemann, D.P. (2007). Ghrelin:Alink between
Robinson, J.A., Wanderling, J.A., and Laska, energy homeostasis and drug abuse? Addiction
E.M. (2009). Randomized, double-blind, Biology 12,15.
placebo-controlled trial of vigabatrin for the Cummings, J.A., Jagannathan, L., Jackson, L.R., and
treatment of cocaine dependence in Mexican Becker, J.B. (2014). Sex differences in the effects
parolees. The American Journal of Psychiatry of estradiol in the nucleus accumbens and stria-
166, 12691277. tum on the response to cocaine:Neurochemistry
Burmeister, J.J., Lungren, E.M., and Neisewander, J.L. and behavior. Drug and Alcohol Dependence
(2003). Effects of fluoxetine and d-fenfluramine 135,2228.
on cocaine-seeking behavior in rats. Czirr, E., and Wyss-Coray, T. (2012). The immu-
Psychopharmacology 168, 146154. nology of neurodegeneration. The Journal of
Carnicella, S., Kharazia, V., Jeanblanc, J., Janak, P.H., Clinical Investigation 122, 11561163.
and Ron, D. (2008). GDNF is a fast-acting potent Dakwar, E., and Levin, F.R. (2009). The emerging
inhibitor of alcohol consumption and relapse. role of meditation in addressing psychiatric ill-
Proceedings of the National Academy of Sciences ness, with a focus on substance use disorders.
of the United States of America 105, 81148119. Harvard Review of Psychiatry 17, 254267.
Brain Homeostasis and Addiction 529

Dalley, J.W., and Roiser, J.P. (2012). Dopamine, sero- Fattore, L., Puddu, M.C., Picciau, S., Cappai, A.,
tonin and impulsivity. Neuroscience 215,4258. Fratta, W., Serra, G.P., and Spiga, S. (2002).
Danovitch, I., and Gorelick, D.A. (2012). State of Astroglial in vivo response to cocaine in mouse
the art treatments for cannabis dependence. dentate gyrus: A quantitative and qualitative
Psychiatric Clinics of North America 35, 309326. analysis by confocal microscopy. Neuroscience
Daws, L.C., Montanez, S., Munn, J.L., Owens, W.A., 110,16.
Baganz, N.L., Boyce-Rustay, J.M., Millstein, R.A., Feltenstein, M.W., and See, R.E. (2008). The neu-
Wiedholz, L.M., Murphy, D.L., and Holmes, A. rocircuitry of addiction: An overview. British
(2006). Ethanol inhibits clearance of brain sero- Journal of Pharmacology 154, 261274.
tonin by a serotonin transporter-independent Filip, M., and Frankowska, M. (2008). GABA(B)
mechanism. The Journal of Neuroscience 26, receptors in drug addiction. Pharmacological
64316438. Reports 60, 755770.
Dawson, A., Miles, M.F., and Damaj, M.I. (2013). Forest, E. (2008). Atypical drugs of abuse. Student
The beta2 nicotinic acetylcholine receptor sub- Doctor Network. http://www.studentdoctor.
unit differentially influences ethanol behavioral net/2008/07/atypical-drugs-of-abuse/
effects in the mouse. Alcohol 47,8594. Forget, B., Coen, K.M., and Le Foll, B. (2009). Inhibition
de Dios, M.A., Herman, D.S., Britton, W.B., Hagerty, of fatty acid amide hydrolase reduces reinstate-
C.E., Anderson, B.J., and Stein, M.D. (2012). ment of nicotine seeking but not break point for
Motivational and mindfulness intervention for nicotine self-administrationcomparison with
young adult female marijuana users. Journal of CB(1) receptor blockade. Psychopharmacology
Substance Abuse Treatment 42,5664. 205, 613624.
Deister, C., and Schmidt, C.E. (2006). Optimizing Fulton, S., Pissios, P., Manchon, R.P., Stiles, L., Frank, L.,
neurotrophic factor combinations for neurite Pothos, E.N., Maratos-Flier, E., and Flier, J.S.
outgrowth. Journal of Neural Engineering 3, (2006). Leptin regulation of the mesoaccumbens
172179. dopamine pathway. Neuron 51, 811822.
Detar, D.T. (2011). Alcoholics anonymous and other Fulton, S., Woodside, B., and Shizgal, P. (2000).
twelve-step programs in recovery. Primary Care Modulation of brain reward circuitry by leptin.
38, 143148. Science 287, 125128.
Devaud, L.L., Walls, S.A., McCulley, W.D., 3rd, and Garg, A., and Aggarwal, B.B. (2002). Nuclear tran-
Rosenwasser, A.M. (2012). Voluntary wheel run- scription factor-kappaB as a target for cancer
ning attenuates ethanol withdrawal-induced drug development. Leukemia 16, 10531068.
increases in seizure susceptibility in male and Goldman, D., Oroszi, G., and Ducci, F. (2005). The
female rats. Pharmacology, Biochemistry, and genetics of addictions: Uncovering the genes.
Behavior 103,1825. Nature Reviews Genetics 6, 521532.
Dinarello, C.A. (2000). Proinflammatory cytokines. Gonzalez, R., Chavez-Pascacio, K., and Meneses, A.
Chest 118, 503508. (2013). Role of 5-HT5A receptors in the consoli-
Dingledine, R., Borges, K., Bowie, D., and Traynelis, dation of memory. Behavioural Brain Research
S.F. (1999). The glutamate receptor ion channels. 252, 246251.
Pharmacological Reviews 51,761. Grant, J.E., and Potenza, M.N. (2006). Compulsive
Diogenes, M.J., Neves-Tome, R., Fucile, S., Martinello, aspects of impulse-control disorders. Psychiatric
K., Scianni, M., Theofilas, P., Lopatar, J., Clinics of North America 29, 539551,x.
Ribeiro, J.A., Maggi, L., Frenguelli, B.G., et al. Grimm, J.W., Lu, L., Hayashi, T., Hope, B.T., Su, T.P.,
(2014). Homeostatic control of synaptic activity and Shaham, Y. (2003). Time-dependent increases
by endogenous adenosine is mediated by adenos- in brain-derived neurotrophic factor protein
ine kinase. Cerebral Cortex 24,6780. levels within the mesolimbic dopamine system
Dohrman, D.P., Diamond, I., and Gordon, A.S. after withdrawal from cocaine:Implications for
(1997). The role of the neuromodulator adenos- incubation of cocaine craving. The Journal of
ine in alcohols actions. Alcohol Health and Neuroscience 23, 742747.
Research World 21, 136143. Gual, A., He, Y., Torup, L., van den Brink, W.,
Duke, A.N., Kaminski, B.J., and Weerts, E.M. Mann, K., and Group, E.S. (2013). A ran-
(2014). Baclofen effects on alcohol seeking, domised, double-blind, placebo-controlled,
self-administration and extinction of seeking efficacy study of nalmefene, as-needed use, in
responses in a within-session design in baboons. patients with alcohol dependence. European
Addiction Biology 19,1626. Neuropsychopharmacology 23, 14321442.
Ernst, E. (2006). Acupuncturea critical analysis. Halpern, J.H., Sherwood, A.R., Hudson, J.I., Gruber,
Journal of Internal Medicine 259, 125137. S., Kozin, D., and Pope, H.G., Jr. (2011). Residual
530 Part IV:Homeostatic Therapies

neurocognitive features of long-term ecstasy Itier, V., and Bertrand, D. (2001). Neuronal nicotinic
users with minimal exposure to other drugs. receptors: From protein structure to function.
Addiction 106, 777786. FEBS Letters 504, 118125.
Halpert, A.G., Olmstead, M.C., and Beninger, R.J. Ito, H.T., and Schuman, E.M. (2008).
(2003). Dimenhydrinate produces a condi- Frequency-dependent signal transmission and
tioned place preference in rats. Pharmacology, modulation by neuromodulators. Frontiers in
Biochemistry, and Behavior 75, 173179. Neuroscience 2, 138144.
Hao, Y., Yang, J.Y., Wu, C.F., and Wu, M.F. (2007). Jerlhag, E., Egecioglu, E., Dickson, S.L., Douhan,
Pseudoginsenoside-F11 decreases morphine- A., Svensson, L., and Engel, J.A. (2007). Ghrelin
induced behavioral sensitization and extracellu- administration into tegmental areas stimulates
lar glutamate levels in the medial prefrontal cor- locomotor activity and increases extracellu-
tex in mice. Pharmacology, Biochemistry, and lar concentration of dopamine in the nucleus
Behavior 86, 660666. accumbens. Addiction Biology 12,616.
Heidbreder, C. (2011). Advances in animal models Jerlhag, E., Egecioglu, E., Landgren, S., Salome, N.,
of drug addiction. Current Topics in Behavioral Heilig, M., Moechars, D., Datta, R., Perrissoud, D.,
Neurosciences 7, 213250. Dickson, S.L., and Engel, J.A. (2009). Requirement
Helms, C.M., Rossi, D.J., and Grant, K.A. (2012). of central ghrelin signaling for alcohol reward.
Neurosteroid influences on sensitivity to etha- Proceedings of the National Academy of Sciences
nol. Frontiers in Endocrinology3,10. of the United States of America 106, 1131811323.
Herman, A.I., and Balogh, K.N. (2012). Jung, K.M., Clapper, J.R., Fu, J., DAgostino, G.,
Polymorphisms of the serotonin transporter and Guijarro, A., Thongkham, D., Avanesian, A.,
receptor genes: Susceptibility to substance abuse. Astarita, G., DiPatrizio, N.V., Frontini, A., etal.
Substance Abuse and Rehabilitation 3,4957. (2012). 2-arachidonoylglycerol signaling in fore-
Higgins, G.A., and Fletcher, P.J. (2003). Serotonin brain regulates systemic energy metabolism. Cell
and drug reward: Focus on 5-HT2C receptors. Metabolism 15, 299310.
European Journal of Pharmacology 480, 151162. Kalivas, P.W. (2009). The glutamate homeosta-
Hiipakka, R.A., and Liao, S. (1998). Molecular sis hypothesis of addiction. Nature Reviews
mechanism of androgen action. Trends in Neuroscience 10, 561572.
Endocrinology and Metabolism 9, 317324. Kelso, M.L., Liput, D.J., Eaves, D.W., and Nixon, K.
Hobson, B.D., ONeill, C.E., Levis, S.C., Monteggia, (2011). Upregulated vimentin suggests new areas
L.M., Neve, R.L., Self, D.W., and Bachtell, of neurodegeneration in a model of an alcohol
R.K. (2013). Adenosine A1 and dopamine d1 use disorder. Neuroscience 197, 381393.
receptor regulation of AMPA receptor phos- Kirchner, H., Heppner, K.M., and Tschop, M.H.
phorylation and cocaine-seeking behavior. (2012). The role of ghrelin in the control of
Neuropsychopharmacology 38, 19741983. energy balance. Handbook of Experimental
Hlzel, B.K., Lazar, S.W., Gard, T., Schuman-Olivier, Z., Pharmacology, 161184.
Vago, D.R., and Ott, U. (2011). How does mind- Kitanaka, J., Kitanaka, N., Hall, F.S., Uhl, G.R.,
fulness meditation work? Proposing mechanisms Tatsuta, T., Morita, Y., Tanaka, K., Nishiyama, N.,
of action from a conceptual and neural perspec- and Takemura, M. (2011). Histamine H3 receptor
tive. Perspectives on Psychological Science 6, agonists decrease hypothalamic histamine levels
537559. and increase stereotypical biting in mice chal-
Hoofnagle, J.H. (2012). Nalmefene. National Institute lenged with methamphetamine. Neurochemical
of Diabetes and Digestive and Kidney Diseases. Research 36, 18241833.
http://livertox.nih.gov/Nalmefene.htm Klok, M.D., Jakobsdottir, S., and Drent, M.L.
Hu, M., Crombag, H.S., Robinson, T.E., and Becker, (2007). The role of leptin and ghrelin in the
J.B. (2004). Biological basis of sex differences regulation of food intake and body weight in
in the propensity to self-administer cocaine. humans: A review. Obesity Reviews: an official
Neuropsychopharmacology 29,8185. journal of the International Association for the
Hurst, R., Rollema, H., and Bertrand, D. (2013). Study of Obesity 8,2134.
Nicotinic acetylcholine receptors: From basic sci- Koob, G.F., and Le Moal, M. (2008). Addiction and
ence to therapeutics. Pharmacology & Therapeutics the brain antireward system. Annual Review of
137, 2254. Psychology 59,2953.
Ikemoto, S. (2007). Dopamine reward circuitry:Two Kronfol, Z., and Remick, D.G. (2000). Cytokines
projection systems from the ventral midbrain to and the brain:Implications for clinical psychia-
the nucleus accumbens-olfactory tubercle com- try. The American Journal of Psychiatry 157,
plex. Brain Research Reviews 56,2778. 683694.
Brain Homeostasis and Addiction 531

LaLumiere, R.T., and Kalivas, P.W. (2008). Matthews, J., and Gustafsson, J.A. (2003). Estrogen
Glutamate release in the nucleus accumbens core signaling: A subtle balance between ER alpha
is necessary for heroin seeking. The Journal of and ER beta. Molecular Interventions 3, 281292.
Neuroscience 28, 31703177. McEwen, B. (2002). Estrogen actions throughout the
Le Moal, M., and Koob, G.F. (2007). Drug addic- brain. Recent Progress in Hormone Research 57,
tion: Pathways to the disease and patho- 357384.
physiological perspectives. European Meeusen, R., Smolders, I., Sarre, S., de Meirleir, K.,
Neuropsychopharmacology 17, 377393. Keizer, H., Serneels, M., Ebinger, G., and
Leggio, L., Garbutt, J.C., and Addolorato, G. (2010). Michotte, Y. (1997). Endurance training effects
Effectiveness and safety of baclofen in the treat- on neurotransmitter release in rat striatum:An
ment of alcohol dependent patients. CNS & in vivo microdialysis study. Acta Physiologica
Neurological Disorders Drug Targets 9,3344. Scandinavica 159, 335341.
Leggio, L., Zywiak, W.H., McGeary, J.E., Edwards, Mellon, S.H., and Griffin, L.D. (2002).
S., Fricchione, S.R., Shoaff, J.R., Addolorato, G., Neurosteroids: Biochemistry and clinical sig-
Swift, R.M., and Kenna, G.A. (2013). A human nificance. Trends in endocrinology and metabo-
laboratory pilot study with baclofen in alcoholic lism:TEM 13,3543.
individuals. Pharmacology, Biochemistry, and Meredith, G.E., Callen, S., and Scheuer, D.A. (2002).
Behavior 103, 784791. Brain-derived neurotrophic factor expression is
Li, T., Ren, G., Lusardi, T., Wilz, A., Lan, J.Q., increased in the rat amygdala, piriform cortex
Iwasato, T., Itohara, S., Simon, R.P., and Boison, and hypothalamus following repeated amphet-
D. (2008). Adenosine kinase is a target for the amine administration. Brain Research 949,
prediction and prevention of epileptogenesis in 218227.
mice. The Journal of Clinical Investigation 118, Messer, C.J., Eisch, A.J., Carlezon, W.A., Jr., Whisler,
571582. K., Shen, L., Wolf, D.H., Westphal, H., Collins,
Lingford-Hughes, A., Watson, B., Kalk, N., and Reid, F., Russell, D.S., and Nestler, E.J. (2000). Role for
A. (2010). Neuropharmacology of addiction GDNF in biochemical and behavioral adapta-
and how it informs treatment. British Medical tions to drugs of abuse. Neuron 26, 247257.
Bulletin 96, 93110. Mo, Q., Lu, S.F., and Simon, N.G. (2006).
Logrip, M.L., Janak, P.H., and Ron, D. (2009). Dehydroepiandrosterone and its metabolites:
Escalating ethanol intake is associated with Differential effects on androgen receptor traf-
altered corticostriatal BDNF expression. Journal ficking and transcriptional activity. The Journal
of Neurochemistry 109, 14591468. of Steroid Biochemistry and Molecular Biology
Lu, L., Dempsey, J., Liu, S.Y., Bossert, J.M., and 99,5058.
Shaham, Y. (2004). A single infusion of Moran, M.M., McFarland, K., Melendez, R.I., Kalivas,
brain-derived neurotrophic factor into the ven- P.W., and Seamans, J.K. (2005). Cystine/gluta-
tral tegmental area induces long-lasting poten- mate exchange regulates metabotropic glutamate
tiation of cocaine seeking after withdrawal. The receptor presynaptic inhibition of excitatory
Journal of Neuroscience 24, 16041611. transmission and vulnerability to cocaine seek-
Lu, L., Liu, Y., Zhu, W., Shi, J., Liu, Y., Ling, W., and ing. The Journal of Neuroscience 25, 63896393.
Kosten, T.R. (2009). Traditional medicine in Morisset, S., Pilon, C., Tardivel-Lacombe, J.,
the treatment of drug addiction. The American Weinstein, D., Rostene, W., Betancur, C.,
Journal of Drug and Alcohol Abuse 35,111. Sokoloff, P., Schwartz, J.C., and Arrang, J.M.
Lukas, S.E., Penetar, D., Berko, J., Vicens, L., Palmer, (2002). Acute and chronic effects of methamphet-
C., Mallya, G., Macklin, E.A., and Lee, D.Y. amine on tele-methylhistamine levels in mouse
(2005). An extract of the Chinese herbal root brain:Selective involvement of the D(2) and not
kudzu reduces alcohol drinking by heavy drink- D(3) receptor. The Journal of Pharmacology and
ers in a naturalistic setting. Alcoholism, Clinical Experimental Therapeutics 300, 621628.
and Experimental Research 29, 756762. Murray, J.E., Everitt, B.J., and Belin, D. (2012).
Lutz, P.E., and Kieffer, B.L. (2013). The multiple fac- N-Acetylcysteine reduces early- and late-stage
ets of opioid receptor function:Implications for cocaine seeking without affecting cocaine taking
addiction. Current Opinion in Neurobiology 23, in rats. Addiction Biology 17, 437440.
473479. Nader, J., Chauvet, C., Rawas, R.E., Favot, L., Jaber, M.,
Maldonado, R., Robledo, P., and Berrendero, F. Thiriet, N., and Solinas, M. (2012). Loss of envi-
(2013). Endocannabinoid system and drug addic- ronmental enrichment increases vulnerability to
tion:New insights from mutant mice approaches. cocaine addiction. Neuropsychopharmacology
Current Opinion in Neurobiology 23, 480486. 37, 15791587.
532 Part IV:Homeostatic Therapies

National Drug Intelligence Center. (2011). The from a community clinic sample. The Journal of
Economic Impact of Illicit Drug Use on Clinical Psychiatry 65,8486.
American Society. Washington, DC: United Raivio, N., Tiraboschi, E., Saarikoski, S.T., Castren,
States Department of Justice. E., and Kiianmaa, K. (2012). Brain-derived
National Institute on Drug Abuse. (2012). neurotrophic factor expression after acute
Commonly abused drugs. http://www.druga- administration of ethanol. European Journal of
buse.gov/drugs-abuse/commonly-abused-drugs/ Pharmacology 687,913.
commonly-abused-drugs-chart Ramesh, D., Ross, G.R., Schlosburg, J.E., Owens,
Neil, J.C., and Kulkarni, J. (2011). Biological basis of R.A., Abdullah, R.A., Kinsey, S.G., Long, J.Z.,
sex differences in psychopharmacology: Preface. Nomura, D.K., Sim-Selley, L.J., Cravatt, B.F.,
Current Topics in Behavioral Neurosciences 8,vvii. et al. (2011). Blockade of endocannabinoid
Nichols, D.E., and Nichols, C.D. (2008). Serotonin hydrolytic enzymes attenuates precipitated opi-
receptors. Chemical Reviews 108, 16141641. oid withdrawal symptoms in mice. The Journal
Nie, H., Rewal, M., Gill, T.M., Ron, D., and Janak, P.H. of Pharmacology and Experimental Therapeutics
(2011). Extrasynaptic delta-containing GABAA 339, 173185.
receptors in the nucleus accumbens dorsomedial Rashidy-Pour, A., Pahlevani, P., Vaziri, A., Shaigani,
shell contribute to alcohol intake. Proceedings of P., Zarepour, L., Vafaei, A.A., and Haghparast,
the National Academy of Sciences of the United A. (2013). Involvement of CB1 receptors in the
States of America 108, 44594464. ventral tegmental area in the potentiation of
Overstreet, D.H., Keung, W.M., Rezvani, A.H., morphine rewarding properties in acquisition
Massi, M., and Lee, D.Y. (2003). Herbal remedies but not expression in the conditioned place pref-
for alcoholism: Promises and possible pitfalls. erence model. Behavioural Brain Research 247,
Alcoholism, Clinical and Experimental Research 259267.
27, 177185. Reddy, D.S. (2010). Neurosteroids:Endogenous role
Pacher, P., Batkai, S., and Kunos, G. (2006). The in the human brain and therapeutic potentials.
endocannabinoid system as an emerging target Progress in Brain Research 186, 113137.
of pharmacotherapy. Pharmacological Reviews Ren, X., Noda, Y., Mamiya, T., Nagai, T., and
58, 389462. Nabeshima, T. (2004). A neuroactive steroid,
Panula, P., and Nuutinen, S. (2013). The histaminer- dehydroepiandrosterone sulfate, prevents the
gic network in the brain:Basic organization and development of morphine dependence and toler-
role in disease. Nature Reviews Neuroscience 14, ance via c-fos expression linked to the extracellu-
472487. lar signal-regulated protein kinase. Behavioural
Parrott, A.C. (2002). Recreational Ecstasy/MDMA, Brain Research 152, 243250.
the serotonin syndrome, and serotonergic neu- Reynolds, M.D., Tarter, R., Kirisci, L., Kirillova, G.,
rotoxicity. Pharmacology, Biochemistry, and Brown, S., Clark, D.B., and Gavaler, J. (2007).
Behavior 71, 837844. Testosterone levels and sexual maturation
Parsegian, A., and See, R.E. (2014). Dysregulation of predict substance use disorders in adolescent
dopamine and glutamate release in the prefrontal boys:Aprospective study. Biological Psychiatry
cortex and nucleus accumbens following meth- 61, 12231227.
amphetamine self-administration and during Robinson, M.B. (1998). The family of
reinstatement in rats. Neuropsychopharmacology sodium-dependent glutamate transporters:
39, 811822. A focus on the GLT-1/EAAT2 subtype.
Pertwee, R.G. (2006). The pharmacology of canna- Neurochemistry International 33, 479491.
binoid receptors and their ligands:An overview. Rogers, G., Elston, J., Garside, R., Roome, C., Taylor,
International Journal of Obesity 30 Suppl 1, R., Younger, P., Zawada, A., and Somerville, M.
S1318. (2009). The harmful health effects of recreational
Pierce, R.C., and Kumaresan, V. (2006). The meso- ecstasy: A systematic review of observational
limbic dopamine system: The final common evidence. Health Technology Assessment 13,
pathway for the reinforcing effect of drugs of iiiiv, ixxii,1315.
abuse? Neuroscience and Biobehavioral Reviews Sarviharju, M., Hyytia, P., Hervonen, A., Jaatinen,
30, 215238. P., Kiianmaa, K., and Korpi, E.R. (2006). Lifelong
Poon, V.Y., Choi, S., and Park, M. (2013). Growth fac- ethanol consumption and brain regional
tors in synaptic function. Frontiers in Synaptic GABAA receptor subunit mRNA expression in
Neuroscience5,6. alcohol-preferring rats. Alcohol 40, 159166.
Raby, W.N., and Coomaraswamy, S. (2004). Sawyer, E.K., Mun, J., Nye, J.A., Kimmel, H.L., Voll,
Gabapentin reduces cocaine use among addicts R.J., Stehouwer, J.S., Rice, K.C., Goodman, M.M.,
Brain Homeostasis and Addiction 533

and Howell, L.L. (2012). Neurobiological changes Smolinska, S., Jutel, M., Crameri, R., and OMahony,
mediating the effects of chronic fluoxetine on L. (2014). Histamine and gut mucosal immune
cocaine use. Neuropsychopharmacology 37, regulation. Allergy 69, 273281.
18161824. Snider, S.E., Hendrick, E.S., and Beardsley, P.M.
Schilt, T., Koeter, M.W., Smal, J.P., Gouwetor, (2013). Glial cell modulators attenuate meth-
M.N., van den Brink, W., and Schmand, B. amphetamine self-administration in the
(2010). Long-term neuropsychological effects of rat. European Journal of Pharmacology 701,
ecstasy in middle-aged ecstasy/polydrug users. 124130.
Psychopharmacology 207, 583591. Sotomayor-Zarate, R., Gysling, K., Busto, U.E.,
Schmaal, L., Veltman, D.J., Nederveen, A., van Cassels, B.K., Tampier, L., and Quintanilla, M.E.
den Brink, W., and Goudriaan, A.E. (2012). (2013). Varenicline and cytisine: Two nicotinic
N-acetylcysteine normalizes glutamate levels acetylcholine receptor ligands reduce etha-
in cocaine-dependent patients: A randomized nol intake in University of Chile bibulous rats.
crossover magnetic resonance spectroscopy Psychopharmacology 227, 287298.
study. Neuropsychopharmacology 37, 21432152. Tan, K.R., Brown, M., Labouebe, G., Yvon, C., Creton,
Schmidt, L.S., Thomsen, M., Weikop, P., Dencker, C., Fritschy, J.M., Rudolph, U., and Luscher, C.
D., Wess, J., Woldbye, D.P., Wortwein, (2010). Neural bases for addictive properties of
G., and Fink-Jensen, A. (2011). Increased benzodiazepines. Nature 463, 769774.
cocaine self-administration in M4 musca- Thomsen, M., Woldbye, D.P., Wortwein, G.,
rinic acetylcholine receptor knockout mice. Fink-Jensen, A., Wess, J., and Caine, S.B. (2005).
Psychopharmacology 216, 367378. Reduced cocaine self-administration in musca-
Schmoutz, C.D., Runyon, S.P., and Goeders, N.E. rinic M5 acetylcholine receptor-deficient mice.
(2014). Effects of inhibitory GABA-active neuros- The Journal of Neuroscience 25, 81418149.
teroids on cocaine seeking and cocaine taking in Tien, L.T., Cai, Z., Rhodes, P.G., and Fan, L.W.
rats. Psychopharmacology 231, 33913400. (2011). Neonatal exposure to lipopolysaccha-
Sebastiao, A.M., and Ribeiro, J.A. (2009). Adenosine ride enhances methamphetamine-induced
receptors and the central nervous system. reinstated behavioral sensitization in adult rats.
Handbook of Experimental Pharmacology, Behavioural Brain Research 224, 166173.
471534. Tien, L.T., Kaizaki, A., Pang, Y., Cai, Z., Bhatt,
Shalev, U., Yap, J., and Shaham, Y. (2001). Leptin A.J., and Fan, L.W. (2013). Neonatal exposure
attenuates acute food deprivation-induced to lipopolysaccharide enhances accumulation
relapse to heroin seeking. The Journal of of alpha-synuclein aggregation and dopamine
Neuroscience 21,RC129. transporter protein expression in the substantia
Shen, Y., Lindemeyer, A.K., Spigelman, I., Sieghart, W., nigra in responses to rotenone challenge in later
Olsen, R.W., and Liang, J. (2011). Plasticity of life. Toxicology 308, 96103.
GABAA receptors after ethanol pre-exposure Tomek, S.E., Lacrosse, A.L., Nemirovsky, N.E.,
in cultured hippocampal neurons. Molecular and Olive, M.F. (2013). NMDA receptor mod-
Pharmacology 79, 432442. ulators in the treatment of drug addiction.
Slattery, D.A., Markou, A., Froestl, W., and Cryan, Pharmaceuticals 6, 251268.
J.F. (2005). The GABAB receptor-positive modu- Uusi-Oukari, M., and Korpi, E.R. (2010). Regulation
lator GS39783 and the GABAB receptor agonist of GABA(A) receptor subunit expression by
baclofen attenuate the reward-facilitating effects pharmacological agents. Pharmacological
of cocaine: Intracranial self-stimulation stud- Reviews 62, 97135.
ies in the rat. Neuropsychopharmacology 30, Vallee, M., Vitiello, S., Bellocchio, L.,
20652072. Hebert-Chatelain, E., Monlezun, S.,
Smith, M.A., and Lynch, W.J. (2011). Exercise as a Martin-Garcia, E., Kasanetz, F., Baillie, G.L.,
potential treatment for drug abuse:Evidence from Panin, F., Cathala, A., etal. (2014). Pregnenolone
preclinical studies. Frontiers in Psychiatry2,82. can protect the brain from cannabis intoxication.
Smith, M.A., Pennock, M.M., Walker, K.L., and Science 343,9498.
Lang, K.C. (2012). Access to a running wheel van den Brink, W., Aubin, H.J., Bladstrom, A.,
decreases cocaine-primed and cue-induced rein- Torup, L., Gual, A., and Mann, K. (2013). Efficacy
statement in male and female rats. Drug and of as-needed nalmefene in alcohol-dependent
Alcohol Dependence 121,5461. patients with at least a high drinking risk
Smith, M.A., and Pitts, E.G. (2012). Wheel running level: Results from a subgroup analysis of two
decreases the positive reinforcing effects of her- randomized controlled 6-month studies. Alcohol
oin. Pharmacological Reports 64, 960964. and Alcoholism 48, 570578.
534 Part IV:Homeostatic Therapies

Vetter-OHagen, C.S., Sanders, K.W., and Spear, Wu, M., Sahbaie, P., Zheng, M., Lobato, R., Boison, D.,
L.P. (2011). Evidence for suppressant effects of Clark, J.D., and Peltz, G. (2013). Opiate-induced
testosterone on sex-typical ethanol intake in changes in brain adenosine levels and narcotic
male Sprague-Dawley rats. Behavioural Brain drug responses. Neuroscience 228, 235242.
Research 224, 403407. Wu, N., Lu, X.Q., Yan, H.T., Su, R.B., Wang, J.F., Liu, Y.,
Vetulani, J. (2001). Drug addiction: Part II. Hu, G., and Li, J. (2008). Aquaporin 4 deficiency
Neurobiology of addiction. Polish Journal of modulates morphine pharmacological actions.
Pharmacology 53, 303317. Neuroscience Letters 448, 221225.
Viveros, M.P., Mendrek, A., Paus, T., Xie, X., Arguello, A.A., Reittinger, A.M., Wells, A.M.,
Lopez-Rodriguez, A.B., Marco, E.M., Yehuda, and Fuchs, R.A. (2012). Role of nicotinic acetyl-
R., Cohen, H., Lehrner, A., and Wagner, E.J. choline receptors in the effects of cocaine-paired
(2012). A comparative, developmental, and clini- contextual stimuli on impulsive decision making
cal perspective of neurobehavioral sexual dimor- in rats. Psychopharmacology 223, 271279.
phisms. Frontiers in Neuroscience6,84. Yamada, K., and Nabeshima, T. (2004). Pro- and
Volkow, N.D., Wang, G.J., Fowler, J.S., and Tomasi, anti-addictive neurotrophic factors and cyto-
D. (2012). Addiction circuitry in the human kines in psychostimulant addiction:Mini review.
brain. Annual Review of Pharmacology and Annals of the New York Academy of Sciences
Toxicology 52, 321336. 1025, 198204.
Wang, H., and Lupica, C.R. (2014). Release of Yan, C.Z., and Hou, Y.N. (2004). Effects of morphine
endogenous cannabinoids from ventral teg- dependence and withdrawal on levels of neuros-
mental area dopamine neurons and the teroids in rat brain. Acta Pharmacologica Sinica
modulation of synaptic processes. Progress 25, 12851291.
in Neuro-Psychopharmacology & Biological Yan, H.T., Wu, N., Lu, X.Q., Su, R.B., Zheng, J.Q.,
Psychiatry 52,2427. and Li, J. (2013a). Aquaporin-4 deficiency atten-
Weinstock, J., Barry, D., and Petry, N.M. (2008). uates opioid dependence through suppress-
Exercise-related activities are associated with ing glutamate transporter-1 down-regulation
positive outcome in contingency management and maintaining glutamate homeostasis. CNS
treatment for substance use disorders. Addictive Neuroscience & Therapeutics 19,1219.
Behaviors 33, 10721075. Yan, Y., Miyamoto, Y., Nitta, A., Muramatsu, S.,
White, A. (2013). Trials of acupuncture for drug Ozawa, K., Yamada, K., and Nabeshima, T.
dependence: A recommendation for hypoth- (2013b). Intrastriatal gene delivery of
eses based on the literature. Acupuncture in GDNF persistently attenuates metham-
Medicine 31, 297304. phetamine self-administration and relapse
Wisniewski, A.B., Brown, T.T., John, M., in mice. The International Journal of
Frankowicz, J.K., Cofranceso, J., Jr., Golub, E.T., Neuropsychopharmacology 16, 15591567.
Ricketts, E.P., and Dobs, A.S. (2007). Yee, A., Loh, H.S., Hisham Hashim, H.M., and Ng,
Hypothalamic-pituitary-gonadal function in C.G. (2014). The prevalence of sexual dysfunc-
men and women using heroin and cocaine, strat- tion among male patients on methadone and
ified by HIV status. Gender Medicine 4,3544. buprenorphine treatments: A meta-analysis
Witkiewitz, K., and Bowen, S. (2010). Depression, study. The Journal of Sexual Medicine 11,2232.
craving, and substance use following a ran- Zernig, G., Ahmed, S.H., Cardinal, R.N., Morgan, D.,
domized trial of mindfulness-based relapse Acquas, E., Foltin, R.W., Vezina, P., Negus, S.S.,
prevention. Journal of Consulting and Clinical Crespo, J.A., Stockl, P., etal. (2007). Explaining
Psychology 78, 362374. the escalation of drug use in substance depen-
Wu, C.F., Liu, Y.L., Song, M., Liu, W., Wang, J.H., dence: Models and appropriate animal labora-
Li, X., and Yang, J.Y. (2003). Protective tory tests. Pharmacology 80, 65119.
effects of pseudoginsenoside-F11 on Zlebnik, N.E., Anker, J.J., and Carroll, M.E. (2012).
methamphetamine-induced neurotoxicity in Exercise to reduce the escalation of cocaine
mice. Pharmacology, Biochemistry, and Behavior self-administration in adolescent and adult rats.
76, 103109. Psychopharmacology 224, 387400.
28
Anxiety and Stress Disorders
E VA M A R A M A R C O A N D M A R A - PA Z V I V E R O S

INTRODUCTION on these brain areas that are key central nodes


in the control of emotional homeostasis and
Neurobiological Basis stress response. The adaptive response to physi-
ofEmotional Homeostasis cal and/or psychological challenges comprises a
All living organisms try to achieve equilibrium peripheral hormonal response through activation
with the changing environment, which is called of the hypothalamic-pituitary-adrenal (HPA)
homeostasis. Emotional homeostasis attempts to axis, together with an autonomic response medi-
achieve a balance in an individuals emotional ated by the sympathetic and parasympathetic
state; however, maintaining stability against nervous systems. These hormonal and autonomic
physical and psychological events that may hap- responses mediate important adaptive functions
pen in everyday life frequently requires a change in preparing the organism for responding to
in homeostasis, known as allostasis. The brain, threat or stress, by increasing vigilance, modu-
through a great variety of complementary and lating memory, mobilizing energy stores, and ele-
often redundant mechanisms, exerts a principal vating cardiovascular function. On the one hand,
role not only in maintaining emotional homeo- glucocorticoid hormones (cortisol in humans
stasis but also in adapting to situations that and corticosterone in rodents), the principle
may threaten survival. Notably, if these adap- endproducts of the HPA axis, act on the brain
tive mechanisms do not cease when the threat to maintain homeostatic balance. First, stress
ends, or are overused by excessive exposure to stimulates the release of corticotropin-releasing
challenges, then the body and brain acquire hormone (CRH) from the paraventricular
cumulative changes, called allostatic load, that nucleus (PVN) of the hypothalamus and amyg-
may increase the risk for psychopathologies, dala. The CRH secretion from the PVN, in turn,
including anxiety and stress-related disorders increases peripheral adrenocorticotropic levels,
(McEwen,2003). which stimulate the adrenal glands to secrete glu-
Emotional processing comprises the cocorticoid hormones. Glucocorticoids activate
evaluation of the emotional salience of the mineralocorticoid and glucocorticoid receptors
stimulus perceived (e.g., appetitive vs. aversive), that, once coupled to the hormone, translocate to
its relationship with previous experiences, and the cell nucleus where they act as regulators of
the context in which it arises. This process also gene transcription. On the other hand, stimula-
includes the behavioral, endocrine, and auto- tion of the lateral nucleus of the hypothalamus
nomic manifestations of the emotional response, activates the sympathetic system, provoking
as well as the subjective feelings that accompany an increase in blood pressure and heart rate,
this response. Abnormal responses to threat or sweating, piloerection, and pupillary dilatation,
stress may lead to fear and anxiety, the emotional whereas the innervation of the parasympathetic
processes that have been the most extensively nervous systems, mainly the vagus and splanch-
studied. In general, the valuation of external nic nerves, from limbic regions is thought to
or internal stimuli is formed by limbic brain mediate visceral symptoms affecting the gastro-
structures, which include the hippocampus, the intestinal and genitourinary systems, also altered
amygdala, and prefrontal cortex (for a review in anxiety disorders (see de Kloet et al., 2005;
see de Kloet et al., 2005; Jankord and Herman, Joels and Baram, 2009; Ulrich-Lai and Herman,
2008; Joels and Baram, 2009). Here we focus 2009, for extended reviews on the subject).
536 Part IV:Homeostatic Therapies

Neural Circuitries hippocampus/ventral subiculum, thus control-


The amygdala is the integration site within the ling the activity of the HPA axis in response to
limbic network where the generation of fear stressors. The ventral hippocampus/ventral sub-
and anxiety occurs in response to environmen- iculum contact PVN projecting neurons in the
tal stimuli. The amygdala is critically involved bed nucleus of the stria terminalis, the medial
in emotion processing; this brain structure preoptic area, the dorsomedial hypothalamus,
receives perceptual informationsimple and and other hypothalamic nuclei. These relay areas
rapidlythrough the sensory thalamus but also are densely populated with GABAergic neu-
highly processed informationmore veridi- rons, allowing for a putative two-neuron relay
cal but slowervia the sensory cortex. Both the between glutamatergic hippocampal outflow
thalamic and the cortical pathway project to the and GABA neurons controlling the HPA stress
lateral nucleus of the amygdala, classically con- response (Herman et al., 2003; Jankord and
sidered the sensory interface of the amygdala Herman,2008).
and a key site of plasticity. In contrast, the lateral Last, the prefrontal cortex has been shown to
nucleus of the amygdala projects, both directly play a key role in the regulation of mood, emo-
and indirectly, to the central nucleus, the out- tion, and stress (Myers-Schulz and Koenigs, 2012;
put region of the amygdala. The central nucleus Shansky and Lipps, 2013). The medial prefrontal
and the medial amygdaloid nucleus have lim- cortex (mPFC) has long been implicated in the
ited projections to the PVN, and also through modulation of anxiety and other emotional behav-
-aminobutyric acid (GABA) neurons to the bed iors. Actually, it is strategically located to modulate
nucleus of the stria terminalis; thus it has been the output of limbic and monoaminergic neuronal
suggested that the activation of the HPA axis may networks and seems to participate in the interpre-
result from a disinhibitory process (Jankord and tation and valuation of the external stimuli, as well
Herman, 2008). Taken together, the amygdala as in the prediction of the social outcomes of the
participates in implicit learning, emotion pro- behavioral response (Nishijo etal., 1988; Damasio,
cessing, and memory storage; it is not only lim- 1998; Baxter et al., 2000). The mPFC modulates
ited to fear. In addition, the amygdala is involved visceral control centers in the brainstem, includ-
in the development of emotional responses to ing cholinergic neurotransmission originating in
the environmental context; in this regard, inputs the basal forebrain (Gaykema etal., 1991), noradr-
from the hippocampus have been related to con- energic (NE) neurons from the locus coeruleus
text conditioning. The amygdala also plays a key (Jodo etal., 1998), dopaminergic neurons project-
role in the emotional modulation of memory, ing from the ventral tegmental area and substantia
emotions influence on attention and percep- nigra (Carr and Sesack, 2000), and serotonergic
tion, social responding, and emotion inhibition (serotonin: 5-hydroxytryptamine [5-HT]) neu-
and regulation (see LeDoux, 2000; Phelps and rons emerging from the dorsal and median raphe
LeDoux, 2005, for review). Human studies have nucleus (Hajos etal., 1998). In addition, the mPFC
further confirmed the implication of the amyg- is a vital component of a distributed extrahypo-
dala in emotional processing. Bursts of electro- thalamic network that modulates activation and
encephalographic activity have been recorded in feedback inhibition of the HPA axis. Different
the amygdala during recollection of specific emo- subregions of the mPFC differentially modulate
tional events (Halgren, 1981), and the electrical the behavioral and systemic response to psycho-
stimulation of the amygdala has been reported logical stress; the prelimbic region of the mPFC
to evoke emotional experiences, mostly fear and suppresses the HPA axis response to acute stress,
anxiety (Gloor etal., 1982), as well as the recol- while the infralimbic cortex activates autonomic
lection of emotionally charged remote memories PVN outputs to promote stress-induced activation
(Brothers,1995). of the HPAaxis.
The hippocampus is involved in the circadian However, neither the prelimbic nor the
glucocorticoid rhythm and inhibition of the HPA infralimbic cortexes directly innervate the PVN;
axis response to stress. Indeed, the hippocampus their connections to modulate the HPA axis relay
can detect a wide range of circulating glucocor- through diverse subcortical nodes (Vertes, 2004;
ticoid concentration given the high expression Holmes and Wellman, 2009). In general, the
of both glucocorticoid and mineralocorticoid amygdala activates the HPA axis in response to
receptors in this brain area (Reul and de Kloet, acute stress whereas the hippocampus and PFC
1986; Herman et al., 1989). Projections to the inhibit the HPA response (Herman et al., 2003;
hypothalamus are largely emitted by the ventral Jankord and Herman, 2008; see Figure28.1).
Anxiety and Stress Disorders 537

External Internal
STIMULUS Environmental mileau

AFFERENT
SYSTEMS

PREFRONTAL
HIPPOCAMPUS
CORTEX
EMOTIONAL
PROCESSING

AMYGDALA

EFFERENT
SYSTEMS PARASYMPATHETIC HYPOTHALAMUS
& SYMPATHETIC PVN
NERVOUS SYSTEM

EMOTIONAL VISCERAL HORMONAL BEHAVIOR


RESPONSES SYMPTOMS RESPONSE RESPONSE

FIGURE 28.1: Scheme of the main neural circuitries involved in emotional processing. PVN, paraventricular
nucleus.

In the past few decades, neuroimaging studies considered to be due to a pathologically hyperac-
have advanced considerably our understanding of tivated and insufficient top-down regulation by
the specific brain regions and circuitries that are frontal brain regions. Worth mentioning, neuro-
affected in anxiety disorders. Indeed, changes in imaging techniques may be considered not only
resting brain metabolism have been described in as a promising tool to reveal the neural circuit-
the scans of patients diagnosed with anxiety dis- ries underlying anxiety disorders but also of great
orders when compared with healthy volunteers. value for the discovery and development of novel
In addition, many studies have been conducted therapeutic tools, as functional neuroimaging
in healthy volunteers after exposure to a fear con- may predict the efficacy and therapeutic value of
ditioning paradigm or during an experimentally drugs (Holzschneider and Mulert,2011).
induced panic attack. One of the most consistent
findings is a hyperactivity of the amygdala dur- Neurochemical Basis
ing symptom provocation that is related to the ofEmotional Homeostasis
experienced symptoms of fear. The anterior cin- A wide variety of neurotransmitter systems are
gulated cortex and the insula are also involved in involved in the complex circuitry that controls
the development and maintenance of anxiety dis- emotional homeostasis. These include monoamin-
orders (Rauch etal., 2003; Bremner, 2004; Damsa ergic transmitters, NE, 5-HT, and dopamine and
etal., 2009). The amygdala is consistently involved the amino acid transmitters, GABA and glutamate.
in studies of social anxiety disorders (SAD), and The neurotransmitter systems that have been best
increased activity in limbic and paralimbic regions studied in relation to stress responsiveness involve
have also been described in SAD, although much the HPA axis and the central NE system. Additional
remains to be investigated within the complexity neurochemical systems participate in the control of
of these repetitive disorders (Freitas-Ferrari etal., stress and emotional responses, including neuro-
2010). Functional neuroimaging studies have peptide Y (NPY), substance P, and opioid and can-
recurrently demonstrated amygdalar hyperacti- nabinoid (CB) systems, among others. Preclinical
vation in posttraumatic stress disorder (PTSD; and clinical studies supporting a role for some of
Brohawn et al., 2010; Vermetten et al., 2007). In these neurochemical agents in emotional homeo-
contrast, hypoactivity of the mPFC, the anterior stasis are reviewed in the next section, as well as
cingulated cortex, and sometimes the hippocam- their potential application as pharmacological
pus, have been identified in a variety of anxi- therapies in the management of anxiety disor-
ety disorders, particularly PTSD and SAD (Shin ders. Overall, the fine-tuning of these circuitries
et al., 2006; Freitas-Ferrari et al., 2010; Hayes under basal conditions, together with their coor-
etal., 2011). Altogether, symptoms of anxiety are dinated response under adverse or stressful
538 Part IV:Homeostatic Therapies

stimuli, guarantees an adequate emotional adap- of cognitive impairments, including basic defi-
tation to the changing environment. However, cits in attention, extinction processes, concen-
abnormalities in the neuroanatomic circuits that tration, and memory (Isaac etal., 2006; Parsons
support emotional regulation and stress respon- and Ressler, 2013). In Europe, a recent report has
siveness and/or in the diversity of chemical neu- indicated that the most frequent mental disor-
rotransmitter systems that control the emotional der is anxiety disorders (14%), with an estimated
homeostasis may be associated with the appear- number of 61.5million of persons affected across
ance of anxiety disorders. Disproportional anxi- all ages (Wittchen etal.,2011).
ety responses, both in intensity or duration, or The fourth edition of the Diagnostic
the arousal of anxiety responses in the absence of and Statistical Manual of Mental Disorders
relevant risksthat is, a dysfunction of emotional (American Psychiatric Association, 1994)groups
homeostasisconstitutes a maladaptive response disorders into diagnostic classes on the basis
to the environment and possibly signal the appear- of the subjective criterion of shared phe-
ance of a psychiatric disorder (i.e., anxiety and/or nomenological features and includes GAD,
stress-related disorders). obsessive-compulsive disorder (OCD), panic
disorder, phobias (including social phobia),
DYSFUNCTIONS OF and PTSD under the category of anxiety disor-
E M O T I O N A L H O M E O S TA S I S : ders. More recently, the American Psychiatric
ANXIETY AND STRESS Association (2013) approved a new edition of
DISORDERS the Diagnostic and Statistical Manual of Mental
Recent surveys demonstrate that anxiety and Disorders, which incorporates significant scien-
stress disorders are the most prevalent of tific advances in more precisely identifying and
psychiatric diseases and have high personal and diagnosing mental disorders. Regarding the area
societal costs (Wittchen et al., 2011). Despite of anxiety, refinements of criteria and symp-
being common, the severity of these disorders can toms across the lifespan constitute the most
vary considerably, from mild worries to incapaci- relevant change. Some disorders included in the
tating symptoms that interfere with functioning broad category of anxiety disorders are now in
(Bernstein and Shaw, 1997). Actually, anxiety three sequential chapters: Anxiety Disorders
disorders categorize a large number of disorders (including specific phobia, social phobia, panic
in which the primary feature is abnormal or inap- disorder and panic attack, GAD, substance/
propriate anxiety. As previously stated, anxiety is medication-induced anxiety disorder, anxiety
an adaptive component of the reactive response to disorder due to another medical condition, etc.),
stress and fear that integrates the flight or fight Obsessive-Compulsive and Related Disorders,
phenomenon, thus preparing the body to either and Trauma- and Stressor-Related Disorders.
fight and protect itself or to flee from a dangerous
situation. However, in the absence of any recog- R E L E VA N C E O F
nizable stimulus or when the stimulus does not T R A N S L AT I O N A L S T U D I E S
warrant such a reaction, these anxiety symptoms FROM ANIMAL MODELS
become pathological, and an anxiety disorder Animal models are of great relevance for under-
might be diagnosed. Indeed, anxiety disorders are standing anxiety and stress disorders (see Hohoff,
marked by excessive fear (and avoidance), often 2009; Cryan and Sweeney, 2011; Neumann etal.,
in response to specific objects or situations and 2011, for more focused reviews on the subject).
in the absence of true danger. Anxiety disorders, Most animal models evaluate unconditioned or
such as panic disorder, social and specific phobia, spontaneous behaviors, involve simple or com-
generalized anxiety disorder (GAD), and PTSD, plex classical and/or operant conditioning pro-
represent a highly prevalent and strongly disa- cedures, or use brain-stimulation paradigms.
bling class of psychiatric disorders (Bekker and General locomotion and exploration in a novel
van Mens-Verhulst, 2007; Shin and Liberzon, environmental context is commonly used to
2010). Notably, cognitive impairments are fre- evaluate unconditioned or spontaneous behav-
quently observed in patients with anxiety disor- ior in animals. Exploratory activity appears to
ders (Gualtieri and Morgan, 2008). More precisely, be a function of general activity level; impulsiv-
PTSD that develops after prolonged inescapable ity or the need to become familiar with a strange
stress experience of exceptional severity (Rubin territory opposes with anxiety or the desire to
et al., 2008) has been associated with a number escape from the unknown and unpredictable.
Anxiety and Stress Disorders 539

As examples of this class, the open field and female rats and the HPA axis negative feedback
hole-board tests are common, as is the social greater (e.g., Louvart et al., 2006; Mazor et al.,
interaction test (File and Hyde, 1979; File and 2009). Notwithstanding, gonadal steroid hor-
Seth, 2003)or the hyponeophagia (Shephard and mones may play a critical role in mediating the
Broadhurst, 1983). Animal models based on con- sex differences in the response to stress as well as
ditioned behaviors typically consist of an operant in anxiety disorders, since sex differences involve
response cued by different light signals. In this differences in gonadal steroid hormone secretion
class we can use the Geller-Seifter conflict test (i.e., estrogen and androgen) and causes differ-
(Sepinwall et al., 1978) or the Vogel water-lick ent effects on neuronal functioning (e.g., Patchev
water test (Vogel et al., 1971). Conditioned pro- and Almeida, 1998; McCormick etal., 2002; Ter
cedures are often subjected to the criticism of Horst etal.,2009).
including food or water deprivation, and, in turn, Targeted mutations leading to anxiety-like
motivation and reward cannot be excluded of the responses in transgenic mice have suggested
discussion of results (see File etal., 2005; Griebel roles for serotonin receptor subtype 1A (5-HT1A),
and Holmes, 2013, for review). The delineation of CRH, GABA, neuropeptide Y, cholecystokinin
specific neural pathways mediating conditioned (CCK), and substance P neural systems in the
and unconditioned fear can logically guide the control of emotional states. A huge volume of
design and conduct of clinical studies investigat- potential anxiolytic drugs has been generated in
ing the neurobiological mechanisms of anxiety animal models; however, the clinical outcome
disorders such as GAD (unconditional fear) and of these investigations has been disappointing,
phobic disorders (conditionedfear). as promising results with novel agents in rodent
Most paradigms claim at least face validity, studies have very rarely translated into effec-
as they are fear inducing, and/or a predictive tiveness in humans (Griebel and Holmes, 2013).
validity in that they are relatively selective for We briefly describe the participation of some of
clinically used anxiolytics. Notwithstanding, the these neurochemical systems in emotionality and
validity of animal models of anxiety has usually anxiety disorders (see, e.g., Nutt, 2001; Charney and
been restricted, implicitly or explicitly, to the Drevets, 2002; Cryan and Sweeney, 2011; Griebel
success or failure of a given model in predicting and Holmes, 2013, for more extensive reviews), and
the clinical anxiolytic potency of pharmacologi- their potential as anxiolytic drugs in the clinic is
cal agents (predictive validity); however, screen- discussed in the corresponding section.
ing may not be the only function of these animal
models. Animal models also provide important GABA and Benzodiazepine Systems
contributions to revealing the neurobiologi- Clinical and preclinical data have established
cal mechanisms of anxiety disorders, as well as that benzodiazepine (BDZ)-receptor agonists
important information on vulnerability factors, exert anxiolytic effects, and the functionality
age-dependent differences, and sexual dimor- of BDZ receptors may be altered in anxiety dis-
phisms, among others. Indeed, animal models orders (Nutt and Malizia, 2001). The BDZ and
are of great value for the investigation of genetic GABA A receptors are part of the same macromo-
and/or environmental factors that may predis- lecular complex; these receptors present different
pose for the development of anxiety disorders. In binding sites that are functionally coupled and
particular, rodent and nonhuman primate stud- regulated in an allosteric manner. Central BDZ
ies of motherinfant interactions are particularly receptors are extensively expressed throughout
compelling, given the important clinical impli- the brain but are most densely concentrated in the
cations of the early life environment. Sex dif- cortical grey matter (Choi etal., 1981). Responses
ferences have also been extensively studied, and similar to those seen in anxiety (i.e., increases in
literature on animal studies has largely docu- heart rate, blood pressure, plasma cortisol, and
mented male versus female differences in behav- chatecolamines) were observed in rodents after
ior, neurobiological parameters, and response to the administration of BDZ-receptor inverse ago-
stress (Toufexis etal., 2006; Simpson and Kelly, nists, such as -carboline-3-carboxylic acid ethy-
2012; Simpson etal., 2012). In general, female ani- lester (Braestrup etal., 1982; Dorow etal., 1983).
mals appear to be more vulnerable than males to Actually, classical BDZs have long been reported
both acute and chronic stress exposure. Actually, to exert anxiolytic-like effects by increasing the
the elevation in plasma corticosterone levels in affinity of GABA A receptors for the neurotrans-
response to stress is higher and more persistent in mitter GABA. Along the same lines, transgenic
540 Part IV:Homeostatic Therapies

mice studies further supported the role of BDZ who developed PTSD. In particular, polymor-
and GABA A receptors in emotional homeosta- phisms at FKBP5, a modulator of glucocorticoid
sis and enabled the understanding of the spe- receptor sensitivity, may influence circulating
cific role of each GABA A-receptor subunit. The cortisol levels and PTSD severity (Yehuda, 2006;
GABA A-receptor 2 subunit seem to mediate, at Sarapas etal.,2011).
least partially, the anxiolytic effect of BDZ ago-
nists, since the anxiolytic action of diazepam Norepinephrine
disappeared in mice lacking the 2 subunit but NeurotransmissionSystem
remained in mice lacking 1 and 3 subunits Abnormalities in the NE system have long been
(Low etal., 2000; McKernan etal., 2000). In con- implicated in the pathophysiology of anxiety dis-
trast, GABA A receptor 1 may mediate the seda- orders (Ressler and Nemeroff, 2000). Exposure to
tive, amnesic, and anticonvulsant effects of BDZs stressful stimuli increases central NE function;
(Rudolph et al., 1999; McKernan et al., 2000). stress exposure increases NE turnover in the
These findings have important implications locus coeruleus, hypothalamus, hippocampus,
for the investigation of the pathophysiology of amygdala, and cerebral cortex (Cassens et al.,
anxiety and also for the development of 1981). The firing rate of locus coeruleus neurons
BDZ-receptor agonists selective for their anxio- also increases during exposure to stress and other
lytic properties and devoid of adverse side effects fear-conditioning stimuli (Rasmussen et al.,
such as sedation or amnesia (for a review see 1986; Abercrombie and Jacobs, 1987). Actually,
Rudolph and Knoflach,2011). NE release seems to play a critical role in fear
learning since the acquisition of fear-conditioned
Serotonin NeurotransmissionSystem responses requires an intact central NE system
The 5-HT system has also been shown to play (Rasmussen etal., 1986; Abercrombie and Jacobs,
a role in fear and anxiety disorders in animal 1987; Bremner etal., 1996; Charney and Deutch,
models and in humans (Ressler and Nemeroff, 1996). Responsiveness of locus coeruleus neu-
2000; Garvey et al., 1995; Iny et al., 1994). rons to future novel stressors can be enhanced by
Multiple studies in animals indicate that stress chronic exposure to some stressful experiences.
exposure increases 5-HT turnover in the mPFC, Therefore, the NE system seems to participate in
nucleus accumbens, amygdala, and lateral hypo- the process by which single or repeated exposure
thalamus (Inoue etal., 1994). The effect of stress to aversive stimuli or pharmacological agents can
in activating 5-HT turnover may stimulate both increase the behavioral sensitivity to subsequent
anxiogenic and anxiolytic pathways within the stressors, the so-called behavioral sensitization.
forebrain, depending on the region involved and This effect may result from a stress-mediated
the 5-HT receptor subtype that is predominantly alteration in the sensitivity of presynaptic 2-NE
stimulated. Graeff etal. (1993) proposed that the synthesis and release.
serotoninergic innervation of the amygdala and
the hippocampus mediate anxiogenic responses Cholecystokinin
by 5-HT2A receptor stimulation, whereas sero- CCK was originally isolated in the gastrointesti-
toninergic innervation of hippocampal 5-HT1A nal system but is found extensively throughout
receptors may mediate responses to aversive the nervous system, with particularly high con-
events. In accordance, 5-HT1A receptor knockout centrations distributed throughout the limbic
mice show behaviors consistent with heightened system. CCK is synthesized as a 115 amino acid
anxiety (Ramboz etal.,1998). preprohormone and is converted into multi-
ple isoforms: the sulphated CCK-8 is predomi-
CRH and Glucocorticoid nant in the central nervous system, and CCK-5
In rodents, CRF1 antagonists generally do not (pentagastrin), and CCK-4 are also relevant. In
show anxiolytic effects under basal conditions, general, CCK binds to two receptors, CCK1 and
but rather they block the heightened anxiety CCK2CCK is one of the neuropeptides impli-
produced by such factors as stress or selective cated in anxiety both in animal models and
breeding for high anxiety (Rotzinger etal., 2010). humans (Woodruff etal., 1991; Harro etal., 1993;
In humans, by examining a population-based Rotzinger et al., 2010; Bowers et al., 2012). CCK
cohort of individuals exposed to the World Trade agonists show anxiogenic effects in a number of
Center collapse, genetic markers related to gluco- animal models; indeed, CCK administration is
corticoid signaling were identified in individuals frequently employed to induce panic attacks in
Anxiety and Stress Disorders 541

animal models (Rotzinger and Vaccarino, 2003). response through distributed actions in limbic
However, the effects of CCK antagonists often and hypothalamic circuits in the brain (Riebe
are not seen unless the system is potentiated. and Wotjak, 2011; Hill and Tasker, 2012) and is
The CCK2 receptor appears to play a key role in involved in the extinction of emotionally aver-
anxiety, but it remains to be seen whether effica- sive memories (Marsicano etal., 2002; Plendl and
cious CCK antagonists can treat anxiety disorders Wotjak, 2010; Gunduz-Cinar et al., 2013) and
or depression. Clinical trials in anxiety disorders habituation and adaptation to stress (Patel et al.,
and CCK antagonists have not been promising. 2005; Hill etal., 2010), processes that are dysregu-
The CCK2 antagonist CI-988 had no effect in lated in PTSD. Taken together, it may seem that
patients with GAD (Adams et al., 1995; Pande reduced AEA signaling could be associated with
etal., 1999), nor did it block the panic-inducing impaired extinction of aversive memories and an
effects of CCK-4 (van Megen et al., 1997). The increase in intrusive symptoms. Regarding the
CCK2 antagonist L-365,260 had no effect in contribution of eCB ligands on emotion, a differ-
patients with panic disorder (Kramer etal.,1995). ential, and maybe opposite, role has been proposed
for AEA and 2-AG. However, much research is
NeuropeptideY needed to clarify the complex role of each com-
NPY is a 36-amino acid peptide that is widely ponent of the eCB in emotional homeostasis and
distributed in the central nervous system. There anxiety disorders.
are currently five identified NPY receptors,
which are also widely distributed in the central PRESENT AND FUTURE
nervous system, particularly throughout the H O M E O S TAT I C T H E R A P I E S
frontal cortex and limbic regions (Redrobe etal., FORANXIETY AND STRESS
2002). NPY has been implicated in anxiety dis- DISORDERS
orders (see Rotzinger et al., 2010, for a review). Benzodiazepines and agents acting on the sero-
In animal models, NPY administration has been tonergic system are currently the main drugs
reported to induce anxiolytic-like effects in vari- employed in the management of anxiety dis-
ety of behavioral models. In contrast, the admin- orders (Sandford et al., 2000; Millan, 2003).
istration of NPY-receptor antagonists enhances However, in recent years a search for new drugs
anxiety, thus providing strong evidence for beyond classical treatments have focused most
NPYs role in modulating anxiety responses. In intensively on the 5-HT, neuropeptide, gluta-
humans, plasma NPY levels have been proposed mate, and eeCB systems (Griebel and Holmes,
to represent a biologic correlate of resilience to or 2013). Selective 5-HT reuptake inhibitors (SSRIs;
recovery from the adverse effects of stress; a bio- e.g., citalopram, paroxetine, fluvoxavine, fluox-
marker of great relevance for the clinical study etine, etc.) and serotonin-norepinephrine reup-
of PTSD (Cohen etal., 2012; Yehuda etal., 2006; take inhibitors (e.g., venlafaxine, duloxetine,
Sajdyk etal.,2008). etc.) are useful first-line agents for most anxiety
disorders, particularly given the frequent comor-
The EndocannabinoidSystem bidity with mood disorders. They both present
The endocannabinoid (eCB) system consists of an extremely broad spectrum of action in anxi-
two CB receptors, one mainly central (CB1 recep- ety disorders. However, their use is limited by a
tor) and a second one (CB2 receptor) tradition- delayed onset and incomplete response in many
ally considered as peripheral although it has been patients. Research on the neurobiological under-
recently described in several brain regions. Two pinnings of anxiety disorders has continued for
principle endogenous ligands have been discov- several decades, and the search for novel phar-
ered: N-arachidonoylethanolamine (AEA) and macological treatments is driven by the growing
2-arachidonoylglyceril (2-AG), together with medical need to improve on the effectiveness
the enzymes involved in their metabolism (see and the side-effect profile of existing drugs (see
Maccarrone, 2010). Over the past few decades, an Ravindran and Stein, 2010; Reinhold etal., 2011;
extent literature on the eCB system has emerged to Griebel and Holmes, 2013, for a more complete
give support to the central role played by the eCB update on anxiety disorders pharmacotherapy).
system in emotional control (Viveros etal., 2005; In general, benzodiazepines are still use-
Moreira and Lutz, 2008; Moreira and Wotjak, ful in the management of panic disorder and
2010; Marco and Laviola, 2012). The eCB system GAD. Serotonergic agents are preferred for the
is known to constrain activation of the stress treatment of OCD, while other antidepressants,
542 Part IV:Homeostatic Therapies

such as tricyclic antidepressants or monoam- signaling to PTSD or will help to elucidate more
ine oxidase inhibitors, are generally reserved as precisely the respective functional roles of 2-AG
second- and third-line strategies due to toler- and AEA in PTSD development and expression.
ability issues. Antidepressant drugs, primarily Despite further studies being needed; prelimi-
acting on monoamine reuptake, are effective for nary data indicate that CB agonists may be effec-
the treatment of a spectrum of anxiety disorders tive in the clinical treatment of PTSD. At present,
including SAD, GAD, panic disorder, and PTSD. a first clinical trial (phase IV) on the efficacy of
Notwithstanding, the anxiolytic effects of antide- delta-9-tetrahydrocannabinol treatment for the
pressant drugs may result from interactions with management of PTSD is being conducted; adult
the GABAergic system. As an example, phenelzine subjects of both genders are being recruited,
induces an anxiolytic-like effect in the rat elevated and first results will be available soon (http://
plus maze, possibly through the elevation of GABA ClinicalTrials.gov/show/NCT00965809).
concentration (Paslawski etal., 1996). Azapirones, In spite of the fact that cannabidiol CBD
including buspirone, are partial 5-HT agonists may not be considered a component of the eCB
with low abuse potential that have been effectively system, notable anti-anxiety properties have
used for GAD. In case of treatment resistance, been extensively reported for this non-psycho-
anticonvulsants and atypical antipsychotics are active phytocannabinoid present in the plant
employed as an adjuvant in the treatment with of Cannabis sativa (Guimares et al., 1990;
antidepressants (Ravindran and Stein,2010). Moreira etal., 2006). CBD not only prevents the
As mentioned, GABA has been recently anxiogenic effects of high doses of tetrahy-
involved in extinction; thus augmenting drocannabinol (Zuardi et al., 1982) but also
extinction-based psychotherapy with GABA- decreases anxiety symptoms in healthy subjects
agonist drug treatment may be a potential submitted to a simulated public-speaking para-
strategy to investigate. In regard to benzodiaz- digm (Zuardi etal., 1993). More recently, using
epine drug development, research has focused a similar paradigm, Bergamaschi et al. (2011)
on the development of selective agonists with showed that CBD reduced public-speaking
increased responsiveness at GABA A-receptor anxiety in treatment-nave social phobic
2-subunit but limited actions to the 1 and 3 patients and successfully decreased subjective
subunits (see Rudolph and Knoflach, 2011). The anxiety in SAD patients (Crippa et al., 2011).
development of the CRH antagonist has also Accordingly, neuroimaging studies have shown
significantly influenced the pharmacotherapy that CBD impairs connectivity between the
of anxiety disorders. Indeed, there is potential prefrontal and subcortical regions (Fusar-Poli
for developing targets for the CRF-2 receptor et al., 2010), attenuates blood oxygenation
and other peptides, such as vasoactive intesti- leveldependent responses to fearful faces in
nal peptide, involved in the regulation of stress the amygdala and cingulate cortex (Fusar-Poli
(Ravindran and Stein,2010). et al., 2009), and decreases activation in the
Furthermore, enhancement of eCB signal- left amygdala-hippocampal complex and left
ing has achieved promising anxiolytic effects in posterior cingulate gyrus (Crippa et al., 2004).
a variety of animal studies, thus emerging as an Although the pharmacological mechanisms
attractive strategy for potential therapeutic appli- involved in CBD effects are still poorly under-
cation (Pacher etal., 2006; Piomelli, 2006; Marco stood (Campos etal., 2012), one of the primary
et al., 2011). In recent years, promising results mechanisms may involve fatty acid amide
have been achieved in the field of the eCB sys- hydrolase inhibition and thus facilitation of
tem as a potential clinical target in the treatment AEA signaling (Bitencourt etal.,2008).
of the PTSD. Actually, drugs acting on the eCB At present, pharmacological therapies
system have been proposed to enhance the effec- are usually combined with concomitant psy-
tiveness of exposure therapy (see Passie et al., chotherapeutic interventions. Indeed, phar-
2012; de Bitencourt et al., 2013; Rabinak and macotherapy should be used as an adjunct to
Phan, 2014; Trezza and Campolongo, 2013, for behavioral or psychotherapeutic interventions.
reviews). Trials examining the effects of agents Among psychotherapeutic interventions, cogni-
that potentiate eCB signaling in PTSD, either tive behavioral therapy, particularly exposure
alone or in conjunction with treatments that therapy, has been shown to be highly effective
modulate glucocorticoids signaling, will provide in the treatment of anxiety disorders (Goossens
proof of principal for the contribution of eCB et al.,2007). Exposure therapy consists of the
Anxiety and Stress Disorders 543

systematic and repeated exposure of patients more likely to experience (i.e., higher rates of
to the anxiety-provoking stimulus or situation severe intimate-partner violence and sexual
until their fear subsides. In this regard, changes assault across the lifespan); the profoundly cor-
in brain structure and function have been dem- rosive and damaging meanings inherent in these
onstrated following successful treatment with kinds of violations; the possibility that women
exposure therapy. Actually, altered patterns in may be prone to heightened levels of emotional
neural functioning were observed after success- reactivity, which could confer additional risk for
ful exposure therapy in a sample of individuals PTSD; and the potential that gender differences in
with spider phobia (an example of specific pho- behavioral and psychological sequelae to trauma
bia); a decrease in the activation of the amygdala influence diagnostic presentation (e.g., internal-
was observed, together with a normalization of izing vs. externalizing symptoms; Pratchett etal.,
insular and anterior cingulated cortex activity 2010). Neuroanatomical sex differences have been
(Goossens et al., 2007). Similarly, recovery from reported in children with maltreatment-related
PTSD was correlated with the activity in the hip- PTSD; boys exhibited larger prefrontal lobe
pocampus and subgenual anterior cingulated cor- cerebrospinal fluid volumes and smaller sple-
tex (Dickie etal.,2011). nium, smaller cerebral volumes and corpus cal-
Furthermore, advances in deep brain stimu- losum, and greater lateral ventricular volume
lation have opened new avenues for the applica- increases than girls. An association between
tion of this novel strategy in the management enhanced brainstem activity and PTSD diagnosis
of anxiety disorders. Depression and OCD have has been described for men but not for women,
benefited from deep brain stimulation since 2005, and the hippocampal fear response was greater
when the US Food and Drug Administration in men compared to their female counterparts
approved vagal nerve stimulation for the man- (De Bellis and Keshavan, 2003). Epinephrine and
agement of treatment-resistant depression; norepinephrine has been associated with PTSD
in 2009 it was approved for refractory OCD symptoms following trauma exposure in men
(Heeramun-Aubeeluck and Lu, 2013). More but not in women (Hawk etal., 2000). Moreover,
recently, a double-blind study demonstrated a sig- sex differences in corticotropin-releasing fac-
nificant improvement of PTSD patients resistant tor (CRF) receptor functionality, which is more
to standard treatment following repeated medial sensitive to low CRF levels but less adaptable
prefrontal cortex deep transcranial magnetic to high levels in women, may possibly increase
stimulation (a wide multicenter study is suggested the risk for PTSD development among women
to substantiate these findings; trial registra- (Bangasser et al., 2010). In addition, sex dif-
tion:ClinicalTrials.gov identifier:NCT00517400; ferences in cortisol and catecholamines levels
Isserles et al., 2013), while striatal deep brain among subjects with PTSD may also be of criti-
stimulation seems to reduce conditioned fear cal relevance. Among PTSD subjects, womens
and enhance extinction memory in rodents salivary cortisol levels decreased during the day
(Rodriguez-Romaguera etal.,2012). while the opposite occurred in men (i.e., a daily
increase in cortisol levels; Freidenberg et al.,
SEX DIFFERENCES IN 2010; Viveros etal.,2012).
PHYSIOLOGICAL AND
PAT H O L O G I C A L E M O T I O N A L ANXIETY DISORDERS
H O M E O S TA S I S DURING THELIFETIME
In general, females are affected approximately Anxiety, as already described, presents as auto-
two to three times more frequently than males nomic hyperactivity, with raised blood pres-
by anxiety disorders (Wittchen et al., 2011). sure and heart frequency, increased sweating,
Accordingly, epidemiological studies report and dryness of mouth and throat, along with
higher rates of PTSD among women compared cognitive and affective states of increased vigi-
to men (Breslau and Anthony, 2007); in addi- lance, distractibility, and apprehension. Motor
tion, symptoms appear to be more severe and fatigue and insomnia are common. In children,
quality-of-life outcomes are significantly worse anxiety disorders include separation anxiety,
in women (Holbrook etal., 2002; Tolin and Foa, avoidance disorders (of strangers), and over-
2006). Sexual differences in PTSD vulnerability anxious disorderin sum, a general apprehen-
may rely on the intimate, interpersonal nature sion of the world. Anxiety disorders appear as
of potentially traumatic events that women are early as in childhood, critically affecting further
544 Part IV:Homeostatic Therapies

neurocognitive development and thus interfering of GAD (Rynn etal., 2001)and SAD (Compton
with school and academic achievement, social etal., 2001)in children and adolescents. In all
functioning, and social integration (Drysdale of these clinical studies, adverse side effects
et al., 2014; Kessler et al., 2005). Anxiety disor- were minimal. In addition, the efficacy of the
ders might persist throughout the lifespan or at SSRI treatment in a pediatric population of
least have an enduring impact. Recent data indi- OCD patients was long-lasting with only mild
cate that GAD and PTSD incidence significantly side effects that tended to disappear with time,
differ by age group; for GAD, evidence suggests mainly nausea, headache, insomnia, abdominal
highest rates (3.4%) among the elderly (65+) and pain, hyperkinesia, nervousness, and vomiting
considerably lower rates (1.7%) in the 14 to 65 age (Cook etal., 2001; Thomsen etal., 2001). Taken
group, while PTSD rates seem to decline by age together, the treatment of anxiety disorders
(1434: 2.9%, 3565: 1.3%, 65+:1.1%; Wittchen in children and adolescents should generally
etal.,2011). involve a multimodal approach that is a com-
bination of pharmacological intervention with
Anxiety Disorders inChildren cognitive behavioral therapy. However, the
and Adolescents clinician must always weight whether the con-
Anxiety disorders are among the most com- dition of the child is sufficiently serious to war-
mon pediatric psychopathologies. The diagnosis rant pharmacological intervention (Velosa and
of anxiety disorders peaks during adolescence Riddle, 2000; Kratochvil et al., 2002; Riddle
(Drysdale et al., 2014; Kessler et al., 2005), and etal.,2013).
the presence of an anxiety disorder in childhood
or adolescence seems to predict its persistence Anxiety Disorders inthe Elderly
into adulthood, as well as the appearance of other An increasing body of evidence suggests that
psychiatric disorders, particularly depression medication and psychotherapy for anxiety dis-
(Stein and Lang,2002). orders in older adults is not as effective as in
The pharmacological intervention of anxiety younger people. Indeed, only a minority of
disorders in children and adolescents was initially old-age patients receives appropriate medica-
based on successful strategies applied in adults. tion. In spite of the fact that anxiety disorders
However, increasing concern about the efficacy are common in later life, the number of clini-
and adequacy of these therapies rapidly rose, and cal trials in the old population is scarce (see
clinical trials in youth have been performed in Voshaar, 2013; Wetherell et al., 2013, for recent
the past few years also considering the appear- updates). Actually, little is known about the
ance of adverse side effects and complications natural course, trajectories, and determinants
(see Kratochvil etal., 2002, for a review). Some of of anxiety disorders in old age, or regarding the
the most relevant studies in the youth are briefly specific needs for interventions in this age group
described here, despite the fact that the psychop- (Riedel-Heller etal., 2006). Therefore, the design
harmacological management of pediatric anxiety of appropriate clinical trials in the elderly is of
disorders is out of the scope of this chapter and critical urgency as well as the development of
can be investigated elsewhere (e.g., Peters and age-specific psychological and pharmacological
Connolly, 2012; Riddle etal.,2013). therapies.
When pharmacotherapy is indicated, the Recently, a European study on mental
SSRIs appear to be particularly suitable for health (Wittchen et al., 2011) highlighted
the treatment of anxiety disorders in children the urgency for incidence longitudinal stud-
and adolescents given their safety and effi- ies especially across defined age cohorts (i.e.,
cacy. Fluvoxamine has arisen as an effective in children and adolescents, adults, and older
therapy for children and adolescents diag- adults) in order to better understand the onset
nosed with social phobia, separation anxiety and natural course of anxiety disorders as well
disorder, or GAD (Research Unit on Pediatric as the identification of critical trajectories.
Psychopharmacology, 2001) as well as for Such data would provide better guidance for
the management of OCD in youth (Riddle preventive trials and targeted early interven-
et al., 2001). Similarly, fluoxetine successfully tions, and they may suggest important cues for
reduced anxiety scores in pediatric patients. preventing the development of co- and multi-
Sertraline was also effective in the management morbidity of anxiety disorders.
Anxiety and Stress Disorders 545

COMORBIDITIES OFANXIETY Greiner, M., Pierce, M. W., Pande, A. C. 1995.


DISORDERS A double-blind, placebo-controlled study of a
It has long been known that anxiety symptoms CCK-B receptor antagonist, CI-988, in patients
and comorbid anxiety are common in depres- with generalized anxiety disorder. J Clin
sive syndromes (Fawcett, 1997; Merikangas etal., Psychopharmacol 15, 428434.
2003; Kessler et al., 2005). Comorbid anxiety American Psychiatric Association. 1994. Diagnostic
disorders have also been associated with a more and Statistical Manual of Mental Disorders. 4th
frequent history of substance abuse and higher ed. (Washington, DC:Author).
ratings for suicidal ideation, and high rates of American Psychiatric Association. 2013. Diagnostic
anxiety disorders have been reported in bipolar and Statistical Manual of Mental Disorders. 5th
disorders (Lee and Dunner, 2008). In both cases, ed. (Washington, DC:Author).
the presence/severity of anxiety symptoms nega- Bangasser, D.A., Curtis, A., Reyes, B.A., Bethea, T.T.,
Parastatidis, I., Ischiropoulos, H., Van Bockstaele,
tively impact the clinical management of the psy-
E. J., Valentino, R. J. 2010. Sex differences in
chiatric disorders of major depression (Fawcett
corticotropin-releasing factor receptor signaling
and Kravitz, 1983)and bipolar disorder (Lee and
and trafficking: Potential role in female vulner-
Dunner, 2008). Given the high prevalence and
ability to stress-related psychopathology. Mol
risk rate of comorbid anxiety disorders in depres-
Psychiatry 15, 877, 896904.
sive and bipolar patients, it is of great relevance Baxter, M.G., Parker, A., Lindner, C.C., Izquierdo,
to evaluate such symptoms in clinical practice, A. D., Murray, E. A. 2000. Control of response
since the presence of comorbidity may eventu- selection by reinforcer value requires interac-
ally serve as a guide to choosing the appropriate tion of amygdala and orbital prefrontal cortex. J
pharmacological therapy. Neurosci 20, 43114319.
Bekker, M.H., van Mens-Verhulst, J. 2007. Anxiety
CONCLUSIONS AND disorders:Sex differences in prevalence, degree,
FUTURE REMARKS and background, but gender-neutral treatment.
At present, despite that several drugs, mainly Gend Med 4 Suppl B, S178S193.
benzodiazepines and serotonergic agents, are Bergamaschi, M.M., Queiroz, R.H., Chagas, M.H.,
available and effective in the management of de Oliveira, D.C., De Martinis, B.S., Kapczinski,
anxiety disorders, the understanding of the F., Quevedo, J., Roesler, R., Schroder, N., Nardi
neural and chemical basis of these disorders A. E., et al. 2011. Cannabidiol reduces the
has enabled the discovery of novel drugs for anxiety induced by simulated public speak-
potential use in the clinic. In a recent European ing in treatment-naive social phobia patients.
study on mental health (Wittchen et al., 2011), Neuropsychopharmacology 36, 12191226.
the authors highlight that early intervention Bernstein, G.A., Shaw, K. 1997. Practice parameters
is critical for an improvement in the primary for the assessment and treatment of children and
diagnosed disorder but may also be particularly adolescents with anxiety disorders. J Am Acad
important for preventing the development of co- Child Adolesc Psychiatry 36, 69S84S.
and multimorbidity since the early treatment Bitencourt, R.M., Pamplona, F.A., Takahashi, R.N.
of a temporally primary disorder may prevent 2008. Facilitation of contextual fear mem-
secondary comorbidity. Moreover, clini- ory extinction and anti-anxiogenic effects of
cians may control for the presence of poten- AM404 and cannabidiol in conditioned rats. Eur
tial comorbidities in the selection of a therapy. Neuropsychopharmacol 18, 849859.
Longitudinal studies along the lifespan are Bowers, M. E., Choi, D. C., Ressler, K. J. 2012.
critical, since age-specific studies should be Neuropeptide regulation of fear and anxiety:
accomplished for the accuracy in the selection Implications of cholecystokinin, endogenous
of appropriate therapies. opioids, and neuropeptide Y. Physiol Behav 107,
699710.
References Braestrup, C., Schmiechen, R., Neef, G., Nielsen, M.,
Abercrombie, E. D., Jacobs, B. L. 1987. Single-unit Petersen, E. N. 1982. Interaction of convulsive
response of noradrenergic neurons in the locus ligands with benzodiazepine receptors. Science
coeruleus of freely moving cats: I. Acutely pre- 216, 12411243.
sented stressful and nonstressful stimuli. J Bremner, J.D. 2004. Brain imaging in anxiety disor-
Neurosci 7, 28372843. ders. Expert Rev Neurother 4, 275284.
Adams, J. B., Pyke, R. E., Costa, J., Cutler, N. R., Bremner, J. D., Krystal, J. H., Southwick, S. M.,
Schweizer, E., Wilcox, C. S., Wisselink, P. G., Charney, D.S. 1996. Noradrenergic mechanisms
546 Part IV:Homeostatic Therapies

in stress and anxiety:II. Clinical studies. Synapse P. K., Filho Busatto, G. 2004. Effects of canna-
23,3951. bidiol (CBD) on regional cerebral blood flow.
Breslau, N., Anthony, J.C. 2007. Gender differences Neuropsychopharmacology 29, 417426.
in the sensitivity to posttraumatic stress disor- Cryan, J. F., Sweeney, F. F. 2011. The age of anxi-
der: An epidemiological study of urban young ety:Role of animal models of anxiolytic action in
adults. J Abnorm Psychol 116, 607611. drug discovery. Br J Pharmacol 164, 11291161.
Brohawn, K.H., Offringa, R., Pfaff, D.L ., Hughes, K.C., Charney, D.S., Deutch, A. 1996. A functional neuro-
Shin, L.M. 2010a. The neural correlates of emo- anatomy of anxiety and fear:Implications for the
tional memory in posttraumatic stress disorder. pathophysiology and treatment of anxiety disor-
Biol Psychiatry 68, 10231030. ders. Crit Rev Neurobiol 10, 419446.
Brothers, L. 1995. Neurophysiology of the perception Charney, D.S., Drevets, W.C. 2002. Neurobiological
of intentions by primates. In: Gazzaniga, M. S. basis of anxiety disorders. In: Davis, K. L.,
(Ed.), The Cognitive Neurosciences (Cambridge, Charney, D. S., Coyle, J. T., Nemeroff, C.
MA:MIT Press), pp. 11071116. (Eds.), Neuropsychopharmacology: The Fifth
Campos, A.C., Moreira, F.A., Gomes, F.V., Del Bel, E.A., Generation Progress (Philadelphia: Lippincott/
Guimaraes, F. S. 2012. Multiple mechanisms Williams & Wilkins), pp. 901930.
involved in the large-spectrum therapeutic Choi, D. W., Farb, D. H., Fischbach, G. D. 1981.
potential of cannabidiol in psychiatric disor- Chlordiazepoxide selectively potentiates GABA
ders. Philos Trans R Soc Lond B Biol Sci 367, conductance of spinal cord and sensory neurons
33643378. in cell culture. J Neurophysiol 45, 621631.
Carr, D. B., Sesack, S. R. 2000. Dopamine termi- Damasio, A.R. 1998. Emotion in the perspective of
nals synapse on callosal projection neurons in an integrated nervous system. Brain Res Brain
the rat prefrontal cortex. J Comp Neurol 425, Res Rev 26,8386.
275283. Damsa, C., Kosel, M., Moussally, J. 2009. Current
Cassens, G., Kuruc, A., Roffman, M., Orsulak, P.J., status of brain imaging in anxiety disorders.
Schildkraut, J.J. 1981. Alterations in brain nor- Curr Opin Psychiatry 22, 96110.
epinephrine metabolism and behavior induced De Bellis, M.D., Keshavan, M.S. 2003. Sex differences
by environmental stimuli previously paired with in brain maturation in maltreatment-related
inescapable shock. Behav Brain Res 2, 387407. pediatric posttraumatic stress disorder. Neurosci
Cohen, H., Liu, T., Kozlovsky, N., Kaplan, Z., Biobehav Rev 27, 103117.
Zohar, J., Mathe, A. A. 2012. The neuropep- de Bitencourt, R. M., Pamplona, F. A., Takahashi,
tide Y (NPY)-ergic system is associated with R.N. 2013. A current overview of cannabinoids
behavioral resilience to stress exposure in an and glucocorticoids in facilitating extinction of
animal model of post-traumatic stress disorder. aversive memories:Potential extinction enhanc-
Neuropsychopharmacology 37, 350363. ers. Neuropharmacology 64, 389395.
Compton, S. N., Grant, P. J., Chrisman, A. K., de Kloet, E. R., Joels, M., Holsboer, F. 2005. Stress
Gammon, P. J., Brown, V. L., March, J. S. 2001. and the brain: From adaptation to disease. Nat
Sertraline in children and adolescents with social Rev Neurosci 6, 463475.
anxiety disorder:An open trial. J Am Acad Child Dickie, E.W., Brunet, A., Akerib, V., Armony, J.L.
Adolesc Psychiatry 40, 564571. 2011. Neural correlates of recovery from
Cook, E.H., Wagner, K.D., March, J.S., Biederman, J., post-traumatic stress disorder: A longitudi-
Landau, P., Wolkow, R., Messig, M. 2001. nal fMRI investigation of memory encoding.
Long-term sertraline treatment of children and Neuropsychologia 49, 17711778.
adolescents with obsessive-compulsive disor- Dorow, R., Horowski, R., Paschelke, G., Amin, M.
der. J Am Acad Child Adolesc Psychiatry 40, 1983. Severe anxiety induced by FG 7142, a
11751181. beta-carboline ligand for benzodiazepine recep-
Crippa, J. A., Derenusson, G. N., Ferrari, T. B., tors. Lancet 2,9899.
Wichert-Ana, L., Duran, F.L., Martin-Santos, R., Drysdale, A. T., Hartley, C. A., Pattwell, S. S.,
Simoes, M.V., Bhattacharyya, S., Fusar-Poli, P., Ruberry, E.J., Somerville, L.H., Compton, S.N.,
Atakan, Z., et al. 2011. Neural basis of anxio- Lee, F.S., Casey, B.J., Walkup, J.T. 2014. Fear and
lytic effects of cannabidiol (CBD) in generalized anxiety from principle to practice:Implications
social anxiety disorder: A preliminary report. J for when to treat youth with anxiety disorders.
Psychopharmacol 25, 121130. Biol Psychiatry 75,1920.
Crippa, J. A., Zuardi, A. W., Garrido, G. E., Fawcett, J. 1997. The detection and consequences of
Wichert-Ana, L., Guarnieri, R., Ferrari, L., anxiety in clinical depression. J Clin Psychiatry
Azevedo-Marques, P.M., Hallak, J.E., McGuire, 58 Suppl 8,3540.
Anxiety and Stress Disorders 547

Fawcett, J., Kravitz, H.M. 1983. Anxiety syndromes in phobic fear normalizes after exposure. Biol
and their relationship to depressive illness. J Clin Psychiatry 62, 11191125.
Psychiatry 44,811. Graeff, F.G., Silveira, M.C., Nogueira, R.L., Audi,
File, S. E., Hyde, J. R. 1979. A test of anxiety that E.A., Oliveira, R.M. 1993. Role of the amygdala
distinguishes between the actions of benzodia and periaqueductal gray in anxiety and panic.
zepines and those of other minor tranquilisers Behav Brain Res 58, 123131.
and of stimulants. Pharmacol Biochem Behav Griebel, G., Holmes, A. 2013. 50years of hurdles and
11,6569. hope in anxiolytic drug discovery. Nat Rev Drug
File, S.E., Lippa, A.S., Beer, B., Lippa, M.T. 2005. Discov 12, 667687.
Animal tests of anxiety. Curr Protoc Pharmacol Gualtieri, C.T., Morgan, D.W. 2008. The frequency
Chapter5, Unit5.38. of cognitive impairment in patients with anxi-
File, S. E., Seth, P. 2003. A review of 25 years of ety, depression, and bipolar disorder: An unac-
the social interaction test. Eur J Pharmacol counted source of variance in clinical trials. J
463,3553. Clin Psychiatry 69, 11221130.
Freidenberg, B. M., Gusmano, R., Hickling, E. J., Guimares, F.S., Chiaretti, T.M., Graeff, F.G., Zuardi,
Blanchard, E. B., Bremner, J. D., Frye, C. 2010. A.W. 1990. Antianxiety effect of cannabidiol in
Women with PTSD have lower basal salivary the elevated plus-maze. Psychopharmacology
cortisol levels later in the day than do men with (Berl) 100, 558559.
PTSD: A preliminary study. Physiol Behav 99, Gunduz-Cinar, O., MacPherson, K. P., Cinar, R.,
234236. Gamble-George, J., Sugden, K., Williams, B.,
Freitas-Ferrari, M. C., Hallak, J. E., Trzesniak, C., Godlewski, G., Ramikie, T. S., Gorka, A. X.,
Filho, A. S., Machado-de-Sousa, J. P., Alapafuja, S. O., et al. 2013. Convergent trans-
Chagas, M. H., Nardi, A. E., Crippa, J. A. lational evidence of a role for anandamide in
2010. Neuroimaging in social anxiety disor- amygdala-mediated fear extinction, threat pro-
der: A systematic review of the literature. Prog cessing and stress-reactivity. Mol Psychiatry 18,
Neuropsychopharmacol Biol Psychiatry 34, 813823.
565580. Hajos, M., Richards, C.D., Szekely, A.D., Sharp, T.
Fusar-Poli, P., Allen, P., Bhattacharyya, S., Crippa, 1998. An electrophysiological and neuroanatom-
J.A., Mechelli, A., Borgwardt, S., Martin-Santos, ical study of the medial prefrontal cortical pro-
R., Seal, M.L., OCarrol, C., Atakan, Z., Zuardi, jection to the midbrain raphe nuclei in the rat.
A.W., McGuire, P. 2010. Modulation of effective Neuroscience 87, 95108.
connectivity during emotional processing by Halgren, E. 1981. The amygdala contribution to
Delta 9-tetrahydrocannabinol and cannabidiol. emotion and memory: Current studies in
Int J Neuropsychopharmacol 13, 421432. humans. In: EBen-Ari, Y. (Ed.), The Amygdala
Fusar-Poli, P., Crippa, J. A., Bhattacharyya, S., Complex (Amsterdam: Elsevier/North Holland
Borgwardt, S. J., Allen, P., Martin-Santos, Biomedical Press), pp. 395408.
R., Seal, M., Surguladze, S. A., OCarrol, C., Harro, J., Vasar, E., Bradwejn, J. 1993. CCK in ani-
Atakan, Z., et al. 2009. Distinct effects of mal and human research on anxiety. Trends
{delta}9-tetrahydrocannabinol and cannabidiol Pharmacol Sci 14, 244249.
on neural activation during emotional process- Hawk, L.W., Dougall, A.L., Ursano, R.J., Baum, A.
ing. Arch Gen Psychiatry 66, 95105. 2000. Urinary catecholamines and cortisol in
Garvey, M.J., Noyes, R.Jr., Woodman, C., Laukes, C. recent-onset posttraumatic stress disorder after
1995. Relationship of generalized anxiety symp- motor vehicle accidents. Psychosom Med 62,
toms to urinary 5-hydroxyindoleacetic acid and 423434.
vanillylmandelic acid. Psychiatry Res. 57,15. Hayes, J.P., LaBar, K.S., McCarthy, G., Selgrade, E.,
Gaykema, R. P., Gaal, G., Traber, J., Hersh, L. B., Nasser, J., Dolcos, F., Morey, R.A. 2011. Reduced
Luiten, P. G. 1991. The basal forebrain cholin- hippocampal and amygdala activity predicts
ergic system: Efferent and afferent connectivity memory distortions for trauma reminders
and long-term effects of lesions. Acta Psychiatr in combat-related PTSD. J Psychiatr Res 45,
Scand Suppl 366,1426. 660669.
Gloor, P., Olivier, A., Quesney, L.F., Andermann, F., Heeramun-Aubeeluck, A., Lu, Z. 2013. Neurosurgery
Horowitz, S. 1982. The role of the limbic system for mental disorders:Areview. Afr J Psychiatry
in experiential phenomena of temporal lobe epi- (Johannesbg) 16, 177181.
lepsy. Ann Neurol 12, 129144. Herman, J. P., Figueiredo, H., Mueller, N. K.,
Goossens, L., Sunaert, S., Peeters, R., Griez, E. J., Ulrich-Lai, Y., Ostrander, M. M., Choi, D. C.,
Schruers, K. R. 2007. Amygdala hyperfunction Cullinan, W. E. 2003. Central mechanisms of
548 Part IV:Homeostatic Therapies

stress integration: Hierarchical circuitry con- during acute and chronic stress. Ann NY Acad
trolling hypothalamo-pituitary-adrenocortical Sci 1148,6473.
responsiveness. Front Neuroendocrinol 24, Jodo, E., Chiang, C., Aston-Jones, G. 1998. Potent
151180. excitatory influence of prefrontal cortex activit y
Herman, J. P., Patel, P. D., Akil, H., Watson, S. J. on noradrenergic locus coeruleus neurons.
1989. Localization and regulation of glucocorti- Neuroscience 83,6379.
coid and mineralocorticoid receptor messenger Joels, M., Baram, T.Z. 2009. The neuro-symphony of
RNAs in the hippocampal formation of the rat. stress. Nat Rev Neurosci 10, 459466.
Mol Endocrinol 3, 18861894. Kessler, R. C., Berglund, P., Demler, O., Jin, R.,
Hill, M.N., McLaughlin, R.J., Bingham, B., Shrestha, L., Merikangas, K.R., Walters, E.E. 2005. Lifetime
Lee, T. T., Gray, J. M., Hillard, C. J., Gorzalka, prevalence and age-of-onset distributions of
B. B., Viau, V. 2010. Endogenous cannabinoid DSM-IV disorders in the National Comorbidity
signaling is essential for stress adaptation. Proc Survey Replication. Arch Gen Psychiatry 62,
Natl Acad Sci USA 107, 94069411. 593602.
Hill, M. N., Tasker, J. G. 2012. Endocannabinoid Kramer, M.S., Cutler, N.R., Ballenger, J.C., Patterson,
signaling, glucocorticoid-mediated nega- W.M., Mendels, J., Chenault, A., Shrivastava, R.,
tive feedback, and regulation of the Matzura-Wolfe, D., Lines, C., Reines, S. 1995. A
hypothalamic-pituitary-adrenal axis. placebo-controlled trial of L-365,260, a CCKB
Neuroscience 204,516. antagonist, in panic disorder. Biol Psychiatry 37,
Hohoff, C. 2009. Anxiety in mice and men:Acom- 462466.
parison. J Neural Transm 116, 679687. Kratochvil, C. J., Harrington, M. J., Burke, W. J.,
Holbrook, T.L., Hoyt, D.B., Stein, M.B., Sieber, W.J. March, J. S. 2002. Pharmacotherapy of child-
2002. Gender differences in long-term post- hood anxiety disorders. Curr Psychiatry Rep 4,
traumatic stress disorder outcomes after major 264269.
trauma:Women are at higher risk of adverse out- LeDoux, J. E. 2000. Emotion circuits in the brain.
comes than men. J Trauma 53, 882888. Annu Rev Neurosci 23, 155184.
Holmes, A., Wellman, C. L. 2009. Stress-induced Lee, J. H., Dunner, D. L. 2008. The effect of anxi-
prefrontal reorganization and executive dys- ety disorder comorbidity on treatment resistant
function in rodents. Neurosci Biobehav Rev 33, bipolar disorders. Depress Anxiety 25,9197.
773783. Louvart, H., Maccari, S., Lesage, J., Leonhardt, M.,
Holzschneider, K., Mulert, C. 2011. Neuroimaging in Dickes-Coopman, A., Darnaudery, M. 2006.
anxiety disorders. Dialogues Clin Neurosci 13, Effects of a single footshock followed by situ-
453461. ational reminders on HPA axis and behaviour
Inoue, T., Tsuchiya, K., Koyama, T. 1994. Regional in the aversive context in male and female rats.
changes in dopamine and serotonin activation Psychoneuroendocrinology 31,9299.
with various intensity of physical and psycholog- Low, K., Crestani, F., Keist, R., Benke, D., Brunig, I.,
ical stress in the rat brain. Pharmacol Biochem Benson, J. A., Fritschy, J. M., Rulicke, T.,
Behav 49, 911920. Bluethmann, H., Mohler, H., et al. 2000.
Iny, L.J., Pecknold, J., Suranyi-Cadotte, B.E., Bernier, B., Molecular and neuronal substrate for the selec-
Luthe, L., Nair, N.P., Meaney, M.J. 1994. Studies tive attenuation of anxiety. Science 290, 131134.
of a neurochemical link between depression, Maccarrone, M. 2010. Endocannabinoid-binding
anxiety, and stress from [3H]imipramine and receptors: Old friends and new comers. Curr
[3H]paroxetine binding on human platelets. Biol Med Chem 17, 13391340.
Psychiatry. 36, 28191. Marco, E. M., Garcia-Gutierrez, M. S.,
Isaac, C.L., Cushway, D., Jones, G.V. 2006. Is post- Bermudez-Silva, F.J., Moreira, F.A., Guimaraes, F.,
traumatic stress disorder associated with specific Manzanares, J., Viveros, M. P. 2011.
deficits in episodic memory? Clin Psychol Rev Endocannabinoid system and psychiatry: In
26, 939955. search of a neurobiological basis for detrimental
Isserles, M., Shalev, A.Y., Roth, Y., Peri, T., Kutz, I., and potential therapeutic effects. Front Behav
Zlotnick, E., Zangen, A. 2013. Effectiveness Neurosci5,63.
of deep transcranial magnetic stimulation Marco, E.M., Laviola, G. 2012. The endocannabinoid
combined with a brief exposure procedure in system in the regulation of emotions throughout
post-traumatic stress disordera pilot study. lifespan: A discussion on therapeutic perspec-
Brain Stimul 6, 377383. tives. J Psychopharmacol 26, 150163.
Jankord, R., Herman, J.P. 2008. Limbic regulation of Marsicano, G., Wotjak, C.T., Azad, S.C., Bisogno, T.,
hypothalamo-pituitary-adrenocortical function Rammes, G., Cascio, M.G., Hermann, H., Tang,
Anxiety and Stress Disorders 549

J., Hofmann, C., Zieglgansberger, W., etal. 2002. Nutt, D. J., Malizia, A. L. 2001. New insights into
The endogenous cannabinoid system controls the role of the GABA(A)-benzodiazepine recep-
extinction of aversive memories. Nature 418, tor in psychiatric disorder. Br J Psychiatry 179,
530534. 390396.
Mazor, A., Matar, M.A., Kaplan, Z., Kozlovsky, N., Pacher, P., Batkai, S., Kunos, G. 2006. The endocan-
Zohar, J., Cohen, H. 2009. Gender-related quali- nabinoid system as an emerging target of phar-
tative differences in baseline and post-stress macotherapy. Pharmacol Rev 58, 389462.
anxiety responses are not reflected in the inci- Pande, A.C., Greiner, M., Adams, J.B., Lydiard, R.B.,
dence of criterion-based PTSD-like behaviour Pierce, M. W. 1999. Placebo-controlled trial of
patterns. World J Biol Psychiatry 10, 856869. the CCK-B antagonist, CI-988, in panic disorder.
McCormick, C. M., Linkroum, W., Sallinen, B. J., Biol Psychiatry 46, 860862.
Miller, N. W. 2002. Peripheral and central sex Parsons, R. G., Ressler, K. J. 2013. Implications of
steroids have differential effects on the HPA axis memory modulation for post-traumatic stress
of male and female rats. Stress 5, 235247. and fear disorders. Nat Neurosci 16, 146153.
McEwen, B. S. 2003. Mood disorders and allostatic Paslawski, T., Treit, D., Baker, G. B., George, M.,
load. Biol Psychiatry 54, 200207. Coutts, R. T. 1996. The antidepressant drug
McKernan, R. M., Rosahl, T. W., Reynolds, D. S., phenelzine produces antianxiety effects in the
Sur, C., Wafford, K.A., Atack, J.R., Farrar, S., plus-maze and increases in rat brain GABA.
Myers, J., Cook, G., Ferris, P., et al. 2000. Psychopharmacology (Berl) 127,1924.
Sedative but not anxiolytic properties of benzo- Passie, T., Emrich, H. M., Karst, M., Brandt, S. D.,
diazepines are mediated by the GABA(A) recep- Halpern, J.H. 2012. Mitigation of post-traumatic
tor alpha1 subtype. Nat Neurosci 3, 587592. stress symptoms by Cannabis resin:Areview of
Merikangas, K. R., Zhang, H., Avenevoli, S., the clinical and neurobiological evidence. Drug
Acharyya, S., Neuenschwander, M., Angst, J. Test Anal 4, 649659.
2003. Longitudinal trajectories of depression and Patchev, V. K., Almeida, O. F. 1998. Gender speci-
anxiety in a prospective community study: The ficity in the neural regulation of the response to
Zurich Cohort Study. Arch Gen Psychiatry 60, stress:New leads from classical paradigms. Mol
9931000. Neurobiol 16,6377.
Millan, M.J. 2003. The neurobiology and control of Patel, S., Roelke, C.T., Rademacher, D.J., Hillard, C.J.
anxious states. Prog Neurobiol 70, 83244. 2005. Inhibition of restraint stress-induced
Moreira, F.A., Aguiar, D.C., Guimaraes, F.S. 2006. neural and behavioural activation by endoge
Anxiolytic-like effect of cannabidiol in the rat nous cannabinoid signalling. Eur J Neurosci 21,
Vogel conflict test. Prog Neuropsychopharmacol 10571069.
Biol Psychiatry 30, 14661471. Peters, T.E., Connolly, S. 2012. Psychopharmacologic
Moreira, F.A., Lutz, B. 2008. The endocannabinoid treatment for pediatric anxiety disorders. Child
system:Emotion, learning and addiction. Addict Adolesc Psychiatr Clin N Am 21, 789806.
Biol 13, 196212. Phelps, E.A., LeDoux, J.E. 2005. Contributions of the
Moreira, F.A., Wotjak, C.T. 2010. Cannabinoids and amygdala to emotion processing: From animal
anxiety. Curr Top Behav Neurosci 2, 429450. models to human behavior. Neuron 48, 175187.
Myers-Schulz, B., Koenigs, M. 2012. Functional Piomelli, D., Tarzia, G., Duranti, A., Tontini, A., Mor,
anatomy of ventromedial prefrontal cor- M., Compton, T.R., Dasse, O., Monaghan, E.P.,
tex: Implications for mood and anxiety disor- Parrott, J.A., Putman, D. 2006. Pharmacological
ders. Mol Psychiatry 17, 132141. profile of the selective FAAH inhibitor KDS-4103
Neumann, I.D., Wegener, G., Homberg, J.R ., Cohen, H., (URB597). CNS Drug Rev 12,2138.
Slattery, D.A., Zohar, J., Olivier, J.D., Mathe, A.A. Plendl, W., Wotjak, C. T. 2010. Dissociation of
2011. Animal models of depression and anxi- within- and between-session extinction of con-
ety:What do they tell us about human condition? ditioned fear. J Neurosci 30, 49904998.
Prog Neuropsychopharmacol Biol Psychiatry 35, Pratchett, L. C., Pelcovitz, M. R., Yehuda, R. 2010.
13571375. Trauma and violence: Are women the weaker
Nishijo, H., Ono, T., Nishino, H. 1988. Single neuron sex? Psychiatr Clin North Am 33, 465474.
responses in amygdala of alert monkey during Rabinak, C. A., Phan, K. L. 2014. Cannabinoid
complex sensory stimulation with affective sig- mo dulation of fear extinction brain cir-
nificance. J Neurosci 8, 35703583. cuits: A novel target to advance anxiety treat-
Nutt, D. J. 2001. Neurobiological mechanisms in ment. Curr Pharm Des 20, 22122217
generalized anxiety disorder. J Clin Psychiatry Ramboz, S., Oosting, R., Amara, D.A., Kung, H.F.,
62 Suppl 11, 2227; discussion28. Blier, P., Mendelsohn, M., Mann, J.J., Brunner, D.,
550 Part IV:Homeostatic Therapies

Hen, R. 1998. Serotonin receptor 1A knock- Rodriguez-Romaguera, J., Do Monte, F. H., Quirk,
out: An animal model of anxiety-related disor- G.J. 2012. Deep brain stimulation of the ventral
der. Proc Natl Acad Sci USA 95, 1447614481. striatum enhances extinction of conditioned
Rasmussen, K., Morilak, D. A., Jacobs, B. L. 1986. fear. Proc Natl Acad Sci USA 109, 87648769.
Single unit activity of locus coeruleus neurons Rotzinger, S., Lovejoy, D. A., Tan, L. A. 2010.
in the freely moving cat: I. During naturalistic Behavioral effects of neuropeptides in rodent
behaviors and in response to simple and complex models of depression and anxiety. Peptides 31,
stimuli. Brain Res 371, 324334. 736756.
Rauch, S. L., Shin, L. M., Wright, C. I. 2003. Rotzinger, S., Vaccarino, F.J. 2003. Cholecystokinin
Neuroimaging studies of amygdala function receptor subtypes: Role in the modulation of
in anxiety disorders. Ann NY Acad Sci 985, anxiety-related and reward-related behaviours
389410. in animal models. J Psychiatry Neurosci 28,
Ravindran, L.N., Stein, M.B. 2010. The pharmaco- 171181.
logic treatment of anxiety disorders:Areview of Rubin, D. C., Berntsen, D., Bohni, M. K. 2008. A
progress. J Clin Psychiatry 71, 839854. memory-based model of posttraumatic stress
Redrobe, J. P., Dumont, Y., Quirion, R. 2002. disorder: Evaluating basic assumptions under-
Neuropeptide Y (NPY) and depression: From lying the PTSD diagnosis. Psychol Rev 115,
animal studies to the human condition. Life Sci 9851011.
71, 29212937. Rudolph, U., Crestani, F., Benke, D., Brunig, I.,
Reinhold, J. A., Mandos, L. A., Rickels, K., Lohoff, Benson, J. A., Fritschy, J. M., Martin, J. R.,
F. W. 2011. Pharmacological treatment of Bluethmann, H., Mohler, H. 1999.
generalized anxiety disorder. Expert Opin Benzodiazepine actions mediated by specific
Pharmacother 12, 24572467. gamma-aminobutyric acid(A) receptor sub-
Research Unit on Pediatric Psychopharmacology. types. Nature 401, 796800.
2001. Fluvoxamine for the treatment of anxiety Rudolph, U., Knoflach, F. 2011. Beyond classical
disorders in children and adolescents. N Engl J benzodiazepines:Novel therapeutic potential of
Med 344, 12791285. GABAA receptor subtypes. Nat Rev Drug Discov
Ressler, K. J., Nemeroff, C. B. 2000. Role of seroto- 10, 685697.
nergic and noradrenergic systems in the patho- Rynn, M. A., Siqueland, L., Rickels, K. 2001.
physiology of depression and anxiety disorders. Placebo-controlled trial of sertraline in the treat-
Depress Anxiety 12 Suppl 1,219. ment of children with generalized anxiety disor-
Reul, J.M., de Kloet, E.R. 1986. Anatomical resolu- der. Am J Psychiatry 158, 20082014.
tion of two types of corticosterone receptor sites Sajdyk, T.J., Johnson, P.L ., Leitermann, R.J., Fitz, S.D.,
in rat brain with in vitro autoradiography and Dietrich, A., Morin, M., Gehlert, D.R ., Urban, J.H.,
computerized image analysis. J Steroid Biochem Shekhar, A. 2008. Neuropeptide Y in the
24, 269272. amygdala induces long-term resilience to
Riddle, M.A., dosReis, S., Reeves, G.M., Wissow, L.S., stress-induced reductions in social responses but
Pruitt, D. B., Foy, J. M. 2013. Pediatric psycho- not hypothalamic-adrenal-pituitary axis activity
pharmacology in primary care: A conceptual or hyperthermia. J Neurosci 28, 893903.
framework. Adolesc Med State Art Rev 24, Sandford, J.J., Argyropoulos, S.V., Nutt, D.J. 2000.
371390. The psychobiology of anxiolytic drugs: Part 1.
Riddle, M.A., Reeve, E.A., Yaryura-Tobias, J.A., Yang, Basic neurobiology. Pharmacol Ther 88,
H.M., Claghorn, J.L., Gaffney, G., Greist, J.H., 197212.
Holland, D., McConville, B. J., Pigott, T., et al. Sarapas, C., Cai, G., Bierer, L. M., Golier, J. A.,
2001. Fluvoxamine for children and adolescents Galea, S., Ising, M., Rein, T., Schmeidler, J.,
with obsessive-compulsive disorder:Arandom- Muller-Myhsok, B., Uhr, M., etal. 2011. Genetic
ized, controlled, multicenter trial. J Am Acad markers for PTSD risk and resilience among
Child Adolesc Psychiatry 40, 222229. survivors of the World Trade Center attacks. Dis
Riebe, C. J., Wotjak, C. T. 2011. Endocannabinoids Markers 30, 101110.
and stress. Stress 14, 384397. Sepinwall, J., Grodsky, F. S., Cook, L. 1978.
Riedel-Heller, S. G., Busse, A., Angermeyer, M. C. Conflict behavior in the squirrel mon-
2006. The state of mental health in old-age across key: Effects of chlordiazepoxide, diazepam and
the old European Uniona systematic review. N-desmethyldiazepam. J Pharmacol Exp Ther
Acta Psychiatr Scand 113, 388401. 204, 88102.
Anxiety and Stress Disorders 551

Shansky, R. M., Lipps, J. 2013. Stress-induced cog- van Megen, H.J., Westenberg, H.G., den Boer, J.A.,
nitive dysfunction: Hormoneneurotransmitter Slaap, B., van Es-Radhakishun, F., Pande, A.C.
interactions in the prefrontal cortex. Front Hum 1997. The cholecystokinin-B receptor antagonist
Neurosci 7,123. CI-988 failed to affect CCK-4 induced symptoms
Shephard, R. A., Broadhurst, P. L. 1983. in panic disorder patients. Psychopharmacology
Hyponeophagia in the Roman rat strains: (Berl) 129, 243248.
Effects of 5-methoxy-N,N-dimethyltryptamine, Velosa, J. F., Riddle, M. A. 2000. Pharmacologic
diazepam, methysergide and the stereoisomers treatment of anxiety disorders in children and
of propranolol. Eur J Pharmacol 95, 177184. adolescents. Child Adolesc Psychiatr Clin N Am
Shin, L. M., Liberzon, I. 2010. The neurocir- 9, 119133.
cuitry of fear, stress, and anxiety disorders. Vermetten, E., Schmahl, C., Southwick, S. M.,
Neuropsychopharmacology 35, 169191. Bremner, J.D. 2007. Positron tomographic emis-
Shin, L. M., Rauch, S. L., Pitman, R. K. 2006. sion study of olfactory induced emotional recall
Amygdala, medial prefrontal cortex, and hip- in veterans with and without combat-related
pocampal function in PTSD. Ann NY Acad Sci posttraumatic stress disorder. Psychopharmacol
1071,6779. Bull 40,830.
Simpson, J., Kelly, J. P. 2012. An investigation of Vertes, R. P. 2004. Differential projections of the
whether there are sex differences in certain infralimbic and prelimbic cortex in the rat.
behavioural and neurochemical parameters in Synapse 51,3258.
the rat. Behav Brain Res 229, 289300. Viveros, M. P., Marco, E. M., File, S. E. 2005.
Simpson, J., Ryan, C., Curley, A., Mulcaire, J., Endocannabinoid system and stress and anxi-
Kelly, J.P. 2012. Sex differences in baseline and ety responses. Pharmacol Biochem Behav 81,
drug-induced behavioural responses in classical 331342.
behavioural tests. Prog Neuropsychopharmacol Viveros, M. P., Mendrek, A., Paus, T.,
Biol Psychiatry 37, 227236. Lopez-Rodriguez, A.B., Marco, E.M., Yehuda, R.,
Stein, M.B., Lang, A.J. 2002. Anxiety and stress dis- Cohen, H., Lehrner, A., Wagner, E. J. 2012.
orders:Course over the lifetime. In:Davis, K.L., A comparative, developmental, and clinical
Charney, D. S., Coyle, J. T., Nemeroff, C. perspective of neurobehavioral sexual dimor-
(Eds.), Neuropsychopharmacology: The Fifth phisms. Front Neurosci6,84.
Generation Progress (Philadelphia: Lippincott/ Vogel, J. R., Beer, B., Clody, D. E. 1971. A simple
Williams & Wilkins). and reliable conflict procedure for testing
Ter Horst, G. J., Wichmann, R., Gerrits, M., anti-anxiety agents. Psychopharmacologia
Westenbroek, C., Lin, Y. 2009. Sex differences 21,17.
in stress responses:Focus on ovarian hormones. Voshaar, R.C. 2013. Lack of interventions for anxiety
Physiol Behav 97, 239249. in older people. Br J Psychiatry 203,89.
Thomsen, P. H., Ebbesen, C., Persson, C. 2001. Wetherell, J. L., Petkus, A. J., Thorp, S. R., Stein,
Long-term experience with citalopram in the M.B., Chavira, D.A., Campbell-Sills, L., Craske,
treatment of adolescent OCD. J Am Acad Child M. G., Sherbourne, C., Bystritsky, A., Sullivan,
Adolesc Psychiatry 40, 895902. G., et al. 2013. Age differences in treatment
Tolin, D.F., Foa, E.B. 2006. Sex differences in trauma response to a collaborative care intervention for
and posttraumatic stress disorder: A quantita- anxiety disorders. Br J Psychiatry 203,6572.
tive review of 25years of research. Psychol Bull Wittchen, H.U., Jacobi, F., Rehm, J., Gustavsson, A.,
132, 959992. Svensson, M., Jonsson, B., Olesen, J., Allgulander,
Toufexis, D. J., Myers, K. M., Davis, M. 2006. The C., Alonso, J., Faravelli, C., et al. 2011. The size
effect of gonadal hormones and gender on anxiety and burden of mental disorders and other
and emotional learning. Horm Behav 50, 539549. disorders of the brain in Europe 2010. Eur
Trezza, V., Campolongo, P. 2013. The endocan- Neuropsychopharmacol 21, 655679.
nabinoid system as a possible target to treat Woodruff, G. N., Hill, D. R., Boden, P., Pinnock,
both the cognitive and emotional features of R., Singh, L., Hughes, J. 1991. Functional role
post-traumatic stress disorder (PTSD). Front of brain CCK receptors. Neuropeptides 19
Behav Neurosci 7,100. Suppl,4556.
Ulrich-Lai, Y. M., Herman, J. P. 2009. Neural reg- Yehuda, R. 2006. Advances in understanding neuro-
ulation of endocrine and autonomic stress endocrine alterations in PTSD and their therapeu-
responses. Nat Rev Neurosci 10, 397409. tic implications. Ann NY Acad Sci 1071, 137166.
552 Part IV:Homeostatic Therapies

Yehuda, R., Brand, S., Yang, R.K. 2006. Plasma neu- cannabidiol on human experimental anxiety. J
ropeptide Y concentrations in combat exposed Psychopharmacology 7,8288.
veterans: Relationship to trauma exposure, Zuardi, A.W., Shirakawa, I., Finkelfarb, E., Karniol,
recovery from PTSD, and coping. Biol Psychiatry I.G. 1982. Action of cannabidiol on the anxiety
59, 660663. and other effects produced by delta 9-THC in
Zuardi, A. W., Cosme, R. A., Graeff, F. G., normal subjects. Psychopharmacology (Berl) 76,
Guimares, F.S. 1993. Effects of ipsapirone and 245250.
29
Malignant Brain Cancer Management
withMetabolic Therapy
T HOM AS N.SE Y FR IED A ND PUR NA MUK HER JEE

G L I O B L A S T O M A M U LT I F O R M E analysis from the cancer genome project, no


Glioblastoma multiforme (GBM) is the most mutation is known that is unique to the GBM and
malignant of the primary brain cancers, with no genetic alterations are seen in major signaling
only about 12% of patients surviving beyond pathways in about 15% of GBM (Parsons et al.,
36 months (long-term survivors) (Fisher and 2008). Moreover, few of the personalized molec-
Buffler, 2005; Krex etal., 2007; Patil etal., 2012; ular markers available are considered important
Stupp et al., 2009). Most GBMs are heterogene- for GBM analysis or therapy (Holdhoff et al.,
ous in cellular composition, consisting of tumor 2012). Recent evidence suggests that the somatic
stem cells, mesenchymal cells, and host stromal mutations seen in cancer cells can arise as down-
cells; hence the name glioblastoma multiforme stream secondary effects of disturbed energy
(Chen et al., 2010; Ohgaki and Kleihues, 2009; metabolism and are unlikely to provide useful
Prestegarden et al., 2010; Rubinstein, 1972; Tso information for therapeutic treatment strategies
et al., 2006). Primary GBM appears to arise de for the majority of GBM patients (Johnson etal.,
novo, while secondary GBM is thought to arise 2014; Poff etal., 2014; Seyfried, 2012a). Reactive
from less malignant glial tumors (Lopes et al., oxygen species (ROS), generated from protracted
1993; Ohgaki and Kleihues, 2009). In addition to disruption of oxidative phosphorylation, cause
the neoplastic cell populations, tumor-associated the nuclear genomic instability and mutations
macrophages/monocytes (TAM) also comprise a seen in tumor cells (Cooke et al., 2003; Iglesias
significant cell population in GBM, sometimes et al., 2012; Seoane et al., 2011). Therefore, the
equaling the number of tumor cells (Morantz genomic approach to brain cancer management
etal., 1979; Nishie etal., 1999; Phillips etal., 1982; is unlikely to produce effective therapies in light
Seyfried, 2001; Seyfried et al., 2010; Shinonaga of the underlying metabolic origin of the disease.
et al., 1988). TAM can indirectly contribute to
tumor progression through release of proinflam- T H E C U R R E N T S TA N D A R D
matory and pro-angiogenic factors (Lewis and OFCARE FORGLIOBLASTOMA
Murdoch, 2005; Nishie et al., 1999; Seyfried, The current standard of care for GBM and many
2001; Seyfried etal., 2010). Neoplastic cells with malignant brain cancers includes maximum sur-
macrophage characteristics might also contribute gical resection, radiation therapy, and chemo-
to the sarcomatous characteristics of many GBM therapy (Fisher and Buffler, 2005; Mason et al.,
(Huysentruyt etal., 2011; Lopes etal., 1993). The 2007; Mrugala, 2013; Stupp etal., 2009). The toxic
neoplastic cells in GBM invade through the neural alkylating agent temozolomide (Temodar) is the
parenchyma well beyond the main tumor mass, most common chemotherapy used for treating
making complete surgical resections exceed- GBM. Most GBM patients also receive periop-
ingly rare (Fisher and Buffler, 2005; Kallenberg erative corticosteroids (dexamethasone) as part
etal., 2009; Talacchi etal., 2010). Moreover, GBM of the standard of care, which is often extended
exhibits the highest incidence of systemic metas- throughout the course of the disease (Chang
tasis among glial neoplasms, while many cells etal., 2005; Koehler, 1995; Seyfried etal., 2010).
appearing as TAM within GBM can actually be The anti-angiogenic drug bevacizumab (Avastin)
neoplastic macrophages/microglia (Huysentruyt is also given often to GBM patients despite the
etal., 2011; Lopes etal., 1993). Despite extensive Food and Drug Administrations removal of
554 Part IV:Homeostatic Therapies
100 et al., 2002; John, 2001; Kiebish et al., 2008;
90 Combined
Radiotherapy
Ordys et al., 2010; Pedersen, 1978; Ramanathan
80
etal., 2005; Roskelley etal., 1943; Villalobo and
70 p < 0.0001 Lehninger, 1979). The ultra structure of mito-
Survival (%)

60
50
chondria in malignant brain tumors differs mark-
40 edly from the ultra structure of normal tissue
30 mitochondria (Arismendi-Morillo, 2009, 2011;
20 Arismendi-Morillo and Castellano-Ramirez,
n = 287
10
n = 286
2008). In contrast to normal mitochondria,
0
0 1 2 3 4 5 6 7
which contain numerous cristae, mitochondria
Time (years) from GBM tissue samples show swelling with
Number at risk partial or total cristolysis (Figure 29.2). Cristae
Combined 254 175 76 39 23 14 6
contain the proteins of the respiratory com-
Radiotherapy 278 144 31 11 6 3 0
plexes and play an essential structural role in
FIGURE 29.1: Kaplan-Meier estimates of over- facilitating energy production through OxPhos
all survival of patients with glioblastoma (Cogliati etal., 2013; Galluzzi etal., 2010; Ordys
multiforme by treatment group. The two patient et al., 2010; Stroud and Ryan, 2013). The struc-
groups included radiotherapy alone (n=278) and radi- tural defects in human glioma mitochondria are
otherapy with temolozomide (n=254). Overall patient also consistent with lipid biochemical defects in
survival has remained largely unchanged from the murine gliomas. We showed that cardiolipin,
study published in 2005 (Stupp etal.,2005). the signature phospholipid of the inner mito-
Reprinted with permission from Stupp etal. (2009). Readers chondrial membrane, was abnormal in five inde-
are referred to the original papers for details. pendently derived mouse brain tumors (Kiebish
etal., 2008; Kiebish etal., 2009). Cardiolipin con-
trols the efficiency of OxPhos, and any alterations
bevacizumab for breast cancer due to toxicity in the content or fatty acid composition of car-
and lack of efficacy (Burton and Dooren, 2011; diolipin will reduce cellular respiration (Chicco
Seyfried etal., 2012). There have been no major and Sparagna, 2007; Claypool and Koehler, 2012;
advances in GBM management for over 50years, Fry and Green, 1981). In addition to these find-
though use of temozolomide has produced ings, Oudard and etal (1997) also indicated that
marginal improvement in patient survival over the high glycolytic activity seen in malignant gli-
radiation therapy alone (Souhami et al., 2004; omas could arise from mitochondrial structural
Stupp et al., 2009). Despite conventional treat- abnormalities. Hence substantial morphological
ments, prognosis remains poor for most patients and biochemical evidence exists showing that
with high-grade brain tumors (Davis et al., respiratory capacity is defective in most tumors,
1998; Fisher and Buffler, 2005; Krex etal., 2007; including gliomas.
Mrugala, 2013; Souhami etal., 2004; Figure 29.1). Based on numerous findings in human gli-
It is our view that the therapies used as part of the oma cell lines and tissues, it is not likely that the
standard of care are responsible in part for the majority of malignant brain tumors are capa-
poor prognosis for patients withGBM. ble of producing adequate amounts of energy
through OxPhos (Arismendi-Morillo, 2009, 2011;
MITOCHONDRIAL Arismendi-Morillo and Castellano-Ramirez,
ABNORMALITIES 2008; Oudard et al., 1996; Oudard et al., 1997;
INMALIGNANT Oudard etal., 1995). Besides these ultrastructure
BR AINTUMORS findings, Renner and colleagues (2010) showed
Substantial evidence collected from numer- that tumor cells isolated from human GBM could
ous investigators over many years indicates that produce adenosine triphosphate (ATP) in the
mitochondrial abnormalities are the hallmark of presence of potassium cyanide. Cyanide blocks
all cancers, including brain cancer (Ordys etal., cytochrome c oxidase and kills normal control
2010; Seyfried et al., 2014a). These mitochon- cells, which obtain energy through OxPhos.
drial abnormalities reduce energy production Mitochondrial energy production in the pres-
through oxidative phosphorylation (OxPhos) ence of cyanide suggests that OxPhos is not likely
(Arismendi-Morillo, 2009; Arismendi-Morillo the origin of the energy produced in these GBM
and Castellano-Ramirez, 2008; Bayley and cells. Similar findings were reported recently
Devilee, 2010; Carew and Huang, 2002; Cuezva for mitochondria in pancreatic tumor cells
Malignant Brain Cancer Management with Metabolic Therapy 555

Normal Mitochondria GBM Mitochondria

inter membrane
matrix cristac inner space outer
membrane membrane

FIGURE29.2: Typical ultrastructure of a normal mitochondrion and a mitochondrion from GBM


tissue. Normal mitochondria contain elaborate cristae, which are extensions of the inner membrane and con-
tain the protein complexes of the electron transport chain necessary for producing ATP through OxPhos. The
mitochondrion from the GBM (m)is enlarged and shows a near total breakdown of cristae (cristolysis) and an
electron-lucent matrix. The absence of cristae in GBM mitochondria indicates that OxPhos would be deficient.
The arrow indicates an inner membrane fold. Bar:0.33m. Method of staining:uranyl acetate/lead citrate. The
GBM mitochondria was reprinted with permission from Arismendi-Morillo and Castellano-Ramirez (2008). The
normal mitochondria and diagram are from http://academic.brooklyn.cuny.edu/biology/bio4fv/page/mito.htm.

(James etal., 2013). These and other studies sug- energy through respiration, (c)cancer cells con-
gest that OxPhos is deficient in malignant glio- tinue to ferment lactate in the presence of oxy-
mas and other malignant cancers and that energy gen, and (d)respiratory insufficiency eventually
through oxidative metabolism alone would be becomes irreversible (Pedersen, 2007; Warburg,
incapable of maintaining viability in gliomacells. 1931, 1956a, 1956b, 1969). Warburg referred
to the phenomenon of enhanced glycolysis in
T H E WA R B U R G E F F E C T cancer cells as aerobic fermentation to high-
INMALIGNANT light the abnormal production of lactate in the
BR AINTUMORS presence of oxygen (Pedersen, 2007; Warburg,
Otto Warburg (1931, 1956a) first proposed that 1931, 1956a, 1956b, 1969). The Warburg effect
all cancers arise from irreversible damage to cel- refers to the aerobic fermentation of cancer cells
lular respiration. As a result, cancer cells increase (Ferreira, 2010; Seyfried, 2012h). Substantial evi-
their capacity to ferment lactate even in the pres- dence exists showing that gliomas produce lactate
ence of oxygen in order to compensate for their (Lichtor and Dohrmann, 1986; Seyfried, 2012g;
insufficient respiration. Although confusion has Seyfried and Mukherjee, 2005). Lactate is the end
surrounded Warburgs hypothesis on the ori- product of pyruvate fermentation. This would
gin of tumor cells (Koppenol etal., 2011; Zu and be expected for any tumor cell with quantitative
Guppy, 2004), his hypothesis has never been for- or qualitative or abnormalities in mitochondria.
mally disproved and remains a credible explana- The large number of mutations found in tumor
tion for the origin of cancer (Cuezva etal., 2004; cells is considered downstream epiphenomena
Ferreira, 2010; Seyfried, 2012h; Seyfried and of damaged or insufficient respiration (Seyfried
Shelton, 2010). Consequently, Warburgs expla- etal., 2014b).
nation for the origin of cancer can no longer be As the result of insufficient respiration,
viewed as a hypothesis but can now be viewed as cancer cells must rely on nonoxidative energy
a theory (Seyfried, 2012a). metabolism to maintain energy balance and via-
The key points of Warburgs theory are bility. Consequently, aerobic fermentation plays a
(a) insufficient respiration initiates tumorigen- role in producing energy through substrate-level
esis and ultimately cancer, (b) energy through phosphorylation in the cytoplasm (glycolysis;
glycolysis gradually compensates for insufficient Seyfried et al., 2014a; Warburg, 1956b). Besides
556 Part IV:Homeostatic Therapies

aerobic fermentation in the cytoplasm, the tricar- mice were corroborated with similar findings in
boxylic acid cycle substrate level phosphorylation humans.
might also produce ATP through nonoxidative Moreover, we found that the expression of
metabolism. It can be difficult to determine, insulin-like growth factor 1 (IGF-1) was also
however, the degree to which mitochondrial dependent on circulating glucose levels (Marsh
ATP production arises from coupled respira- etal., 2008a; Seyfried etal., 2003). IGF-1 is a cell
tion or from tricarboxylic acid cycle substrate surface receptor linked to rapid tumor growth
level phosphorylation (Chinopoulos etal., 2010; through the PI3K/Akt signaling pathway (Marsh
Phillips et al., 2009; Schwimmer et al., 2005; et al., 2008a). The association of plasma IGF-1
Seyfried, 2012d). A protracted reliance on non- levels with tumor growth rate is due in part to
oxidative energy metabolism involving glucose elevated levels of blood glucose. These findings
and amino acid fermentation with substrate in animal models and in brain cancer patients
level phosphorylation can cause genomic insta- indicate that tumor growth rate and prognosis is
bility and other recognized hallmarks of cancer dependent to a significant extent on circulating
(Seyfried etal., 2014a). Emerging evidence indi- glucose levels. Glucose is the prime fuel for gly-
cates that the function of DNA repair enzymes colysis, which drives growth of most brain cancer
and the integrity of the nuclear genome are (Oudard etal., 1996; Oudard etal., 1997; Seyfried
dependent to a large extent on the energy derived and Mukherjee, 2005). As long as circulating glu-
from normal respiration (Chandra and Singh, cose levels remain elevated, brain tumor growth
2011; Delsite et al., 2003; Kulawiec et al., 2008; will be difficult to manage.
Lu etal., 2009; Rasmussen etal., 2003; Smiraglia In addition to glucose, glutamine is also
etal., 2008; Veatch etal., 2009; Yang etal., 2010). suggested to play an important role in tumor
In other words, the Warburg effect and genomic energy metabolism (Alberghina et al., 2014;
instability ultimately arise from a protracted DeBerardinis and Cheng, 2010; Gao etal., 2009;
insufficiency of OxPhos (Seyfried et al., 2014a). Wise et al., 2008; Yuneva, 2008). Recent stud-
It remains speculative whether respiratory insuf- ies suggest that inhibition of the mitochondrial
ficiency is irreversible, as Warburg suggested, or OxPhos, and more specifically of Complex I, is
might be reversed through metabolic therapy. able to promote glutamine utilization (Fendt
It is difficult to imagine, however, how respira- et al., 2013). Glutamine is also a major energy
tion or the mitochondria cristolysis seen in GBM fuel for immune cells (Newsholme et al., 2003).
could be easily reversed (Figure29.2). As neoplastic myeloid cells can contribute to the
GBM tumor cell population, glutamine becomes
ROLE OFGLUCOSE AND an important fuel for GBM growth and inva-
G L U TA M I N E I N B R A I N sion (Huysentruyt et al., 2011). In contrast to
TUMOR PROGRESSION extracranial tissues where glutamine is the most
Glucose is the predominant fuel of the brain, available amino acid, glutamine is tightly regu-
but it also fuels tumor cell glycolysis as well as lated in the brain through its involvement in the
serving as a precursor for glutamate synthesis glutamate-glutamine cycle of neurotransmis-
(McKenna etal., 2006; Seyfried and Mukherjee, sion (Hawkins, 2009; McKenna et al., 2006).
2005; Seyfried et al., 2003; Warburg, 1956a). Glutamate is a major excitatory neurotransmitter
Using linear regression analysis, we showed that must be cleared rapidly following synaptic
that the growth rate of the CT-2A experimen- release in order to prevent excitotoxic damage
tal mouse astrocytoma was directly dependent to neurons (Hawkins, 2009; Takano etal., 2001).
on blood glucose levels (Seyfried et al., 2003). Glial cells possess transporters for the clear-
The higher the blood glucose levels, the faster ance of extracellular glutamate, which is then
the tumors grew. As glucose levels fall, tumor metabolized to glutamine for delivery back to
size and growth rate fall. Hyperglycemia not neurons. Neurons metabolize the glutamine to
only contributes to rapid tumor cell growth but glutamate, which is then repackaged into synap-
also enhances white matter damage in patients tic vesicles for future release (Hawkins, 2009). The
receiving radiation therapy (Szerlip etal., 2011). glutamate-glutamine cycle maintains low extra-
Hyperglycemia is also linked to poor prognosis cellular levels of both glutamate and glutamine
in humans with malignant brain cancer (Champ in normal neural parenchyma. Disruption of the
and Klement, 2015; Derr et al., 2009; McGirt glutamate-glutamine cycle can cause neurotox-
et al., 2008). In other words, our findings in icity and provide neoplastic GBM cells access
Malignant Brain Cancer Management with Metabolic Therapy 557

to glutamine, as we recently described (Seyfried 2014b). It is well documented that neurotoxicity


etal., 2014b). from mechanical trauma (surgery), radiotherapy,
and chemotherapy will increase tissue inflamma-
CYTOMEGALOVIRUS:AN tion and glutamate levels (Di Chiro etal., 1988;
O N C O M O D U L AT O R O F B R A I N Kallenberg et al., 2009; Lee et al., 2010; Monje
T U M O R E N E R G Y M E TA B O L I S M etal., 2007). Local astrocytes rapidly clear extra-
Many cancers including GBM are infected with cellular glutamate, metabolizing it to glutamine
human cytomegalovirus (HCMV), which acts for release to neurons. In the presence of dead
as an oncomodulator of tumor progression or dying neurons, however, surviving tumor
(Dziurzynski et al., 2012; Hawkins and Croul, cells and the TAMs will use astrocyte-derived
2011; Michaelis etal., 2009). Products of the virus glutamine for their energy and growth. TAMs
can damage mitochondria in the infected tumor also release pro-angiogenic and growth fac-
cells, thus contributing to a further dependence tors, which further stimulate tumor progres-
on glucose and glutamine for energy metabo- sion (Seyfried, 2001, 2012b; Seyfried etal., 2010).
lism (Bozidis etal., 2010; Cobbs, 2013; Seyfried, Radiation damage to tumor cell mitochondria
2012c; Williamson and Colberg-Poley, 2009; Yu will hasten a dependence on glucose and glu-
etal., 2011). The virus often infects cells of mono- tamine for growth and survival (Seyfried and
cyte/macrophage origin, which are considered Shelton, 2010; Warburg, 1956a). The HCMV
the origin of many metastatic cancers, includ- infection in the neoplastic GBM cells will further
ing GBM (Dziurzynski etal., 2011; Huysentruyt accelerate tumor cell growth through increased
etal., 2011; Munzarova etal., 1991; Pawelek and metabolism of glucose and glutamine (Yu etal.,
Chakraborty, 2008; Seyfried and Huysentruyt, 2011). Tumor radiation will also upregulate
2013). Indeed, we proposed that neoplastic the PI3K/Akt signaling pathway, which drives
microglia/macrophages were the most invasive glioma glycolysis and chemotherapeutic drug
cells within GBM (Huysentruyt etal., 2011). GBM resistance (Elstrom et al., 2004; Kargiotis et al.,
malignancy is correlated with the titer of HCMV 2010; Seyfried and Shelton, 2010; Xu etal., 2005;
infection: the higher the titer, the greater the Zhuang et al., 2009). In contrast to normal glia
malignancy (Dziurzynski et al., 2012). HCMV that metabolize glutamate to glutamine, neoplas-
infection increases glucose and glutamine metab- tic glioma cells secrete glutamate. It is not clear if
olism seen in tumor cells (Chambers etal., 2010; the secreted glutamate is derived from glutamine
Yu etal., 2011). Human fibroblasts infected with in the necrotic microenvironment or is synthe-
HCMV require glutamine for ATP production. sized from glucose. Glioma glutamate secre-
Infected cells starved for glutamine beginning 24 tion is thought to contribute in part to neuronal
hours postinfection failed to produce infectious excitotoxicity and tumor expansion (Takano
virions (Chambers et al., 2010). Thus HCMV et al., 2001). Any treatment that increases neu-
infection activates the mechanisms needed to rotoxicity will reduce patient survival (Lawrence
switch the anapleurotic substrate from glucose etal., 2011). The standard of care increases cen-
to glutamine (Chambers et al., 2010). We sug- tral nervous system toxicity. These observations
gest that HCMV infection will contribute to the indicate that the current standard of care creates
progression of GBM through an oncomodulatory a metabolic environment that would facilitate
effect on tumor cell energy metabolism. GBM progression.
In addition to the enhancing effects of radia-
D OE S T H E C U R R E N T STA N DA R D tion and HCMV on GBM energy metabolism,
OF C A R E ACC EL ER AT E G BM most GBM patients are also given high-dose
R EC U R R E NC E A N D PRO G R E SSION glucocorticoids (dexamethasone) (Chang etal.,
T H ROUG H EF F EC T S ON E N ERGY 2005). Although dexamethasone is given to
M E TA BOL ISM? reduce radiation-associated brain swelling and
It is our view that the current standard of care tumor edema, dexamethasone significantly
for GBM and other malignant brain cancers con- elevates blood glucose levels (Hans etal., 2006;
tributes to tumor recurrence and progression. Harris etal., 2013; Kargiotis etal., 2010; Lukins
This prediction comes from new information and Manninen, 2005; Noch and Khalili, 2009).
describing how the standard of care can enhance Glucose fuels tumor cell glycolysis as well as
the availability of glucose and glutamine within serving as a precursor for glutamate synthesis
the tumor microenvironment (Seyfried et al., (McKenna etal., 2006; Seyfried and Mukherjee,
558 Part IV:Homeostatic Therapies

2005; Seyfried et al., 2003; Warburg, 1956a). tumor cells and the inevitable recurrence of the
Any therapy that elevates blood glucose will tumor. The process is illustrated in Figure29.3.
not be in the best interest of the GBM patient. Although the existing standard of care for
Many GBM patients are also treated with the malignant brain cancer will increase patient sur-
anti-angiogenic drug bevacizumab (Avastin). vival over the short term (months) compared to
Bevacizumab targets leaky blood vessels, thus the no therapy option (Lawrence et al., 2012),
enhancing hypoxia and radiation-induced it is clear how this therapeutic strategy can accel-
necrosis in the tumor microenvironment. erate the energy metabolism of surviving tumor
Increased hypoxia will further enhance tumor cells. Moreover, the malignant phenotype of
cell glycolysis and select for those tumor cells brain tumor cells that survive radiotherapy is
with the greatest invasive properties (de Groot often greater than that of the cells from the origi-
et al., 2010; Iwamoto et al., 2009; Reardon nal tumor (Kargiotis etal., 2010). Treatments that
et al., 2011). Bevacizumab also exacerbates increase tumor energy metabolism will facilitate
radiation-induced necrosis, which will create a tumor cell growth and survival, thus decreas-
more favorable environment for tumor recur- ing overall patient survival. The data show that
rence (Jeyaretna et al., 2011). Viewed collec- only about 1.1% of GBM patients (6/532) become
tively, these findings further illustrate how the long-term survivors following radiation therapy
current standard of care will create a microen- (Figure 29.1). While GBM is certainly a deadly
vironment that facilitates the energy needs of disease, it remains to be determined if the current

Dexamethasone-induced
Radiation-induced
necrosis Hyperglycaemia

Dead and
dying neurons
Gln
Glu
Gln
RAC
Glu Tumour
Proinflammatory
cytokines
Glu Glu Proangiogenic factors
Gln: Glu cycle
Glu

Dying Gln TAM


Gln
neurons

Glu

Monocytes
from circulation

FIGURE 29.3: How the standard of care can accelerate brain tumor growth and recurrence.
GBM and other high-grade brain tumors consist of multiple neoplastic cell types as well as tumor-associated mac-
rophages (TAMs), which release proinflammatory and pro-angiogenic factors. All these cells will use glucose and
glutamine (Gln) as major metabolic fuels for their growth and survival. Recent evidence suggests that nearly all
GBM are infected with human cytomegalovirus, which enhances glucose and glutamine metabolism in the tumor
cells. Increased glutamate (Glu) concentrations will arise after radiation/drug-induced necrosis. Reactive astro-
cytes (RA) take up and metabolize glutamate to glutamine, whereas hyperglycemia will arise after corticosteroid
(dexamethasone) therapy. Together, these standard treatments will provide a microenvironment that facilitates
tumor cell growth, survival, and the likelihood of tumor recurrence (Seyfried etal.,2010).
With permission from Lancet Oncology.
Malignant Brain Cancer Management with Metabolic Therapy 559

standard of care makes the disease even worse becomes a prime target for GBM management.
(Fisher and Buffler, 2005; Stupp etal., 2009). This However, most normal cells of the brain also
situation is even more disturbing, as the standard need glycolytic pathway products, such as pyru-
of care is not considered curative but only pallia- vate, for energy production through OxPhos. It
tive (Preusser etal.,2011). therefore becomes important to protect normal
brain cells from drugs or therapies that disrupt
EXPLOITING MITOCHONDRIAL glycolytic pathways or cause systemic reduc-
DYSFUNCTION FOR THE tion of glucose. It is well known that ketones
M E TA B O L I C M A N A G E M E N T can replace glucose as an energy metabolite and
OF GBM can protect the brain from severe hypoglyce-
GBM, like most cancers, is primarily a disease mia (Drenick etal., 1972; VanItallie and Nufert,
of energy metabolism (Seyfried, 2012a; Seyfried 2003; Veech, 2004). Hence the shift in energy
etal., 2012; Seyfried etal., 2011). Rational strate- metabolism associated with a low carbohydrate,
gies for GBM management should therefore be high-fat ketogenic diet (KD) administered in
found in therapies that specifically target tumor restricted amounts (KD-R) can protect normal
cell energy metabolism. As glucose is the major brain cells from glycolytic inhibition and the
fuel for tumor energy metabolism through lac- brain from hypoglycemia.
tate fermentation, the restriction of glucose It is important to mention that the therapeu-
tic benefit for the KD is best when the diet is con-
sumed in moderately restricted amounts rather
8 5 than in unrestricted amounts. Blood glucose lev-
els are lower while ketone levels are higher when
Diet
4 the diet is consumed in restricted amounts than
Plasma [Ketones] (mM)

in unrestricted amounts (Seyfried et al., 2003;


Plasma [Glucose] (mM)

Initiation
4
Managed Growth 3 Zhou etal., 2007). Moreover, a prolonged unre-
Unmanaged stricted consumption of the KD can cause dyslip-
Growth
2
idemia, insulin resistance, and elevated glucose
2 levels. Moderate restriction of intake will reduce
[Glucose] these conditions (Meidenbauer etal., 2014). The
[Ketones] 1
mild body weight loss associated with the dietary
restriction also creates a healthier homeostatic
0
0 3 10 17 24 30
0.0 state. This contrasts with the weight loss associ-
Days of Treatment ated with radiation and chemotherapies, which
are toxic to the body. Hence, the KD-R can
FIGURE29.4: Relationship of circulating levels enhance general physiological health while plac-
of glucose and ketones (-hydroxybutyrate, ing metabolic stress on tumorcells.
-OHB) to tumor management. The glucose and When systemic glucose availability becomes
ketone values are within normal physiological ranges limiting, most normal cells of the body will tran-
under fasting conditions in humans and will produce sition their energy metabolism to fats and ketone
anti-angiogenic, anti-inflammatory, and pro-apoptotic bodies. Ketone bodies are generated almost exclu-
effects. We refer to this state as the zone of metabolic sively in liver hepatocytes largely from fatty acids
management. Metabolic stress will be greater in tumor of triglyceride origin during periods of fasting
cells than in normal cells when the whole body enters (Krebs etal., 1971; VanItallie and Nufert, 2003).
the metabolic zone. The values for blood glucose in mg/ Arestriction of total caloric intake will facilitate a
dl can be estimated by multiplying the mM values by reduction in blood glucose and insulin levels and
18. The glucose and ketone levels predicted for tumor an elevation in ketone bodies (-hydroxybutyrate
management in human cancer patients are 3.1 to 3.8 and acetoacetate). Most tumor cells are unable to
mM (5565 mg/dl) and 2.5 to 7.0 mM, respectively. use ketone bodies for energy due to abnormalities
These ketone levels are well below the levels associ- in mitochondria structure or function (Seyfried,
ated with ketoacidosis (blood ketone values greater 2012e; Seyfried et al., 2012). Ketone bodies can
than 15 mmol). Elevated ketones will sustain meta- also be toxic to some cancer cells (Maurer etal.,
bolic pressure on tumor cells, buffer daily fluctuations 2011; Skinner et al., 2009). Nutritional ketosis
in blood glucose levels, and protect the brain from induces metabolic stress on tumor tissue that
hypoglycemia. is selectively vulnerable to glucose deprivation
Modified from a previous version (Seyfried etal.,2008). (Seyfried et al., 2012). Hence, metabolic stress
560 Part IV:Homeostatic Therapies

will be greater in tumor cells than in normal cells are reduced (Cahill and Veech, 2003; Morris,
when the whole body is transitioned away from 2005; VanItallie and Nufert, 2003; Veech et al.,
glucose and to ketone bodies for energy. 2001). Ketone bodies have neuroprotective and
The metabolic shift from glucose metabo- anti-inflammatory action against a number of
lism to ketone body metabolism creates an anti- neurological and neurodegenerative diseases
angiogenic, anti-inflammatory, and pro-apoptotic (Maalouf et al., 2009; Seyfried, 2014). Ketone
environment within the tumor microenviron- body metabolism reduces oxygen free radicals
ment (Jiang and Wang, 2013; Mukherjee et al., while enhancing metabolic efficiency of normal
2004; Mukherjee et al., 2002; Mulrooney et al., cells (Milder and Patel, 2012; Stafford etal., 2010;
2011; Seyfried, 2012e; Seyfried et al., 2003). The Veech et al., 2001). It is also important to rec-
general concept of a survival advantage for tumor ognize that circulating ketone levels will rarely
cells over normal cells occurs when fermentable exceed 7 to 9mmol in most nondiabetic patients
fuels are abundant but not when they become since excess ketones will be excreted in the urine
limiting (Seyfried, 2012f). Figure 29.4 illustrates (Veech etal., 2001). Hence, ketones are consid-
the changes in whole-body levels of blood glu- ered good medicine for several neurological
cose and ketone bodies (hydroxybutyrate) and neurodegenerative diseases in patients with
that are predicted to metabolically stress tumor normal physiology (Cahill and Veech, 2003;
cells while enhancing the metabolic efficiency Freeman and Kossoff, 2010; Seyfried, 2014;
of normal cells. This therapeutic strategy was VanItallie and Nufert,2003).
illustrated previously in cancer patients and in The KD, in addition to lowering glucose
preclinical models (Champ et al., 2014; Chang availability to the tumor microenvironment,
etal., 2013; Fine etal., 2012; Nebeling etal., 1995; could potentially lower brain glutamine lev-
Simone et al., 2013; Stafford et al., 2010; Zhou els, thus restricting availability of this energy
etal., 2007; Zuccoli etal.,2010). metabolite for tumor growth (Bergman et al.,
2010; Yudkoff et al., 2007). Previous studies
THE CALORIE-RESTRICTED with refractory pediatric astrocytoma and adult
KETOGENIC DIET AS A GBM suggest that KDs are therapeutically effec-
N O N T O X I C M E TA B O L I C tive against malignant brain cancer (Nebeling
THER APY FORBR AIN CANCER etal., 1995; Zuccoli etal., 2010). The KD-R could
Emerging evidence suggests that metabolic be even more therapeutic if combined with
therapies using KDs that lower glucose levels nontoxic drugs that also target glycolysis (e.g.,
can help retard GBM growth in younger and 2-deoxyglucose or 3-bromopyruvate; Ko et al.,
older patients (Nebeling et al., 1995; Rieger 2004; Marsh et al., 2008b). The KD-R could
et al., 2014; Woolf and Scheck, 2015; Zuccoli also further enhance GBM survival when com-
et al., 2010). We showed that reduced intake of bined with the anti-HCMV drug, valganciclovir
either a high-carbohydrate diet or a high-fat, (Valcyte). HCMV infection of tumor cells can
low-carbohydrate KD had similar effects in contribute to elevated metabolism of glucose
reducing aggressive brain tumor growth in mice and glutamine (Yu et al., 2011). Recent evi-
(Zhou etal., 2007). The macronutrient composi- dence indicates that valganciclovir can enhance
tion of commercial KDs for tumor treatment has survival of GBM patients (Soderberg-Naucler
been described in detail (Abdelwahab etal., 2012; et al., 2013). However, this finding is contro-
Zhou etal., 2007). Restricted diets are those that versial, and further studies will be needed to
deliver fewer total calories in order to lower circu- verify these initial observations (Cobbs, 2014;
lating glucose and insulin levels (Seyfried etal., Solomon et al., 2014). On the other hand, we
2003). Moreover, the KD-R has some advantages think the KD-R could possibly reduce HCMV
over regular diet restriction as it delivers the load and activation state through an effect of
optimum metabolic stress to the cancer cells and viral energy metabolites.
protects normal cells by reducing the glucose Poff et al. (2014) also recently showed a
and elevating the ketone level further (Seyfried synergistic interaction between the KD and
etal., 2005). Nebeling and colleagues (1995) first hyperbaric oxygen therapy (HBO2T). The KD
showed that the KD was an effective nontoxic reduces glucose for glycolytic energy, while also
management for advanced stage astrocytoma reducing nicotinamide adenine dinucleotide
in children. Ketone bodies (-hydroxybutyrate phosphate-oxidase levels for anti-oxidant poten-
and acetoacetate) become an alternative fuel for tial through the pentose-phosphate-pathway.
brain energy metabolism when glucose levels HBO2T will increase ROS in the tumor cells
Malignant Brain Cancer Management with Metabolic Therapy 561

while the ketones will protect normal cells CONCLUSIONS AND HOPE
against ROS damage and from the potential for FORTHEFUTURE
central nervous system oxygen toxicity (Veech, As long as brain cancer is viewed as something
2004; Veech et al., 2001). Glucose deprivation other than a metabolic disease, it is unlikely
will enhance oxidative stress in tumor cells, that there will be major progress in improv-
while increased oxygen can reduce tumor cell ing progression-free survival for the major-
proliferation (Chen etal., 2009; Spitz etal., 2000). ity of those with the disease (Seyfried, 2012b).
In contrast to radiation therapy, which also kills The current standard of care for GBM offers lit-
tumor cells through ROS production (Harrison tle hope of improved quality of life or long-term
and Blackwell, 2004), the KD + HBO2T will patient survival (Lawrence et al., 2012). If GBM
kill tumor cells without causing toxic collateral becomes viewed as a metabolic disease, however,
damage to normal cells. Some KDs might also we might anticipate major advances in treatment
enhance the therapeutic action of radiation ther- and substantial enhancement of progression-free
apy against brain and lung tumors (Abdelwahab survival. Metabolic therapy targets the abnor-
etal., 2012; Allen etal., 2013). It will be impor- malities in tumor cell energy metabolism without
tant to compare and contrast the therapeutic causing toxicity. Unfortunately, there is a criti-
efficacy of conventional radiation therapy with cal lack of clinical trials using metabolic therapy
HBO2T when used with the KD-R. It would also as an alternative to the current standard of care.
be important to determine if HBO2T can even- Nevertheless, we remain hopeful that the meta-
tually replace radiation therapy for managing bolic approach to brain cancer management using
malignant brain cancer. the KD-R together with synergistic drugs and pos-
According to the evidence presented here, sibly HBO2T will offer the best chance for quality
the KD-R can represent a viable nontoxic option of life and longer term survival for GBM patients.
to the current standard of care for managing Metabolic therapy should become the new stand-
malignant brain cancer. The KD-R can tar- ard of care if the goal is to improve the overall sur-
get tumor cells globally without harming nor- vival and quality of life of GBM patients.
mal neurons and glia. The blood-brain barrier
is less of an issue with the KD-R therapy than ACK NOWLEDGMENTS
with conventional therapies. Although KD-R This work was supported in part from National
therapy could be a more rational approach to Institutes of Health grants (HD-39722, NS-55195,
malignant brain cancer management than is and CA-102135), a grant from the American
the current standard of cancer, the KD-R is not Institute of Cancer, and the Boston College
without some shortcomings. Compliance can ExpenseFund.
be a major obstacle in attempting to implement
References
the KD-R (Seyfried et al., 2012). Some people
can have difficulty in maintaining blood glu- Abdelwahab, M.G., Fenton, K.E., Preul, M.C., Rho,
J. M., Lynch, A., Stafford, P., and Scheck, A. C.
cose and ketones in the ranges needed to target
(2012). The ketogenic diet is an effective adjuvant
angiogenesis and to control tumor growth and
to radiation therapy for the treatment of malig-
inflammation. Considerable patient discipline
nant glioma. PLoS One 7, e36197.
and motivation is required for implementing
Alberghina, L., Gaglio, D., Moresco, R.M., Gilardi,
the KD-R as a therapy. Many neurooncolo-
M.C., Messa, C., and Vanoni, M. (2014). A sys-
gists are also unfamiliar with the principles
tems biology road map for the discovery of drugs
and concepts about how the therapy controls targeting cancer cell metabolism. Curr Pharm
tumor growth or how metabolic therapy can be Des 20, 26482666.
used as an alternative to the standard of care. Allen, B.G., Bhatia, S.K., Buatti, J.M., Brandt, K.E.,
Consequently, some patients might be discour- Lindholm, K. E., Button, A. M., Szweda, L. I.,
aged from using the KD-R. Nevertheless, we Smith, B.J., Spitz, D.R., and Fath, M.A. (2013).
remain hopeful that the metabolic approach Ketogenic diets enhance oxidative stress and
to brain cancer management using the KD-R radio-chemo-therapy responses in lung cancer
together with synergistic drugs, valganciclovir, xenografts. Clin Cancer Res 19, 39053913.
and possibly HBO2T will offer the best chance Arismendi-Morillo, G. (2009). Electron micros-
for quality of life and longer term survival for copy morphology of the mitochondrial network
GBM patients. Neurooncologists should test in human cancer. Int J Biochem Cell Biol 41,
this hypothesis. 20622068.
562 Part IV:Homeostatic Therapies

Arismendi-Morillo, G. (2011). Electron microscopy diagnosed malignant glioma. JAMA 293,


morphology of the mitochondrial network in 557564.
gliomas and their vascular microenvironment. Chen, R., Nishimura, M. C., Bumbaca, S. M.,
Biochim Biophys Acta 1807, 602608. Kharbanda, S., Forrest, W. F., Kasman, I. M.,
Arismendi-Morillo, G.J., and Castellano-Ramirez, Greve, J. M., Soriano, R. H., Gilmour, L. L.,
A. V. (2008). Ultrastructural mitochon- Rivers, C. S., et al. (2010). A hierarchy of
drial pathology in human astrocytic tumors: self-renewing tumor-initiating cell types in glio-
Potentials implications pro-therapeutics strat- blastoma. Cancer Cell 17, 362375.
egies. J Electron Microsc (Tokyo) 57,3339. Chen, Y., Cairns, R., Papandreou, I., Koong, A., and
Bayley, J.P., and Devilee, P. (2010). Warburg tumours Denko, N. C. (2009). Oxygen consumption can
and the mechanisms of mitochondrial tumour regulate the growth of tumors, a new perspective
suppressor genes. Barking up the right tree? Curr on the Warburg effect. PLoS One 4,e7033.
Opin Genet Dev 20, 324329. Chicco, A.J., and Sparagna, G.C. (2007). Role of car-
Bergman, C., Kashiwaya, Y., and Veech, R.L. (2010). diolipin alterations in mitochondrial dysfunc-
The effect of pH and free Mg2+ on ATP linked tion and disease. Am J Physiol Cell Physiol 292,
enzymes and the calculation of Gibbs free C3344.
energy of ATP hydrolysis. J Phys Chem B 114, Chinopoulos, C., Gerencser, A.A., Mandi, M., Mathe,
1613716146. K., Torocsik, B., Doczi, J., Turiak, L., Kiss, G.,
Bozidis, P., Williamson, C. D., Wong, D. S., and Konrad, C., Vajda, S., et al. (2010). Forward
Colberg-Poley, A.M. (2010). Trafficking of UL37 operation of adenine nucleotide translocase dur-
proteins into mitochondrion-associated mem- ing F0F1-ATPase reversal:Critical role of matrix
branes during permissive human cytomegalovi- substrate-level phosphorylation. FASEB J 24,
rus infection. J Virol 84, 78987903. 24052416.
Burton, T. M., and Dooren, J. C. (2011). Key FDA Claypool, S. M., and Koehler, C. M. (2012). The
approval yanked for avastin. Wall Street Journal. complexity of cardiolipin in health and disease.
November19. Trends Biochem Sci 37,3241.
Cahill, G.F., Jr., and Veech, R.L. (2003). Ketoacids? Cobbs, C. S. (2013). Cytomegalovirus and brain
Good medicine? Trans Am Clin Climatol Assoc tumor: Epidemiology, biology and therapeutic
114, 149161; discussion 162143. aspects. Curr Opin Oncol 25, 682688.
Carew, J. S., and Huang, P. (2002). Mitochondrial Cobbs, C. S. (2014). Does valganciclovir have a
defects in cancer. Mol Cancer1,9. role in glioblastoma therapy? Neuro Oncol 16,
Chambers, J. W., Maguire, T. G., and Alwine, J. C. 330331.
(2010). Glutamine metabolism is essential for Cogliati, S., Frezza, C., Soriano, M.E., Varanita, T.,
human cytomegalovirus infection. J Virol 84, Quintana-Cabrera, R., Corrado, M., Cipolat, S.,
18671873. Costa, V., Casarin, A., Gomes, L.C., etal. (2013).
Champ, C.E., and Klement, R.J. (2015). Commentary Mitochondrial cristae shape determines respira-
on Strong adverse prognostic impact of hyper- tory chain supercomplexes assembly and respira-
glycemic episodes during adjuvant chemo- tory efficiency. Cell 155, 160171.
radiotherapy of glioblastoma multiforme. Cooke, M. S., Evans, M. D., Dizdaroglu, M.,
Strahlenther Onkol. 191, 281282. and Lunec, J. (2003). Oxidative DNA dam-
Champ, C.E., Palmer, J.D., Volek, J.S., Werner-Wasik, age:Mechanisms, mutation, and disease. FASEB
M., Andrews, D.W., Evans, J.J., Glass, J., Kim, L., J 17, 11951214.
and Shi, W. (2014). Targeting metabolism with a Cuezva, J. M., Chen, G., Alonso, A. M., Isidoro, A.,
ketogenic diet during the treatment of glioblas- Misek, D. E., Hanash, S. M., and Beer, D. G.
toma multiforme. J Neurooncol 117, 125131. (2004). The bioenergetic signature of lung ade-
Chandra, D., and Singh, K. K. (2011). Genetic nocarcinomas is a molecular marker of cancer
insights into OXPHOS defect and its role in can- diagnosis and prognosis. Carcinogenesis 25,
cer. Biochim Biophys Acta 1807, 620625. 11571163.
Chang, H.T., Olson, L.K., and Schwartz, K.A. (2013). Cuezva, J. M., Krajewska, M., de Heredia, M. L.,
Ketolytic and glycolytic enzymatic expression Krajewski, S., Santamaria, G., Kim, H., Zapata,
profiles in malignant gliomas: Implication for J. M., Marusawa, H., Chamorro, M., and Reed,
ketogenic diet therapy. Nutr Metab 10,47. J. C. (2002). The bioenergetic signature of can-
Chang, S.M., Parney, I.F., Huang, W., Anderson, F.A ., cer:Amarker of tumor progression. Cancer Res
Jr., Asher, A.L., Bernstein, M., Lillehei, K.O., 62, 66746681.
Brem, H., Berger, M. S., and Laws, E. R. Davis, F. G., Freels, S., Grutsch, J., Barlas, S., and
(2005). Patterns of care for adults with newly Brem, S. (1998). Survival rates in patients with
Malignant Brain Cancer Management with Metabolic Therapy 563

primary malignant brain tumors stratified by glutamine metabolism is a function of the


patient age and tumor histological type: An alpha-ketoglutarate to citrate ratio in cells. Nat
analysis based on Surveillance, Epidemiology, Comm 4,2236.
and End Results (SEER) data, 19731991. J Ferreira, L. M. (2010). Cancer metabolism: The
Neurosurg 88,110. Warburg effect today. Exp Mol Pathol 89,
de Groot, J. F., Fuller, G., Kumar, A. J., Piao, Y., 372380.
Eterovic, K., Ji, Y., and Conrad, C. A. (2010). Fine, E. J., Segal-Isaacson, C. J., Feinman, R. D.,
Tumor invasion after treatment of glioblastoma Herszkopf, S., Romano, M. C., Tomuta, N.,
with bevacizumab:Radiographic and pathologic Bontempo, A.F., Negassa, A., and Sparano, J.A.
correlation in humans and mice. Neuro Oncol (2012). Targeting insulin inhibition as a meta-
12, 233242. bolic therapy in advanced cancer:Apilot safety
DeBerardinis, R. J., and Cheng, T. (2010). Qs and feasibility dietary trial in 10 patients.
next: The diverse functions of glutamine in Nutrition 28, 10281035.
metabolism, cell biology and cancer. Oncogene Fisher, P. G., and Buffler, P. A. (2005). Malignant
29, 313324. gliomas in 2005:Where to go from here? JAMA
Delsite, R. L., Rasmussen, L. J., Rasmussen, A. K., 293, 615617.
Kalen, A., Goswami, P. C., and Singh, K. K. Freeman, J. M., and Kossoff, E. H. (2010). Ketosis
(2003). Mitochondrial impairment is accompa- and the ketogenic diet, 2010:Advances in treat-
nied by impaired oxidative DNA repair in the ing epilepsy and other disorders. Adv Pediatr 57,
nucleus. Mutagenesis 18, 497503. 315329.
Derr, R.L ., Ye, X., Islas, M.U., Desideri, S., Saudek, C.D., Fry, M., and Green, D.E. (1981). Cardiolipin require-
and Grossman, S.A. (2009). Association between ment for electron transfer in complex Iand III of
hyperglycemia and survival in patients with the mitochondrial respiratory chain. J Biol Chem
newly diagnosed glioblastoma. J Clin Oncol 27, 256, 18741880.
10821086. Galluzzi, L., Morselli, E., Kepp, O., Vitale, I.,
Di Chiro, G., Oldfield, E., Wright, D.C., De Michele, D., Rigoni, A., Vacchelli, E., Michaud, M., Zischka,
Katz, D. A., Patronas, N. J., Doppman, J. L., H., Castedo, M., and Kroemer, G. (2010).
Larson, S. M., Ito, M., and Kufta, C. V. (1988). Mitochondrial gateways to cancer. Mol Aspects
Cerebral necrosis after radiotherapy and/or intra- Med 31,120.
arterial chemotherapy for brain tumors: PET Gao, P., Tchernyshyov, I., Chang, T.C., Lee, Y.S., Kita,
and neuropathologic studies. Am J Roentgenol K., Ochi, T., Zeller, K.I., De Marzo, A.M., Van
150, 189197. Eyk, J.E., Mendell, J.T., and Dang, C.V. (2009).
Drenick, E. J., Alvarez, L. C., Tamasi, G. C., and c-Myc suppression of miR-23a/b enhances mito-
Brickman, A.S. (1972). Resistance to symptom- chondrial glutaminase expression and glutamine
atic insulin reactions after fasting. J Clin Invest metabolism. Nature 458, 762765.
51, 27572762. Hans, P., Vanthuyne, A., Dewandre, P.Y., Brichant, J.F.,
Dziurzynski, K., Chang, S. M., Heimberger, A. B., and Bonhomme, V. (2006). Blood glucose con-
Kalejta, R.F., McGregor Dallas, S.R., Smit, M., centration profile after 10 mg dexamethasone
Soroceanu, L., and Cobbs, C.S. (2012). Consensus in non-diabetic and type 2 diabetic patients
on the role of human cytomegalovirus in glio- undergoing abdominal surgery. Br J Anaesth 97,
blastoma. Neuro Oncol 14, 246255. 164170.
Dziurzynski, K., Wei, J., Qiao, W., Hatiboglu, M.A., Harris, D., Barts, A., Connors, J., Dahl, M., Elliott, T.,
Kong, L.Y., Wu, A., Wang, Y., Cahill, D., Levine, N., Kong, J., Keane, T., Thompson, D., Stafford, S., Ur, E.,
Prabhu, S., etal. (2011). Glioma-associated cyto- and Sirrs, S. (2013). Glucocorticoid-induced
megalovirus mediates subversion of the mono- hyperglycemia is prevalent and unpredictable for
cyte lineage to a tumor propagating phenotype. patients undergoing cancer therapy: An obser-
Clin Cancer Res 17, 46424649. vational cohort study. Cur Onocol 20, e532538.
Elstrom, R.L ., Bauer, D.E ., Buzzai, M., Karnauskas, R., Harrison, L., and Blackwell, K. (2004). Hypoxia and
Harris, M.H., Plas, D.R ., Zhuang, H., Cinalli, R.M., anemia:Factors in decreased sensitivity to radia-
Alavi, A., Rudin, C. M., and Thompson, C. B. tion therapy and chemotherapy? Oncologist 9
(2004). Akt stimulates aerobic glycolysis in can- Suppl 5,3140.
cer cells. Cancer Res 64, 38923899. Hawkins, C., and Croul, S. (2011). Viruses and
Fendt, S.M., Bell, E.L ., Keibler, M.A., Olenchock, B.A., human brain tumors: Cytomegalovirus enters
Mayers, J. R., Wasylenko, T. M., Vokes, N. I., the fray. J Clin Invest 121, 38313833.
Guarente, L., Vander Heiden, M. G., and Hawkins, R.A. (2009). The blood-brain barrier and
Stephanopoulos, G. (2013). Reductive glutamate. Am J Clin Nutr 90, 867S874S.
564 Part IV:Homeostatic Therapies

Holdhoff, M., Ye, X., Blakeley, J.O., Blair, L., Burger, P.C., supporting the Warburg theory of cancer. J Lipid
Grossman, S. A., and Diaz, L. A., Jr. (2012). Res 49, 25452556.
Use of personalized molecular biomarkers in Kiebish, M.A., Han, X., Cheng, H., and Seyfried, T.N.
the clinical care of adults with glioblastomas. J (2009). In vitro growth environment produces
Neurooncol 110, 279285. lipidomic and electron transport chain abnor-
Huysentruyt, L. C., Akgoc, Z., and Seyfried, T. N. malities in mitochondria from non-tumorigenic
(2011). Hypothesis:Are neoplastic macrophages/ astrocytes and brain tumours. ASN Neuro 1,
microglia present in glioblastoma multiforme? e00011.
ASN Neuro 3, e00064. Ko,Y.H.,Smith,B.L.,Wang,Y.,Pomper,M.G.,Rini,D.A.,
Iglesias, P., Salas, A., and Costoya, J.A. (2012). The Torbenson, M.S., Hullihen, J., and Pedersen, P.L.
maintenance of mitochondrial genetic stability is (2004). Advanced cancers:Eradication in all cases
crucial during the oncogenic process. Commun using 3-bromopyruvate therapy to deplete ATP.
Integr Biol 5,3438. Biochem Biophys Res Commun 324, 269275.
Iwamoto, F. M., Abrey, L. E., Beal, K., Gutin, P. H., Koehler, P. J. (1995). Use of corticosteroids in
Rosenblum, M.K., Reuter, V.E., DeAngelis, L.M., neuro-oncology. Anticancer Drugs 6,1933.
and Lassman, A.B. (2009). Patterns of relapse and Koppenol, W. H., Bounds, P. L., and Dang, C. V.
prognosis after bevacizumab failure in recurrent (2011). Otto Warburgs contributions to current
glioblastoma. Neurology 73, 12001206. concepts of cancer metabolism. Nat Rev Cancer
James, A.D., Chan, A., Erice, O., Siriwardena, A.K., 11, 325337.
and Bruce, J.I. (2013). Glycolytic ATP fuels the Krebs, H. A., Williamson, D. H., Bates, M. W.,
plasma membrane calcium pump critical for Page, M.A., and Hawkins, R.A. (1971). The role
pancreatic cancer cell survival. J Biol Chem 288, of ketone bodies in caloric homeostasis. Adv
3600736019. Enzyme Reg 9, 387409.
Jeyaretna, D. S., Curry, W. T., Jr., Batchelor, T. T., Krex, D., Klink, B., Hartmann, C., von Deimling, A.,
Stemmer-Rachamimov, A., and Plotkin, S. R. Pietsch, T., Simon, M., Sabel, M., Steinbach, J.P.,
(2011). Exacerbation of cerebral radiation necro- Heese, O., Reifenberger, G., et al. (2007).
sis by bevacizumab. J Clin Oncol 29, e159e162. Long-term survival with glioblastoma multi-
Jiang, Y.S., and Wang, F.R. (2013). Caloric restric- forme. Brain 130, 25962606.
tion reduces edema and prolongs survival in a Kulawiec, M., Safina, A., Desouki, M. M., Still, I.,
mouse glioma model. J Neurooncol 114,2532. Matsui, S.I., Bakin, A., and Singh, K.K. (2008).
John, A. P. (2001). Dysfunctional mitochondria, Tumorigenic transformation of human breast
not oxygen insufficiency, cause cancer cells to epithelial cells induced by mitochondrial DNA
produce inordinate amounts of lactic acid: The depletion. Cancer Biol Ther 7, 17321743.
impact of this on the treatment of cancer. Med Lawrence, Y. R., Mishra, M. V., Werner-Wasik, M.,
Hypotheses 57, 429431. Andrews, D. W., Showalter, T. N., Glass, J.,
Johnson, B. E., Mazor, T., Hong, C., Barnes, M., Shen, X., Symon, Z., and Dicker, A. P. (2012).
Aihara, K., McLean, C. Y., Fouse, S. D., Improving prognosis of glioblastoma in the 21st
Yamamoto, S., Ueda, H., Tatsuno, K., et al. century: Who has benefited most? Cancer 118,
(2014). Mutational analysis reveals the origin and 42284234.
therapy-driven evolution of recurrent glioma. Lawrence, Y.R., Wang, M., Dicker, A.P., Andrews, D.,
Science 343, 189193. Curran, W.J., Jr., Michalski, J.M., Souhami, L.,
Kallenberg, K., Bock, H. C., Helms, G., Jung, K., Yung, W.K., and Mehta, M. (2011). Early toxic-
Wrede, A., Buhk, J. H., Giese, A., Frahm, J., ity predicts long-term survival in high-grade
Strik, H., Dechent, P., and Knauth, M. (2009). glioma. Br J Cancer 104, 13651371.
Untreated glioblastoma multiforme: Increased Lee, W.H., Sonntag, W.E., Mitschelen, M., Yan, H.,
myo-inositol and glutamine levels in the contra- and Lee, Y.W. (2010). Irradiation induces region-
lateral cerebral hemisphere at proton MR spec- ally specific alterations in pro-inflammatory
troscopy. Radiology 253, 805812. environments in rat brain. Int J Radiat Biol 86,
Kargiotis, O., Geka, A., Rao, J.S., and Kyritsis, A.P. 132144.
(2010). Effects of irradiation on tumor cell sur- Lewis, C., and Murdoch, C. (2005). Macrophage
vival, invasion and angiogenesis. J Neurooncol responses to hypoxia: Implications for tumor
100, 323338. progression and anti-cancer therapies. Am J
Kiebish, M. A., Han, X., Cheng, H., Chuang, J. H., Pathol 167, 627635.
and Seyfried, T.N. (2008). Cardiolipin and elec- Lichtor, T., and Dohrmann, G.J. (1986). Respiratory
tron transport chain abnormalities in mouse patterns in human brain tumors. Neurosurgery
brain tumor mitochondria: Lipidomic evidence 19, 896899.
Malignant Brain Cancer Management with Metabolic Therapy 565

Lopes, M.B. S., Vanbenberg, S.R ., and Scheithauer, B.W. Meidenbauer, J.J., Ta, N., and Seyfried, T.N. (2014).
(1993). The World Health Organization clas- Influence of a ketogenic diet, fish-oil, and calorie
sification of nervous system tumors in experi- restriction on plasma metabolites and lipids in
mental neuro-oncology. In: Molecular Genetics C57BL/6J mice. Nutr Metab 11,23.
of Nervous System Tumors, A. H. Levine and Michaelis, M., Doerr, H. W., and Cinatl, J. (2009).
H. H.Schmidek, eds. (NewYork:John Wiley & The story of human cytomegalovirus and can-
Sons), pp.136. cer: Increasing evidence and open questions.
Lu, J., Sharma, L.K., and Bai, Y. (2009). Implications Neoplasia 11,19.
of mitochondrial DNA mutations and mitochon- Milder, J., and Patel, M. (2012). Modulation of oxi-
drial dysfunction in tumorigenesis. Cell Res 19, dative stress and mitochondrial function by the
802815. ketogenic diet. Epilepsy Res 100, 295303.
Lukins, M. B., and Manninen, P. H. (2005). Monje, M. L., Vogel, H., Masek, M., Ligon, K. L.,
Hyperglycemia in patients administered dexa- Fisher, P.G., and Palmer, T.D. (2007). Impaired
methasone for craniotomy. Anesth Analg 100, human hippocampal neurogenesis after treat-
11291133. ment for central nervous system malignancies.
Maalouf, M., Rho, J.M., and Mattson, M.P. (2009). Ann Neurol 62, 515520.
The neuroprotective properties of calorie restric- Morantz, R.A., Wood, G.W., Foster, M., Clark, M.,
tion, the ketogenic diet, and ketone bodies. Brain and Gollahon, K. (1979). Macrophages in experi-
Res Rev 59, 293315. mental and human brain tumors:Part2. Studies
Marsh, J., Mukherjee, P., and Seyfried, T.N. (2008a). of the macrophage content of human brain
Akt-dependent proapoptotic effects of dietary tumors. J Neurosurg 50, 305311.
restriction on late-stage management of a phos- Morris, A.A. (2005). Cerebral ketone body metabo-
phatase and tensin homologue/tuberous sclero- lism. J Inherit Metab Dis 28, 109121.
sis complex 2-deficient mouse astrocytoma. Clin Mrugala, M.M. (2013). Advances and challenges in
Cancer Res 14, 77517762. the treatment of glioblastoma:Aclinicians per-
Marsh, J., Mukherjee, P., and Seyfried, T.N. (2008b). spective. Disc Med 15, 221230.
Drug/diet synergy for managing malignant Mukherjee, P., Abate, L. E., and Seyfried, T. N.
astrocytoma in mice: 2-deoxy-D-glucose and (2004). Antiangiogenic and proapoptotic effects
the restricted ketogenic diet. Nutr Metab of dietary restriction on experimental mouse
(Lond)5,33. and human brain tumors. Clin Cancer Res 10,
Mason, W.P., Maestro, R.D., Eisenstat, D., Forsyth, P., 56225629.
Fulton, D., Laperriere, N., Macdonald, D., Perry, Mukherjee, P., El-Abbadi, M. M., Kasperzyk, J. L.,
J., and Thiessen, B. (2007). Canadian recommen- Ranes, M.K., and Seyfried, T.N. (2002). Dietary
dations for the treatment of glioblastoma multi- restriction reduces angiogenesis and growth in
forme. Curr Oncol 14, 110117. an orthotopic mouse brain tumour model. Br J
Maurer, G.D., Brucker, D.P., Baehr, O., Harter, P.N., Cancer 86, 16151621.
Hattingen, E., Walenta, S., Mueller-Klieser, W., Mulrooney, T. J., Marsh, J., Urits, I., Seyfried,
Steinbach, J.P., and Rieger, J. (2011). Differential T. N., and Mukherjee, P. (2011). Influence of
utilization of ketone bodies by neurons and caloric restriction on constitutive expression of
glioma cell lines: A rationale for ketogenic diet NF-kappaB in an experimental mouse astrocy-
as experimental glioma therapy. BMC Cancer toma. PLoS One 6, e18085.
11,315. Munzarova, M., Rejthar, A., and Mechl, Z. (1991). Do
McGirt, M. J., Chaichana, K. L., Gathinji, M., some malignant melanoma cells share antigens
Attenello, F., Than, K., Ruiz, A.J., Olivi, A., and with the myeloid monocyte lineage? Neoplasma
Quinones-Hinojosa, A. (2008). Persistent outpa- 38, 401405.
tient hyperglycemia is independently associated Nebeling, L.C., Miraldi, F., Shurin, S.B., and Lerner,
with decreased survival after primary resection E. (1995). Effects of a ketogenic diet on tumor
of malignant brain astrocytomas. Neurosurgery metabolism and nutritional status in pediatric
63, 286291; discussion291. oncology patients: Two case reports. J Am Coll
McKenna, M. C., Gruetter, R., Sonnewald, U., Nutr 14, 202208.
Waagepetersen, H.S., and Schousboe, A. (2006). Newsholme, P., Lima, M. M., Procopio, J.,
Energy metabolism of the brain. In: Basic Pithon-Curi, T.C., Doi, S.Q., Bazotte, R.B., and
Neurochemistry: Molecular, Cellular, and Curi, R. (2003). Glutamine and glutamate as vital
Medical Aspects, G.J. Siegel, R.W. Albers, S.T. metabolites. Braz J Med Biol Res 36, 153163.
Bradey, and D.P. Price, eds. (NewYork:Elsevier Nishie, A., Ono, M., Shono, T., Fukushi, J., Otsubo,
Academic Press), pp. 531557. M., Onoue, H., Ito, Y., Inamura, T., Ikezaki, K.,
566 Part IV:Homeostatic Therapies

Fukui, M., etal. (1999). Macrophage infiltration synthetase is a phosphate target for the activa-
and heme oxygenase-1 expression correlate with tion of mitochondrial metabolism. Biochemistry
angiogenesis in human gliomas. Clin Cancer Res 48, 71407149.
5, 11071113. Phillips, J.P., Eremin, O., and Anderson, J.R. (1982).
Noch, E., and Khalili, K. (2009). Molecular mecha- Lymphoreticular cells in human brain tumours
nisms of necrosis in glioblastoma: The role of and in normal brain. Br J Cancer 45,6169.
glutamate excitotoxicity. Cancer Biol Ther 8, Poff, A.M., Ari, C., Arnold, P., Seyfried, T.N., and
17911797. DAgostino, D. P. (2014). Ketone supplementa-
Ohgaki, H., and Kleihues, P. (2009). Genetic altera- tion decreases tumor cell viability and prolongs
tions and signaling pathways in the evolution of survival of mice with metastatic cancer. Intl J
gliomas. Cancer Sci 100, 22352241. Cancer 135, 17111720.
Ordys, B.B., Launay, S., Deighton, R.F., McCulloch, J., Prestegarden, L., Svendsen, A., Wang, J., Sleire, L.,
and Whittle, I.R. (2010). The role of mitochon- Skaftnesmo, K.O., Bjerkvig, R., Yan, T., Askland, L.,
dria in glioma pathophysiology. Mol Neurobiol Persson, A., Sakariassen, P.O., and Enger, P.O.
42,6475. (2010). Glioma cell populations grouped by
Oudard, S., Arvelo, F., Miccoli, L., Apiou, F., different cell type markers drive brain tumor
Dutrillaux, A. M., Poisson, M., Dutrillaux, B., growth. Cancer Res 70, 42744279.
and Poupon, M.F. (1996). High glycolysis in gli- Preusser, M., de Ribaupierre, S., Wohrer, A., Erridge,
omas despite low hexokinase transcription and S.C., Hegi, M., Weller, M., and Stupp, R. (2011).
activity correlated to chromosome 10 loss. Br J Current concepts and management of glioblas-
Cancer 74, 839845. toma. Ann Neurol 70,921.
Oudard, S., Boitier, E., Miccoli, L., Rousset, S., Ramanathan, A., Wang, C., and Schreiber, S.L. (2005).
Dutrillaux, B., and Poupon, M. F. (1997). Perturbational profiling of a cell-line model of
Gliomas are driven by glycolysis: Putative roles tumorigenesis by using metabolic measurements.
of hexokinase, oxidative phosphorylation and Proc Natl Acad Sci USA 102, 59925997.
mitochondrial ultrastructure. Anticancer Res 17, Rasmussen, A. K., Chatterjee, A., Rasmussen, L. J.,
19031911. and Singh, K.K. (2003). Mitochondria-mediated
Oudard, S., Poirson, F., Miccoli, L., Bourgeois, Y., nuclear mutator phenotype in Saccharomyces
Vassault, A., Poisson, M., Magdelenat, H., cerevisiae. Nucleic Acids Res 31, 39093917.
Dutrillaux, B., and Poupon, M. F. (1995). Reardon, D. A., Desjardins, A., Peters, K.,
Mitochondria-bound hexokinase as target for ther- Gururangan, S., Sampson, J., Rich, J. N.,
apy of malignant gliomas. Int J Cancer 62, 216222. McLendon, R., Herndon, J.E., 2nd, Marcello, J.,
Parsons, D.W., Jones, S., Zhang, X., Lin, J.C., Leary, R.J., Threatt, S., et al. (2011). Phase II study of met-
Angenendt, P., Mankoo, P., Carter, H., Siu, I.M., ronomic chemotherapy with bevacizumab for
Gallia, G.L., etal. (2008). An integrated genomic recurrent glioblastoma after progression on bev-
analysis of human glioblastoma multiforme. acizumab therapy. J Neurooncol 103, 371379.
Science 321, 18071812. Renner, C., Asperger, A., Seyffarth, A.,
Patil, C.G., Yi, A., Elramsisy, A., Hu, J., Mukherjee, D., Meixensberger, J., Gebhardt, R., and Gaunitz, F.
Irvin, D.K., Yu, J.S., Bannykh, S.I., Black, K.L., (2010). Carnosine inhibits ATP production in
and Nuno, M. (2012). Prognosis of patients with cells from malignant glioma. Neurol Res 32,
multifocal glioblastoma:Acase-control study. J 101105.
Neurosurg 117, 705711. Rieger, J., Bahr, O., Maurer, G. D., Hattingen, E.,
Pawelek, J. M., and Chakraborty, A. K. (2008). The Franz, K., Brucker, D., Walenta, S., Kammerer,
cancer cellleukocyte fusion theory of metasta- U., Coy, J. F., Weller, M., and Steinbach, J. P.
sis. Adv Cancer Res 101, 397444. (2014). ERGO: A pilot study of ketogenic diet
Pedersen, P.L. (1978). Tumor mitochondria and the in recurrent glioblastoma. Intl J Onocol 44,
bioenergetics of cancer cells. Prog Exp Tumor 18431852.
Res 22, 190274. Roskelley, R.C., Mayer, N., Horwitt, B.N., and Salter,
Pedersen, P. L. (2007). Warburg, me and hexoki- W.T. (1943). Studies in cancer. VII. Enzyme defi-
nase 2: Multiple discoveries of key molecular ciency in human and experimental cancer. J Clin
events underlying one of cancers most common Invest 22, 743751.
phenotypes, the Warburg Effect, i.e., elevated Rubinstein, L. J. (1972). Tumors of the Central
glycolysis in the presence of oxygen. J Bioenerg Nervous System (Washington, DC: Armed
Biomembr 39, 211222. Forces Institute of Pathology).
Phillips, D., Aponte, A.M., French, S.A., Chess, D.J., Schwimmer, C., Lefebvre-Legendre, L., Rak, M.,
and Balaban, R. S. (2009). Succinyl-CoA Devin, A., Slonimski, P. P., di Rago, J. P., and
Malignant Brain Cancer Management with Metabolic Therapy 567

Rigoulet, M. (2005). Increasing mitochondrial disease: Implications for novel therapeutics.


substrate-level phosphorylation can rescue Carcinogenesis 35, 515527.
respiratory growth of an ATP synthase-deficient Seyfried, T. N., Flores, R., Poff, A. M., DAgostino,
yeast. J Biol Chem 280, 3075130759. D. P., and Mukherjee, P. (2014b). Metabolic
Seoane, M., Mosquera-Miguel, A., Gonzalez, T., therapy: A new paradigm for managing malig-
Fraga, M., Salas, A., and Costoya, J. A. (2011). nant brain cancer. Cancer Letters 356, 2 Pt A,
The mitochondrial genome is a genetic sanctu- 289300.
ary during the oncogenic process. PLoS One 6, Seyfried, T. N., and Huysentruyt, L. C. (2013). On
e23327. the origin of cancer metastasis. Crit Rev Onocog
Seyfried, T.N. (2001). Perspectives on brain tumor 18,4373.
formation involving macrophages, glia, and neu- Seyfried, T. N., Kiebish, M., Mukherjee, P., and
ral stem cells. Perspect Biol Med 44, 263282. Marsh, J. (2008). Targeting energy metabolism in
Seyfried, T. N. (2012a). Cancer as a Metabolic brain cancer with calorically restricted ketogenic
Disease: On the Origin, Management, and diets. Epilepsia 49 Suppl 8, 114116.
Prevention of Cancer (Hoboken, NJ:John Wiley & Seyfried, T.N., Marsh, J., Shelton, L.M., Huysentruyt,
Sons). L.C., and Mukherjee, P. (2012). Is the restricted
Seyfried, T. N. (2012b). Cancer treatment strate- ketogenic diet a viable alternative to the standard
gies. In:Cancer as a Metabolic Disease:On the of care for managing malignant brain cancer?
Origin, Management, and Prevention of Cancer Epilepsy Res 100, 310326.
(Hoboken, NJ:John Wiley & Sons), pp. 227289. Seyfried, T.N., and Mukherjee, P. (2005). Targeting
Seyfried, T. N. (2012c). Genes, respiration, viruses, energy metabolism in brain cancer:Review and
and cancer. In:Cancer as a Metabolic Disease:On hypothesis. Nutr Metab (Lond)2,30.
the Origin, Management, and Prevention of Seyfried, T.N., Mukherjee, P., Adams, E., Mulrooney,
Cancer (Hoboken, NJ: John Wiley & Sons), pp. T.J., and Abate, L.E. (2005). Metabolic control of
145176. brain cancer:Role of glucose and ketones. Proc
Seyfried, T.N. (2012d). Is mitochondrial glutamine Amer Assoc Cancer Res 46,267.
fermentation a missing link in the metabolic the- Seyfried, T. N., Mukherjee, P., Kalamian, M., and
ory of cancer? In:Cancer as a Metabolic Disease: Zuccoli, G. (2011). The restricted ketogenic
On the Origin, Management, and Prevention of diet: An alternative treatment strategy for glio-
Cancer (Hoboken, NJ: John Wiley & Sons), pp. blastoma multiforme. In: Treatment Stratagies
133144. Oncology, R. Holcroft, ed. (London:Cambridge
Seyfried, T. N. (2012e). Metabolic management of Rse. Ctr.), pp.2435.
cancer. In:Cancer as a Metabolic Disease:On the Seyfried, T.N., Sanderson, T.M., El-Abbadi, M.M.,
Origin, Management, and Prevention of Cancer McGowan, R., and Mukherjee, P. (2003). Role of
(Hoboken, NJ:John Wiley & Sons), pp. 291354. glucose and ketone bodies in the metabolic con-
Seyfried, T. N. (2012f). Nothing in cancer biol- trol of experimental brain cancer. Br J Cancer 89,
ogy makes sense except in the light of evolu- 13751382.
tion. In:Cancer as a Metabolic Disease:On the Seyfried, T.N., and Shelton, L.M. (2010). Cancer as a
Origin, Management, and Prevention of Cancer metabolic disease. Nutr Metab (Lond)7,7.
(Hoboken, NJ:John Wiley & Sons), pp. 261275. Seyfried, T. N., Shelton, L. M., and Mukherjee, P.
Seyfried, T. N. (2012g). Respiratory dysfunction in (2010). Does the existing standard of care
cancer cells. In Cancer as a Metabolic Disease: increase glioblastoma energy metabolism?
On the Origin, Management, and Prevention Lancet Oncol 11, 811813.
of Cancer (Hoboken, NJ: John Wiley & Shinonaga, M., Chang, C. C., Suzuki, N., Sato, M.,
Sons), pp. 73105. and Kuwabara, T. (1988). Immunohistological
Seyfried, T. N. (2012h). The Warburg dispute. In: evaluation of macrophage infiltrates in brain
Cancer as a Metabolic Disease: On the Origin, tumors. Correlation with peritumoral edema. J
Management, and Prevention of Cancer, Neurosurg 68, 259265.
(Hoboken, NJ:John Wiley & Sons), pp. 107117. Simone, B.A., Champ, C.E., Rosenberg, A.L., Berger,
Seyfried, T.N. (2014). Ketone strong:Emerging evi- A. C., Monti, D. A., Dicker, A. P., and Simone,
dence for a therapeutic role of ketone bodies in N. L. (2013). Selectively starving cancer cells
neurological and neurodegenerative diseases. J through dietary manipulation: Methods and
Lipid Res 55, 18151817. clinical implications. Future Oncol 9, 959976.
Seyfried, T. N., Flores, R. E., Poff, A. M., and Skinner, R., Trujillo, A., Ma, X., and Beierle, E. A.
DAgostino, D.P. (2014a). Cancer as a metabolic (2009). Ketone bodies inhibit the viability of
568 Part IV:Homeostatic Therapies

human neuroblastoma cells. J Pediatr Surg 44, impacting volumetric white matter changes
212216; discussion216. following whole brain radiation therapy. J
Smiraglia, D.J., Kulawiec, M., Bistulfi, G.L., Gupta, Neurooncol 103, 111119.
S. G., and Singh, K. K. (2008). A novel role for Takano, T., Lin, J.H., Arcuino, G., Gao, Q., Yang, J.,
mitochondria in regulating epigenetic modi- and Nedergaard, M. (2001). Glutamate release
fication in the nucleus. Cancer Biol Ther 7, promotes growth of malignant gliomas. Nat Med
11821190. 7, 10101015.
Soderberg-Naucler, C., Rahbar, A., and Stragliotto, G. Talacchi, A., Turazzi, S., Locatelli, F., Sala, F.,
(2013). Survival in patients with glioblastoma Beltramello, A., Alessandrini, F., Manganotti, P.,
receiving valganciclovir. N Engl J Med 369, Lanteri, P., Gambin, R., Ganau, M., etal. (2010).
985986. Surgical treatment of high-grade gliomas in
Solomon, I.H., Ramkissoon, S.H., Milner, D.A., Jr., motor areas:The impact of different supportive
and Folkerth, R.D. (2014). Cytomegalovirus and technologies:A171-patient series. J Neurooncol
glioblastoma:Areview of evidence for their asso- 100, 417426.
ciation and indications for testing and treatment. Tso, C.L ., Shintaku, P., Chen, J., Liu, Q., Liu, J., Chen, Z.,
J Neuropathol Exp Neurol 73, 994998. Yoshimoto, K., Mischel, P. S., Cloughesy, T. F.,
Souhami, L., Seiferheld, W., Brachman, D., Liau, L. M., and Nelson, S. F. (2006). Primary
Podgorsak, E.B., Werner-Wasik, M., Lustig, R., glioblastomas express mesenchymal stem-like
Schultz, C.J., Sause, W., Okunieff, P., Buckner, J., properties. Mol Cancer Res 4, 607619.
et al. (2004). Randomized comparison of ste- VanItallie, T. B., and Nufert, T. H. (2003).
reotactic radiosurgery followed by conventional Ketones: Metabolisms ugly duckling. Nutr Rev
radiotherapy with carmustine to conventional 61, 327341.
radiotherapy with carmustine for patients with Veatch, J.R., McMurray, M.A., Nelson, Z.W., and
glioblastoma multiforme: Report of Radiation Gottschling, D. E. (2009). Mitochondrial dys-
Therapy Oncology Group 9305 protocol. Int J function leads to nuclear genome instability via
Radiat Oncol Biol Phys 60, 853860. an iron-sulfur cluster defect. Cell 137, 12471258.
Spitz, D.R., Sim, J.E., Ridnour, L.A., Galoforo, S.S., Veech, R.L. (2004). The therapeutic implications of
and Lee, Y.J. (2000). Glucose deprivation-induced ketone bodies: The effects of ketone bodies in
oxidative stress in human tumor cells:Afunda- pathological conditions:Ketosis, ketogenic diet,
mental defect in metabolism? Ann NY Acad Sci redox states, insulin resistance, and mitochon-
899, 349362. drial metabolism. Prostaglandins Leukot Essent
Stafford, P., Abdelwahab, M. G., Kim do, Y., Preul, Fatty Acids 70, 309319.
M.C., Rho, J.M., and Scheck, A.C. (2010). The Veech, R.L., Chance, B., Kashiwaya, Y., Lardy, H.A.,
ketogenic diet reverses gene expression patterns and Cahill, G.F., Jr. (2001). Ketone bodies, poten-
and reduces reactive oxygen species levels when tial therapeutic uses. IUBMB Life 51, 241247.
used as an adjuvant therapy for glioma. Nutr Villalobo, A., and Lehninger, A.L. (1979). The proton
Metab (Lond)7,74. stoichiometry of electron transport in Ehrlich
Stroud, D. A., and Ryan, M. T. (2013). Mitochondria: ascites tumor mitochondria. J Biol Chem 254,
Organization of respiratory chain complexes 43524358.
becomes cristae-lized. Curr Biol 23, R969R971. Warburg, O. (1931). The Metabolism of Tumours
Stupp, R., Hegi, M.E ., Mason, W.P., van den Bent, M.J., (NewYork:Richard R.Smith).
Taphoorn, M. J., Janzer, R. C., Ludwin, S. K., Warburg, O. (1956a). On the origin of cancer cells.
Allgeier, A., Fisher, B., Belanger, K., etal. (2009). Science 123, 309314.
Effects of radiotherapy with concomitant and Warburg, O. (1956b). On the respiratory impairment
adjuvant temozolomide versus radiotherapy in cancer cells. Science 124, 269270.
alone on survival in glioblastoma in a ran- Warburg, O. (1969). Revidsed Lindau lec-
domised phase III study: 5-year analysis of the tures: The prime cause of cancer and
EORTC-NCIC trial. Lancet Oncol 10, 459466. preventionParts 1 & 2. In Meeting of the
Stupp, R., Mason, W.P., van den Bent, M.J., Weller, M., Nobel-Laureates D. Burk, ed. (Lindau, Lake
Fisher, B., Taphoorn, M.J., Belanger, K., Brandes, Constance, Germany:K.Triltsch).
A. A., Marosi, C., Bogdahn, U., et al. (2005). Williamson, C.D., and Colberg-Poley, A.M. (2009).
Radiotherapy plus concomitant and adjuvant Access of viral proteins to mitochondria via
temozolomide for glioblastoma. N Engl J Med mitochondria-associated membranes. Rev Med
352, 987996. Virol 19, 147164.
Szerlip, N., Rutter, C., Ram, N., Yovino, S., Kwok, Y., Wise, D.R., DeBerardinis, R.J., Mancuso, A., Sayed,
Maggio, W., and Regine, W. F. (2011). Factors N., Zhang, X. Y., Pfeiffer, H. K., Nissim, I.,
Malignant Brain Cancer Management with Metabolic Therapy 569

Daikhin, E., Yudkoff, M., McMahon, S. B., Yudkoff, M., Daikhin, Y., Melo, T. M., Nissim, I.,
and Thompson, C. B. (2008). Myc regulates a Sonnewald, U., and Nissim, I. (2007). The keto-
transcriptional program that stimulates mito- genic diet and brain metabolism of amino
chondrial glutaminolysis and leads to gluta- acids: Relationship to the anticonvulsant effect.
mine addiction. Proc Natl Acad Sci USA 105, Annu Rev Nutr 27, 415430.
1878218787. Yuneva, M. (2008). Finding an Achilles heel of
Woolf, E.C., and Scheck, A.C. (2015). The ketogenic cancer:The role of glucose and glutamine metab-
diet for the treatment of malignant glioma. J olism in the survival of transformed cells. Cell
Lipid Res. 56,510. Cycle 7, 20832089.
Xu, R. H., Pelicano, H., Zhou, Y., Carew, J. S., Zhou, W., Mukherjee, P., Kiebish, M. A., Markis,
Feng, L., Bhalla, K. N., Keating, M. J., and W. T., Mantis, J. G., and Seyfried, T. N. (2007).
Huang, P. (2005). Inhibition of glycolysis in The calorically restricted ketogenic diet, an effec-
cancer cells: A novel strategy to overcome tive alternative therapy for malignant brain can-
drug resistance associated with mitochondrial cer. Nutr Metab (Lond)4,5.
respiratory defect and hypoxia. Cancer Res 65, Zhuang, W., Qin, Z., and Liang, Z. (2009). The role of
613621. autophagy in sensitizing malignant glioma cells
Yang, D., Wang, M.T., Tang, Y., Chen, Y., Jiang, H., to radiation therapy. Acta Biochim Biophys Sin
Jones, T.T., Rao, K., Brewer, G.J., Singh, K.K., (Shanghai) 41, 341351.
and Nie, D. (2010). Impairment of mitochon- Zu, X.L., and Guppy, M. (2004). Cancer metabolism:
drial respiration in mouse fibroblasts by Facts, fantasy, and fiction. Biochem Biophys Res
oncogenic H-RAS(Q61L). Cancer Biol Ther 9, Commun 313, 459465.
122133. Zuccoli, G., Marcello, N., Pisanello, A., Servadei, F.,
Yu, Y., Clippinger, A.J., Pierciey, F.J., Jr., and Alwine, J.C. Vaccaro, S., Mukherjee, P., and Seyfried, T. N.
(2011). Viruses and metabolism: Alterations (2010). Metabolic management of glioblastoma
of glucose and glutamine metabolism medi- multiforme using standard therapy together
ated by human cytomegalovirus. AdvVirus Res with a restricted ketogenic diet: Case report.
80,4967. Nutr Metab (Lond)7,33.
30
Obesityand Diabetes
Nature, Nurture, andBeyond

CHARLESMOBBS

OBESITY AND stored resources at night (nocturnal animals, of


DIABETESEPIDEMIOLOGY course, do the inverse but the logic is the same).
A N D H E A LT H C O N S E Q U E N C E S Thus almost all organisms store bioenergetic
Biological evolution is largely the history of opti- potential when nutritional resources are in excess
mizing extraction of bioavailable energy from of current need (usually during the day), then use
the environment, since energy availability (or the stored form when environmental resources
the lethal extraction of those resources by preda- are inadequate for current need (usually at night).
tors) has been a rate-limiting factor for survival. Although this minimal rhythm of storage and use
Akey characteristic of the bioenergenetic econ- of stored resources is dictated by the daily cycle of
omy is the flexibility to adapt to the availability sunlight, organisms must also adapt to the possi-
of nutritional resources. Notably, this includes bility of longer periods of nutritional deficit (e.g.,
the capacity to use alternate substrates to pro- during winter or drought). Thus energy resources
duce adenosine triphosphate (ATP), depending must be stored in excess to those needed during
on availability. Since glucose produced from the nonacquisition phase of theday.
photosynthesis is the ultimate source of all bio- On the other hand, other constraints limit
energetics potential, most cells and organisms the optimum degree of nutritional stores. For
use glucose preferentially to make ATP when it animals, excess storage of nutrients in prey spe-
is available. When it is less available than other cies would reduce mobility to escape predation,
substrates, cells and organisms quickly adapt whereas excess nutrients in predators would
to metabolize those substrates. A classic exam- reduce mobility for predation (though in the
ple of this flexibility is the lac operon, which is latter case this might be self-limiting). Mobility
activated to metabolize lactose when it is present is also required for many other functions in
in the absence of glucose, and which, however, animals, including migration for reproduction
is tellingly inhibited when glucose is present (especially in birds and insects). Thus for animals
(Lewis, 2005). As described in this chapter, such homeostatic mechanisms have evolved to main-
metabolic flexibility may be relevant to obesity tain an optimum balance between storing nutri-
on low-carbohydrate diets (Brehm et al., 2003; tional resources for use during nutritional deficit
Kennedy etal.,2007). and maintaining mobility.
In addition to adapting to the presence of Several lines of evidence suggest that adipos-
alternate sources of bioenergetic potential, cells ity is highly regulated. One such line of evidence
and organisms must also be able to survive in is the relative stability of body weight:although
the absence of a continuous intake of nutritional most people can lose weight by limiting caloric
resources. Of particular relevance is the daily intake, almost all of this weight is gained back
cycle of sunlight that drives the daily cycle of met- over the next year (Davis et al., 2009). Of note
abolic economy. Aclassic example is the adapta- is that the weight gained back is almost exactly
tion of plants to the dark, since the production of the weight lost, indicating that total adiposity is
bioenergetics potential via photosynthesis ceases highly regulated (Davis et al., 2009). Similarly,
without sunlight. However, diurnal animals gen- after removal of adipose tissue by lipectomy, adi-
erally follow the same metabolic logic, acquiring posity is eventually restored to the original level
nutritional resources during the day and using with remarkable precision (Lopez et al., 2007).
Malignant Brain Cancer Management with Metabolic Therapy 571

In fact these and similar observations moti- amputations. More important, interventional
vated the lipostatic hypothesis, that adiposity studies have clearly demonstrated that interven-
is defended, such that perturbation by either tions leading to weight loss substantially reduce
overfeeding or underfeeding is quickly followed the risk of a variety of pathologies, notably diabe-
by homeostatic responses to return to the initial tes, and interventions that reduce blood glucose
weight (Lopez etal.,2007). similarly substantially reduce the risk of dia-
Although it has been argued that, for humans betic complications. In view of the major health
at least, homeostatic mechanisms to maintain threat represented by the dramatic increase in
nutritional resources are more important than the prevalence of obesity in the past 20years, it
mechanisms to prevent excess storage, this is is naturally of great interest to understand the
unconvincing, since human survival has his- physiological mechanisms regulating adipos-
torically also required a high degree of mobil- ity in humans. Of particular interest, in view of
ity, possibly more than most species (Pijl, 2011). the observation that adipose levels are typically
Nevertheless, nutritional deficiency has arguably defended, it is of great interest to understand
been the major cause of human mortality and why average adiposity has robustly increased in
morbidity throughout history (given the evidence humans in the past 20years.
that malnutrition impairs immune responses to
infectious diseases). Certainly the specter of mal- ROLE OFTHE VENTROMEDIAL
nutrition has framed the distinctly pessimistic HYPOTHALAMUS INENERGY
Malthusean view of human progress, a view that BALANCE AND GLUCOSE
continues to exert robust traction eventoday. H O M E O S TA S I S
Thus from a historical perspective it is shock- The first clue to the mechanisms regulating adi-
ing that in the waning of the 20th century, which posity actually came from clinical observations.
saw so many triumphs of public health, includ- In these cases, published around 1900 (Bray,
ing antibiotics, public hygiene, and the green 1993), it was reported that patients with tumors
revolution, the major threat to global health was in the hypothalamic/pituitary area exhibited a
the phenomenon of excess storage of nutritional syndrome of obesity and sterility. These obser-
resources, apparently overriding homeostatic vations remained anecdotal until a classic study
mechanisms evolved to prevent precisely this published in 1940 using the relatively recently
phenomenon. Nevertheless, it is now clear that invented stereotactic instrument (Hetherington
the worldwide epidemic in obesity and associ- & Ranson, 1940)]. This study, which used a stere-
ated disorders is a major threat to global health otaxic instrument to guide electrode placement,
(James, 2008) and plausibly will decrease life allowing electrolytic lesion of a specific and veri-
expectancy in the US population for the first fiable lesion in the brain, was among the first to
time in history (Olshansky etal., 2005). Indeed, demonstrate the function of a specific part of
it is becoming increasingly clear that obesity is the in the control of a physiological function.
a major risk factor for a remarkably broad range Specifically, lesions targeted to the ventromedial
of diseases and thus will constitute an increasing nucleus of the hypothalamus in rats (but prob-
burden on public finances (Daviglus etal., 2003; ably always lesioning adjacent arcuate nuclear
Fontaine et al., 2003; Oster et al., 1999; Peeters areas as well) produced profoundly increased
etal., 2003; Thompson etal., 1999). The main dis- adiposity relative to sham-lesioned rats. The
ease for which obesity is a major risk factor is type increased adiposity was associated with increased
2 diabetes (Overweight, obesity, and health risk, food intake. These studies were repeated in many
2000), although this perhaps begs the question of species many times over many years with reli-
in what sense type 2 diabetes is a disease. Thus, ably the same results. A particularly relevant
in themselves, elevated adiposity and elevated variation on these studies was to pair-feed the
blood glucose would plausibly be protective, in lesioned rats so that their food intake was the
the sense that their opposites are lethal. Certainly same as (nonobese) sham-lesioned rats. These
epidemiological studies have overwhelmingly pair-fed rats still became obese, only more slowly
confirmed that elevated blood glucose (and its (Hetherington & Ranson, 1940). Thus it became
marker hemogloblin A1c) are major risk fac- clear that neurons in the area of the ventrome-
tors for a wide variety of pathologies, especially dial hypothalamus (VHM) control not only food
kidney failure, retinopathy leading to blindness, intake but also, apparently, metabolic rate as well.
and peripheral neuropathy leading to a wide Subsequent studies demonstrated that lesions in
variety of pathological consequences including this area of the hypothalamus, especially if they
572 Part IV:Homeostatic Therapies

included the area ventral to the ventromedial area between the ventromedial nucleus and the
nucleus (i.e., the arcuate nucleus) produced not arcuate nucleus (Bergen et al., 1998). Of par-
only sterility (as was observed in humans) but ticular interest, in some mouse strains the GTG
also loss of function of all neuroendocrine sys- lesion produces not only obesity but diabetes as
tems, including glucocorticoid secretion (the well (Matsuo & Shino, 1972). It is not clear why
hypothalamus-pituitary-adrenal axis), growth GTG specifically targets neurons in the ventro-
hormone secretion, and thyroid secretion, simi- medial nucleus (although it was subsequently
lar to effects of fasting (Ahima etal.,1996). shown to also target neurons in the nucleus
An even more informative variation on the of the solitary tract/area postrema [Powley &
VMH lesion studies were the remarkable stud- Prechtl, 1986]). However, GTG did not produce
ies by Hervey, who connected rats given VMH lesions in diabetic mice (Debons etal., 1974), sug-
lesions to normal rats using parabiosis (Hervey, gesting that GTG may target neurons specifically
1959). Remarkably, the rats with the VMH lesions adapted to sense glucose as part of their function
gained weight, as expected, but the control rats to sense and regulate energy balance and glucose
lost almost all fat and frequently starved to death homeostasis. As described later, these observa-
(Hervey, 1959). These studies were the first to tions supported and motivated the glucostat
clearly suggest that adiposity is controlled by a hypothesis (Mayer, 1953) and subsequent dis-
circulating factor acting on hypothalamic neu- covery of glucose-regulated hypothalamic neu-
rons. As described later, this hypothesis has been rons (Oomura etal., 1969). Because of these and
largely confirmed by genetic analysis, although later results described later, the VMH has been a
many questions remain. major focus of studies investigating the causes of
Sadly, the original clinical observations that obesity (Yi, Scherer, & Tschop, 2011). Specifically,
hypothalamic tumors in humans are often asso- a major hypothesis animating the field has been
ciated with obesity have been robustly replicated. that impairments of neurons in the VMH (with
Aparticularly common lesion of this type occurs later emphasis specifically, including the arcuate
in children, generally in the form of hypotha- nucleus) to properly sense (and thus to underes-
lamic craniopharyngioma (Brabant et al., 1996; timate) signals of nutritional sufficiency may be a
Roth etal., 1998), a nonmalignant, slow-growing major contributor to obesity and diabetes (King,
tumor that nevertheless can cause a range of 2006; Myers & Olson, 2012). As described later,
debilitating symptoms, including hyperphagia this hypothesis has been in many ways supported
and obesity. In some cases, the hyperphagia may and still animates much research in the field,
resolve after resection of the tumor, but in oth- although other brain areas are increasingly of
ers hyperphagia and obesity may be caused by interest (Myers & Olson,2012).
the treatment to remove the craniopharyngioma. A similar protocol, neonatal injection with
In any case, these fortunately rare cases substan- monosodium glutamate, produces a lesion
tially corroborate the unique role of the hypo- somewhat similar to the that produced by GTG,
thalamus in regulating human energy balance. although even more ventral, completely destroy-
A pharmacological method to produce obe- ing the arcuate nucleus while damaging only a part
sity by hypothalamic lesion was discovered ser- of the ventromedial nucleus in mice (Bergen etal.,
endipitously in the course of toxicity screening 1998). Interestingly, however, in Chinese hamsters
of the compound gold-thioglucose (GTG), which this protocol produces robust damage to both the
was being successfully developed as a treat- arcuate nucleus and the ventromedial nucleus and
ment for rheumatoid arthritis. During toxicity causes diabetes in the absence of obesity (Komeda,
screening it was observed that after i.p. injec- Yokote, & Oki, 1980). This was an early indication
tion of GTG at doses near but not quite LD50, that this area of the brain controls glucose metab-
many mice became obese after treatment (Owen, olism as well as energy balance, a concept that was
Parson, & Crispell, 1953). When this observation subsequently amply corroborated.
was made, it was clear that hypothalamic lesions Following on these and related observa-
cause obesity in a range of species, so investiga- tions, particularly observations supporting the
tors assessed if the obesity produced by GTG was glucostat hypothesis, several studies from the
associated with hypothalamic lesions. In fact i.p. Sherwin group demonstrated that inhibiting
injection of GTG reliably lesions the ventrome- glucose metabolism in the ventromedial nucleus
dial nucleus of the hypothalamus (Mori & Perry, rapidly stimulated counterregulatory responses
1967), although careful analysis demonstrated that increase blood glucose whereas local infu-
that the center of the lesion was in the cell-poor sion of glucose or lactate in the presence of
Malignant Brain Cancer Management with Metabolic Therapy 573

systemic hypoglycemia prevented counter- their idiosyncratic dependence on genetic back-


regulatory responses (Borg et al., 1994; 1997; ground led to much confusion in the course of
1999; 2003). These studies clearly demonstrated these early studies. On the C57Bl/6J background,
glucose-sensing neurons in the VMH play a however, the obesity associated with the agouti
major role in peripheral glucose homeostasis. gene was quite robust and entailed hyperphagia.
As with VMH lesions, however, even when agouti
GENETIC BASIS OFOBESITY mice were pair-fed with wild-type mice, they
AND DIABETES INMICE became obese, although at a slower rate. These
Throughout the 20th century and even more so observations clearly suggested a potentially
in the 21st century, researchers have sought the hypothalamic locus for the obesity-producing
causes of disease by genetic analysis. They have effects of the agouti gene, but the link between
also used genetics to clarify mechanisms causing coat color and energy balance was completely
obesity (although, as described earlier, hypotha- obscure for manyyears.
lamic lesions caused by environmental pertur- Two other single genes causing obesity, though
bations have guided much research). Anecdotal segregating as a Mendelian single recessive gene,
observation certainly supports that obesity runs were discovered through a completely differ-
in families, and more rigorous research compar- ent method. Jackson Laboratories has been the
ing identical to fraternal twins has consistently largest breeder of mice for scientific research for
suggested that body mass index is roughly 50% much of the 20th century. As part of its program
heritable (Hjelmborg etal., 2008; Maes, Neale, & it has instituted a systematic screening program
Eaves, 1997). Until recently, however, tools were of each mouse for visible phenotypes, the purpose
not available to isolate the specific genes involved of which was to discover spontaneous mutations.
in human obesity. The program was inspired by the highly produc-
However, tools for the genetic analysis of tive studies of Morgan and colleagues study-
complex traits, and the availability of single gene ing induced mutations in Drosophila (Morgan,
polymorphisms determining these traits, were 1911). This program has produced some of the
developed much earlier for mice than humans. most informative mutations (leading ultimately
Mice have long been the species of choice for to the discovery of the relevant genes) in biomed-
genetic studies in mammals because for hun- ical research. One of the genes, initially called the
dreds, if not thousands, of years, mice have been obese or ob gene, was originally discovered in a
the subject of recreational breeding by mouse complex cross, but eventually the gene was bred
fanciers, who generally bred for esthetically to the standard C57Bl/6J background (Coleman
pleasing (cute) traits, including hair color. & Hummel, 1973). Homozygous ob/ob mice on
One such mouse variant was the yellow or agouti the C57Bl/6J were robustly obese and hyper-
mouse, selected for its esthetically pleasing coat phagic; as with VMH lesions and with the agouti
color. This coat color was determined by a single mutation, pair-fed ob/ob mice still became obese
gene that segregated as a single Mendelian domi- but at a slower rate (Coleman & Hummel, 1973).
nant gene, suspected as early at 1927, although Furthermore, ob/ob mice exhibited a wide range
rigorous analysis of one such prominent gene, of phenotypes, including sterility, suggesting that
called yellow lethal, did not begin until about the gene involved was involved in a wide range
1950 (Morgan, 1950). This simple heredity pat- of physiological functions (Coleman & Hummel,
tern, coupled with the simple phenotype, made 1973). On the other hand, hypothalamic lesions
this gene of great interest to mammalian geneti- also produced a similar wide range of functions,
cists. Eventually this gene was bred onto the but the relation of the ob gene to hypothalamic
standard C57Bl/6J background, which provided function was completely obscure for decades.
a stark contrast:in the presence of the gene, mice A mutation in another gene produced pheno-
were pale yellow, but in the absence of the gene, types similar to mutations in the ob gene but
mice were black. Also of increasing interest, was designated the diabetes (db) gene because
though at the time possibly underappreciated, on the earliest backgrounds studied, the obe-
the yellow color segregated with a variety of other sity was associated with robust hyperglycemia
phenotypes, including increased susceptibility to (Hummel, Dickie, & Coleman, 1966), compared
various cancers and obese phenotypes (Morgan, to the relatively mild hyperglycemia produced by
1950). This obese phenotype had been noted pre- the ob gene. However, further breeding demon-
viously but was not robust on other genetic back- strated that the ob gene and the db gene produced
grounds. The complexity of the phenotypes and essentially identical phenotypes when placed
574 Part IV:Homeostatic Therapies

on the same genetic backgrounds (Coleman & polymorphisms, had developed substantially to
Hummel, 1973; 1975). Nevertheless, the same the point that single-gene mutations causing a
studies clearly demonstrated that the obese and specific phenotypes could be determined by posi-
the diabetes genes were physically distinct. The tional cloning. The first mutation known to cause
similarity of the phenotypes produced by muta- obesity to be isolated using this approach was the
tions in these distinct genes suggested that their Agouti (viable Yellow) gene (Bultman, Michaud,
functions might be part of the same system. & Woychik, 1992; Miller et al., 1993). Isolation
Following the pioneering studies of Hervey of this gene surprisingly demonstrated that the
(1959), Coleman developed the hypothesis that mutation was not in the coding region of the gene
the ob gene and the db gene code for a hormone but in the promoter, leading to ectopic expression
and its receptor (without specifying which was of the peptide coding for the gene (Miller etal.,
which). As indicated earlier, Herveys studies 1993). The wild-type version of the Agouti gene
had shown that when obese rats with lesions in is normally expressed almost exclusively in hair
the VMH were joined by parabiosis with normal follicles and the epidermis (Miller et al., 1993).
rats, the normal rats rapidly lost weight whereas Since mechanisms regulating coat color have
the obese rats retained their obesity. These stud- been extensively elucidated, it seemed plausible
ies suggested that a circulating satiety factor that enhanced expression of the peptide coded
normally acts on the hypothalamus, but when by the Agouti gene would likely interfere with
the hypothalamus is damaged it can no longer these mechanisms. Since the product appeared to
respond to this factor, leading to increased levels be a secreted peptide, the most plausible hypoth-
of the factor in proportion to increased adipos- esis was that the peptide served to antagonize
ity. To address this hypothesis, this group car- the action of alpha-melanin-stimulating hor-
ried out a parabiotic analysis, first linking the mone (-MSH), whose action at the melanocor-
circulatory systems of db/db mice to wild-type tin receptor is known to promote the formation
mice (Coleman & Hummel, 1969). In Colemans of eumelanin, the main compound producing
now-classic studies, whereas db/db mice contin- dark coat color. Thus it was of great interest that
ued to gain weight, the wild-type mice connected shortly after the discovery of the agouti peptide,
to db/db mice starved to death (Coleman & it was shown to block the activity of -MSH at the
Hummel, 1969), just as in Herveys VMH-lesioned melanocortin receptor, reducing the production
rats (Hervey, 1959), leading Coleman to conclude of cAMP and eumelanin (Lu etal.,1994).
that db/db mice fail to sense a peripheral signal of While these studies did not directly demon-
satiety or adiposity, thus leading to an overabun- strate the cause of the obesity, the authors also
dance of this signal, which when reaching the reported that the agouti protein antagonized the
circulation system of wild-type mice led to star- binding of -MSH to the homologous melanocor-
vation. In a subsequent study, Coleman linked tin receptor, the MCR4 receptor (Lu etal., 1994).
ob/ob mice to db/db mice, leading to death by The authors suggested that obesity might there-
starvation in the ob/ob mice, whereas the db/db fore be caused by the action of the agouti gene
mice continued to gain weight (Coleman, 1973). to antagonize VMH -MSH acting outside the
Furthermore, when ob/ob mice were linked to skin. The obvious hypothesis was that -MSH was
wild-type mice, both the wild-type and ob/ob acting on the MC4R receptor in the hypothala-
mice continued to gain weight, although ob/ob mus. However, at that point there had been lit-
mice gained weight somewhat more slowly than tle evidence that -MSH played an important role
when not linked to wild-type mice (Coleman, in regulating energy balance. There were reports
1973). From these studies Coleman concluded of modest effects to reduce food intake by -MSH
that the obese mouse is unable to produce suf- (Poggioli, Vergoni, & Bertolini, 1986; Tsujii &
ficient satiety factor to regulate its food con- Bray, 1989), but these effects were not substan-
sumption, whereas the diabetes mouse produces tially distinguishable from the modestly anorec-
satiety factor, but cannot respond to it because tic effects of many other peptides.
of a defective satiety center (Coleman, 1973). Shortly after the cloning of the Agouti gene,
These predictions were borne out by subsequent the obese gene was also cloned by positional clon-
molecular genetic studies, but. as described later, ing (Zhang etal., 1994). The cloning of the obese
the actual situation is probably more complicated gene was somewhat more technically challeng-
thanthat. ing because the mutation in this gene that causes
By the 1990s genetic techniques, particu- obesity is recessive, in contrast to the dominant
larly the present of restriction enzyme length character of the Agouti gene. In any case the
Malignant Brain Cancer Management with Metabolic Therapy 575

likely function of the protein encoded by the observe a clinically meaningful weight loss after
obese gene was immediately clear from the cod- treatment with leptin (Heymsfield et al., 1999).
ing sequence, which indicated a secreted poly- Of particular interest was that Agouti mice were
peptide made predominantly in adipose tissue characterized by relatively elevated expressions
(Zhang etal., 1994). Thus the cloning of the obese of leptin in adipose tissue, raising the possibil-
gene immediately suggested that this gene coded ity that leptin acts through the melanocortin
for the circulating product originally suggested system and that these mice are relatively resist-
by the parabiosis studies of Hervey and especially ant to leptin (Mizuno etal., 1996). The parabiotic
Coleman. The studies of Hervey suggested that studies of Coleman suggested that the product of
the product of the obese gene would act in the the obese gene product (now referred to as leptin)
VMH, and the studies of Coleman suggested that might be the hypothetical feedback humoral
the product would act via a receptor. factor(s) (presumably a hormone or hormones)
These hypotheses were quickly corroborated. that accounts for the starvation that occurs in
First, the protein product encoded by the obese the control animal linked to the obese animal.
gene was demonstrated to reduce food intake However mice expressing transgenic leptin,
and body weight when injected into either ob/ob whose levels are on the order of those observed
mice or wild-type mice but had no effect in db/db in db/db mice, do not starve to death and indeed
mice (Halaas etal., 1995); thus the coded protein eventually increase adiposity to almost normal
was christened leptin, based on the Greek word levels (Qiu et al., 2001). This suggests that rats
for light or, as the authors indicated, thin. with hypothalamic lesions, and (leptin recep-
Based on the published sequence, Tartaglia etal. tor deficient) db/db mice, express other factors
labeled leptin and screened various tissues and in addition to leptin that account for the starva-
cell types for binding activity, eventually clon- tion phenotype. In view of the inefficacy of leptin
ing a gene from choroid plexus that bound the treatment in human obesity (Heymsfield et al.,
peptide (Tartaglia etal., 1995). Of particular note 1999), the search for such factors would appear to
was that the gene mapped near the diabetes locus, be particularly compelling.
coded for a single membrane receptor similar to A major breakthrough in the field came from
the IL-6 cytokine receptor and a variant of which the demonstration that targeted disruption of the
was expressed in the hypothalamus (Tartaglia MC4R receptor, which is not associated with skin
etal., 1995). Shortly thereafter the diabetes gene color, causes obesity in mice (Huszar etal., 1997).
was cloned by positional cloning and shown to Although this genetic study was somewhat at
code for the same protein as reported by Tartaglia odds with the pharmacological studies suggest-
etal. (1995); Lee etal. (1996); Chen etal. (1996). ing that -MSH (the ligand for the MCR4 recep-
Once leptin was available for study, it was tor) produces relatively modest reductions in
quickly demonstrated that, consistent with the food intake, subsequent studies amply corrobo-
hypothalamic localization of the leptin recep- rated the importance of the melanocortin system
tor, the hormone was particularly effective when in the regulation of energy balance and glucose
infused directly into the third ventricle of the homeostasis. A key set of observations were
hypothalamus, which largely targets the ven- based on the regulation of the gene that codes for
tromedial/arcuate hypothalamic area (Schwartz -MSH, proopiomelanocortin (POMC). This gene
et al., 1996). In particular, infusion of leptin codes for the precursor protein that is proteolyti-
into the ventromedial nucleus produced a rapid cally processed into several active neuropeptides,
reduction in blood glucose (Minokoshi, Haque, including beta-endorphin, -MSH, and cortico-
& Shimazu,1999). tropin (also known as ACTH), depending on the
While the 1990s were a high point in the field tissue (hence the rather complex name). The gene
of obesity, the hopes that leptin might be the for this precursor peptide was originally isolated
equivalent of insulin, and that obesity is caused from pituitary tumor cells on the basis of its cod-
by a deficiency that could be treated by hormone ing for ACTH but was then shown to also code
replacement, were quickly disappointed. In fact, for beta-endorphin (Roberts & Herbert,1977).
it was clear almost at once that obesity is almost The POMC precursor in the pituitary, from
always associated with increased, rather than which it was originally cloned, is processed to
decreased, leptin secretion, suggesting that obe- produced ACTH, which is then released into
sity, if anything, is characterized by leptin resist- systemic circulation to stimulate glucocorti-
ance, analogous to insulin resistance (Frederich coid production when acting on the adrenal
et al., 1995). Subsequent clinical trials failed to glands through the MCR2 receptor. Interestingly
576 Part IV:Homeostatic Therapies

Cushings disease, a condition characterized by discovered by more conventional pharmacologi-


hyperactivity of the adrenal gland and thus of cal methods. The first evidence was that NPY,
elevated glucocorticoid secretion, can be caused which was originally discovered as a fairly abun-
by pituitary tumors that hypersecrete ACTH. dant peptide in the whole brain with no clear
Cushings disease is characterized most notably function, is expressed in particularly high levels
by insulin resistance (sometimes diabetes) asso- in the hypothalamus, leading to assessment of its
ciated with syndromic deposition of adipose tis- effect on food intake (Clark et al., 1984). These
sue in unusual areas such as the upper back. Thus studies demonstrated that infusing NPY into
this product of the POMC gene can, under unu- the third ventricle stimulated food intake (Clark
sual circumstances, drive a metabolic syndrome etal., 1984). This observation was not particularly
including obesity. While beta-endorphin is also remarkable since the infusion of many neuro-
produced by processing POMC in the pitui- peptides into the third ventricle either stimulate
tary, the function of pituitary beta-endorphin is or inhibit food intake, and the significance of
notknown. these pharmacological effects are rarely clear.
POMC is also expressed in the VMH as well However, in a key follow-up study, Sanacora etal.
as brainstem, where it is differentially processed demonstrated that NPY expressed specifically in
to produce -MSH rather than ACTH (-MSH the arcuate nucleus was relatively induced both
is derived from a further cleavage of ACTH, in genetically obese Zucker rats (which were sub-
releasing the N-terminal peptide). The func- sequently shown to be characterized by a muta-
tion of hypothalamic POMC had been obscure tion in the leptin receptor [Takaya etal.,1996])
(a popular hypothesis was that the key pep- and by fasting (Sanacora et al., 1990). Thus the
tide was beta-endorphin, which might regulate regulation of NPY is precisely inverted relative
reproductive function), but after it became unex- to POMC (Mizuno et al., 1998; Schwartz et al.,
pectedly clear that the receptor for -MSH (the 1997; Thornton etal., 1997). However, in contrast
MC4R receptor) plays a key role in regulating to POMC, complete genetic ablation of NPY pro-
energy balance, a role for hypothalamic POMC duces little if any metabolic phenotype (e.g., on
in energy balance was assessed. The key obser- food intake, body weight, hyperphagic response
vations were that expression of hypothalamic to fasting, and weight loss in response to leptin)
POMC was reduced in ob/ob (leptin-deficient) although such mice exhibit an enhanced suscep-
and db/db (leptin receptor deficient) mice, as well tibility to seizures (Erickson, Clegg, & Palmiter,
as by fasting (Mizuno etal., 1998; Schwartz etal., 1996). These studies raise major concerns about
1997; Thornton etal., 1997), supporting previous the physiological importance of NPY for the
observations suggesting that leptin acts through regulation of energy balance. Nevertheless, abla-
the hypothalamic melanocortin system (Mizuno tion of NPY does reduce obese phenotypes in the
et al., 1996). Subsequent studies demonstrated complete absence of leptin (Erickson, Hollopeter,
that ablation of neuronal POMC, leaving pitui- & Palmiter, 1996), similar to effects of transgenic
tary POMC intact, produces massive obese phe- restoration of POMC (Mizuno etal.,2003).
notypes (Smart, Tolle, & Low,2006). Similar to NPY in hypothalamic location,
A key observation linking POMC to the leptin regulation, and function is the neuropeptide
system was that transgenic restoration of POMC Agouti-related peptide (AgRP) (Ollmann et al.,
in ob/ob mice reversed obese phenotypes by 1997). The gene for this neuropeptide was dis-
about 50% and completely reversed impairments covered in a search of a plausible hypothalamic
in glucose homeostasis in ob/ob mice independ- homolog to the wild-type Agouti gene, which in
ent of improvements in body weight and food wild-type mice is expressed principally in the
intake (Mizuno et al., 2003). Subsequent stud- skin, not the brain (Miller et al., 1993). In con-
ies using alternative approaches generally cor- trast, AgRP is expressed in the hypothalamus;
roborated these studies (Balthasar et al., 2004; of particular interest, AgRP also antagonizes
Berglund etal.,2012). the MC4R receptor, and, even more impres-
Another neuropeptide expressed in the VMH sively, transgenic overexpression causes obesity
(more precisely in the medial arcuate nucleus) and without changing coat color (Ollmann et al.,
implicated in the regulation of energy balance is 1997). In contrast to NPY, which is expressed
neuropeptide Y (NPY). In contrast to the lep- in many, if not most, areas of the brain, AgRP is
tin system and the melanocortin system, whose expressed almost exclusively in the medial aspect
role in energy balance was largely discovered by of the arcuate nucleus and in fact co-expresses
genetic analysis, the role of the NPY system was in NPY neurons in that area (Hahn etal., 1998).
Malignant Brain Cancer Management with Metabolic Therapy 577

Furthermore, both NPY and AgRP are induced lateral aspect of the arcuate nucleus as well as
by fasting and in genetically obese mice (Mizuno substantially lateral to and outside of the bound-
& Mobbs, 1999). It was therefore greatly disap- aries of this nucleus (Bergen etal., 1998). While
pointing that genetic ablation of both NPY and MSG did reduce POMC expression (presum-
AgRP had no evident effect on energy balance ably by destroying the medial POMC neurons),
(Qian et al., 2002). Nevertheless, ablation of it appears that the lateral POMC neurons were
AgRP neurons in adult, though not neonatal, spared (Bergen etal., 1998), and of course MSG
neurons leads to dramatic reduction in food did not produce the robust obesity produced by
intake (Luquet et al., 2005), suggesting that at genetic ablation of POMC (Smart, Tolle, & Low,
least in adults something produced by these neu- 2006) or the relevant MC4R receptor (Huszar
rons (though apparently neither AgRP nor NPY) et al., 1997). In contrast, GTG reduced total
plays an important in regulating energy balance POMC by about the same amount (presumably
under normal circumstances. targeting mainly POMC neurons outside the
Another key neuropeptide, expressed in the arcuate and near the boundary of the VMN) and
lateral hypothalamus, is melanocyte concen- also of course produced extremely robust obe-
trating hormone (MCH) (Qu et al., 1996). This sity and hyperphagia (Bergen etal., 1998). These
neuropeptide is elevated in the lateral hypothala- data clearly suggested that a set of neurons GTG
mus of genetically obese ob/ob mice, and central lesions, presumably targeting the larger part of
infusion increases food intake (Qu et al., 1996). the VMN in addition to POMC neurons, plays
Furthermore, in contrast to the lack of pheno- a key role in regulating energy balance (Bergen
type by ablation of NPY and AgRP, ablation of et al., 1998). However, in contrast to the NPY/
MCH produces a lean (though not starving) phe- AgRP and POMC, the relevant neuropeptides
notype (Shimada et al., 1998), and overexpres- (if any) remained elusive. Nevertheless, the
sion produces modest obese phenotype (Ludwig observations may be explained at least in part
etal.,2001). by evidence that VMN neurons directly regulate
An unexpected consequence of the remarka- (activate) POMC neurons (Sternson, Shepherd, &
ble discoveries in the 1990s was a shift away from Friedman,2005).
an emphasis on the ventromedial nucleus and A series of molecular manipulations has led
toward the arcuate nucleus, primarily because to a better understanding of the nature of these
NPY/AgRP and POMC neurons were reported VMN neurons that play a key role in regulating
to be expressed in the arcuate nucleus. This was energy balance. Akey breakthrough was the rec-
puzzling because many previous studies had ognition that the transcription factor steroid fac-
demonstrated that lesions of the arcuate nucleus, tor 1 is highly expressed in VMN neurons (as well
especially by neonatal exposure to i.p. monoso- as adrenal and gonadal glands), ablation of which
dium glutamate (MSG), produced only very mild (with appropriate steroid replacement to main-
obesity, without hyperphagia, compared to the tain viability) causes obesity, eventually leading
far more robust obesity with hyperphagia pro- to mice with twice the weight of wild-type mice
duced by VMN lesions produced by electrolytic (Majdic etal., 2002). On the other hand, the obe-
destruction or GTG (Bunyan, Murrell, & Shah, sity in these mice develops relatively late and is
1976). Of particular pertinence, MSG lesions did not characterized by robust hyperphagia so dif-
not produce hypophagia, as would be expected fers from the classical robust obesity characteris-
if NPY/AgRP neurons play an important role in tic of VMN lesions, for example, by GTG (Majdic
enhancing food intake and producing pro-obese etal.,2002).
phenotypes, consistent with the lack of metabolic An unexpected line of investigation arose
phenotypes produced by ablating both NPY and from the observation that chronic infusion of
AgRP (Bunyan, Murrell, & Shah,1976). the neurotrophin brain-derived neurotrophic
On the other hand, these apparently para- factor (BDNF) reduces body weight (Lapchak &
doxical observations were not as simple as they Hefti, 1992), although as discussed previously
appeared. First, although it is widely assumed such pharmacological effects are widely reported
that POMC neurons are confined to the arcuate for many neuropeptides (e.g., NPY), so in itself
nucleus, this is not the case. NPY/AgRP neurons this observation generated limited interest at
are confined entirely to the medial aspect of the the time. However, in contrast to NPY, genetic
arcuate nucleus and thus are almost completely ablation of BDNF (heterozygous only because
eliminated by neonatal MSG (Bergen etal., 1998). homozygous ablation is lethal) also led to the
In contrast, POMC neurons are expressed in the expected hyperphagia and weight gain (Kernie,
578 Part IV:Homeostatic Therapies

Liebl, & Parada, 2000). Of even greater interest, Nolan, 1997) and ghrelin (Kojima et al., 1999),
forebrain-specific postnatal ablation of BDNF also report more short-term status of nutritional
produced an even more robust hyperphagia resources. However, the physiological signifi-
and obesity (Xu et al., 2003), almost as great as cance of these signals remains to be determined,
observed in leptin-deficient mice (the gold stand- since >95% reduction of brain insulin recep-
ard for genetic obesity). This was of particular tors produces no effect on body weight in male
interest because other than the hippocampus, mice and only an extremely marginal increase
the hypothalamus expresses the highest levels in female mice (Bruning etal., 2000). Similarly,
of BDNF, and in the hypothalamus, BDNF is ablation of ghrelin or its receptor produces lit-
expressed almost exclusively in the VMN and tle if any effect on energy balance though there
is induced by nutritional stimulation possibly is some effect on glucose homeostasis (Sun etal.,
downstream of POMC neurons via the MC4R 2008). Therefore, among hormonal signals, leptin
(Xu etal., 2003)as well as glucose (Unger etal., is by far the most implicated in regulating energy
2007). Furthermore, ablation of BDNF in VMN balance, although many lines of evidence argue
recapitulates the obesity observed after forebrain against a clear role of the leptin system in the
ablation of BDNF (Unger et al., 2007), whereas vast majority of human obesity (e.g., leptin sup-
infusion of BDNF into the VMH reduces food plementation has no significant effect in obese
intake and increases energy expenditure (Wang humans [Heymsfield etal.,1999]).
etal., 2010). The role of BNDF became even more As indicated, the hypothesis that glucose-
difficult to resolve when it was discovered that the sensing neurons in the VMN play an important
form of BDNF most relevant to energy balance is role in energy balance, as described by the glu-
produced in the dendrites, not the cell body, of costat hypothesis (Mayer, 1953), animated much
VMN neurons, where it plays a key role in medi- research in metabolism for several decades, but
ating responses to leptin (Liao etal.,2012). this hypothesis had been largely abandoned by
However, in contrast to neuropeptides, whose the 1980s (Even & Nicolaidis, 1986) and effec-
mode of action is presumably via G-protein cou- tively abandoned after the discovery of leptin.
pled receptors, BDNF is thought to act primarily However, an important role for hypothalamic
through trophic effects via trkB, the BDNF recep- nutrient sensing in regulating energy balance
tor (Lobo etal., 2010). This raised the hypothesis arose unexpectedly from studies on the role of
that BDNF in dendrites from VMN neurons acts fatty acid synthase in cancer (Loftus etal., 2000).
to stabilize connections with neurons whose pro- Based on the hypothesis that cancer cells must
jections express the trkB receptor (Liao et al., synthesize fatty acids at a high rate to allow for
2012). Since trkB receptors in dopamine-receptor membrane and organelle replication, Kuhajda
1 expressing neurons originating in the nucleus et al. synthesized C75, a potent inhibitor of
accumbens appear to play a key role in mediat- fatty acid synthase with promising anti-tumor
ing reward, and these neurons project to the lat- properties (Kuhajda et al., 2000). In course of
eral hypothalamus as do dendrites from VMN characterizing the physiological effects of this
neurons, we hypothesized that BDNF in VMN compound, Loftus etal. observed that C75 sub-
dendrites plays a key role linking food-induced stantially reduced food intake, usually an indi-
reward with food intake and obesity (Schwartz & cation of toxicity (Kuhajda etal., 2000). To their
Mobbs, 2012). Such a hypothesis might explain credit however, these investigators examined in
why atypical antipsychotic drugs robustly pro- detail the mechanism of anorexia, and found
mote obesity in patients given these drugs, which the mechanism to require elevated hypotha-
otherwise remains a major medical mystery lamic malonyl-CoA, an expected consequence
(Pramyothin & Khaodhiar,2010). of inhibiting fatty acid synthase, which normally
uses malonyl-CoA in synthesizing fatty acids
(Kuhajda et al., 2000). Since a classic effect of
HYPOTHALAMIC malonyl-CoA (which is normally exported to the
NUTRITIONAL SIGNALS: cytoplasm when glucose metabolism is active)
S U B S T R AT E C O M P E T I T I O N is to reduce carnitine palmitoyltransferase 1A
AS A CAUSE OF OBESIT Y (Cpt1a), the rate-limiting enzyme for beta oxi-
In addition to the robust effects of leptin, which dation of fatty acids, these investigators boldly
serves as a signal to hypothalamic neurons to hypothesized that inhibiting hypothalamic beta
report adipose stores (Ahima & Flier, 2000), oxidation inhibits food intake and body weight,
two other hormonal signals, insulin (Woods & probably in interaction with hypothalamic
Malignant Brain Cancer Management with Metabolic Therapy 579

glucose-sensing neurons (Kuhajda et al., 2000). produced by a high-fat diet, at least in rodents
This was bold because the standard view, based (Surwit etal.,1988).
on studies of cortical neurons, was (and gen- It should be noted that the effects of dietary
erally still is) that neurons cannot support composition on obesity are complex, as we have
beta oxidation (i.e., metabolism of fatty acids), previously described (Mobbs etal., 2007). Thus,
although astrocytes can (Edmond et al., 1987). in mice, a high-fat diet with relatively normal
Nevertheless, based on studies in the periph- carbohydrate and protein concentrations pro-
ery, Ruderman et al. had hypothesized that duces obesity relative to a high-carbohydrate,
such substrate competition might indeed exist low-fat diet (Surwit etal., 1988), but a ketogenic
in glucose-sensing neurons and thus play a role diet even higher in fat with low carbohydrates
in the physiological function of these neurons and protein actually reduces adiposity (Kennedy
(Ruderman etal.,1999). etal., 2007). Asimilar diet in humans also facili-
The hypothesis that enhanced beta oxidation tates weight loss (Brehm etal., 2003). The mecha-
in hypothalamic neurons promotes obese pheno- nisms mediating these effects are not known
types was supported by the report that inhibition but may depend on the unique metabolic state
of hypothalamic Cpt1a (also called liver-specific produced by ketogenesis (Kennedy et al., 2007;
Cpt1) reduced food intake and hepatic glu- Mobbs etal.,2007).
cose production (Obici et al., 2003). Similarly,
pro-obese phenotypes of ghrelin are mediated GENETIC OBESITY
in part by induction of hypothalamic beta oxi- INHUMANS
dation (Andrews etal., 2008). Specifically, ghre- Genetic studies in mice, implicating leptin and the
lin induced hypothalamic expression of Cpt1(a), melanocortin system, led to the discovery (using
and inhibition of Cpt1a activity with etomoxir the candidate gene approach) that mutations in
blocked the effects of ghrelin to increase food these systems also play a key role in the regula-
intake (Andrews et al., 2008). Consistent with tion of human adiposity. For example, mutations
these observations, we reported that fasting and in leptin (Montague etal., 1997), the leptin recep-
hypoglycemia produces changes in gene expres- tor (Clement etal., 1998), the MCR4 receptor (Yeo
sion (including elevation of Cpt1a) indicating a etal., 1998), and POMC itself (Challis etal., 2002;
shift away from glucose utilization and toward Jackson etal., 1999)can cause obesity in humans.
beta oxidation (Mobbs et al., 2004; Poplawski The most common cause of single-gene mutations
etal., 2010; Poplawski, Mastaitis, & Mobbs, 2011). that cause obesity in humans arise in the MC4R
We suggested that at least some of these effects are receptor (Yeo etal.,1998).
due to activation by hypoglycemia and/or fast- Noncandidate gene approaches, especially
ing of PPAR- (Poplawski etal., 2010; Poplawski, more recent studies with very high power
Mastaitis, & Mobbs, 2011), whose activation (>200,000 individuals), such as genome-wide
during fasting (by free fatty acids) mediates the association studies (GWAS), have corroborated
activation of Cpt1a and inhibition of glucose that obesity is indeed heritable but that polymor-
metabolism (Muoio etal., 2002). Since PPAR- in phisms in no single gene can account for more
principle activates the same genes as PPAR- (they than a very small amount (<1%) of the variance
both target genes with PPAR response ele- in body mass index. The main candidate for
ments, this hypothesis could also accommodate human obesity that has arisen from these stud-
evidence that hypothalamic PPAR- promotes ies is the fat mass and obesity associated (FTO)
obese phenotypes, especially under conditions gene (Hinney, Vogel, & Hebebrand, 2010). While
of excess free fatty acids (e.g., on a high-fat diet) polymorphisms in this gene are the most reliably
(Diano etal., 2011; Ryan etal.,2011). associated with adiposity in humans (Hinney,
In sum, these data support the hypoth- Vogel, & Hebebrand, 2010), the functional sig-
esis that metabolism of glucose and free fatty nificance of the FTO polymorphism is unclear,
acids in specialized nutrient-sensing hypo- including whether increased or decreased activ-
thalamic neurons are in competition, with ity of FTO is associated with obesity. One line of
fatty acid metabolism (beta oxidation) driving evidence suggests that fasting reduces hypotha-
pro-obese phenotypes and glucose oxidation lamic FTO and glucose induces it (i.e., regulation
opposing obese phenotypes. Since free fatty similar to BDNF and POMC), so it seems likely
acids activate fatty acid oxidation (presum- that decreased activity of FTO (whatever it is
ably at the expense of glucose oxidation), this doing) is associated with obesity. Nevertheless,
could account for the phenomenon of obesity although FTO is presently the most promising
580 Part IV:Homeostatic Therapies

genetic basis of human obesity, the functional we were before the heroic age of discovery in the
basis of this risk remains to be determined. regulation of energy balance and glucose homeo-
A commonly held hypothesis is that human stasis. In particular, it is clear from GWAS stud-
obesity is actually quite simple to explain:calo- ies that no single polymorphism can account for
ries consumed are simply greater than calories any but a tiny fraction of the variance of adipos-
expended over time, thus leading to the storage ity, and even aggregating all the genes implicated
of excess calories that accumulate over time, by GWAS, no common mechanisms emerge as
due at least in part to an inherent bias toward particularly clear or dominant. In any case, the
weight gain (Schwartz etal., 2003). Thus between best evidence for the total combined effect of all
ages 50 and 70, average weight gain is about 0.8 polymorphisms on adiposity is likely to be no
pounds/year (Mozaffarian etal., 2011). This plau- more than about 50% of variance of adiposity
sibly reflects that caloric consumption each year (Hjelmborg et al., 2008; Maes, Neale, & Eaves,
is very slightly greater than caloric expenditure 1997). Thus it is unclear that focusing on poly-
(indeed, it is remarkable that the difference is not morphisms in individual genes is likely to provide
greater). While this is an appealing simple poten- substantial insight into causes of human obesity.
tial mechanism, it is difficult to reconcile with the It might be assumed that the increase in the
evidence that while it is relatively easy for most prevalence of obesity over the past 20 years,
people to lose substantial weight in a supervised clearly not due to changing genotypes, must be
program, weight is regained relatively quickly. due to changing environment, most likely in the
A typical example is a recent study examin- increased palatability and reduced cost of pro-
ing weight gain in patients who had within the cessed food. While this would appear to be plau-
last year lost about 17 pounds due to intentional sible, there is at present no clear evidence that
weight loss, largely for health reasons (Sherwood any specific environmental factor (specifically
et al., 2013). Patients were then encouraged to including sucrose or fructose) has significantly
maintain weight loss by following a manual and contributed to the remarkable increase in obesity
log book promoting standard weight loss strat- observed in the past 20years.
egies, in addition to two follow-up phone calls We therefore suggest that an underappreci-
(Sherwood et al., 2013). These patients gained ated phenomenon may provide the most promis-
about 10 pounds in the following 2years; that is, ing avenue to understand human obesity:a major
they gained weight over five times faster than the source of variance in adiposity that appears to
rate at which they likely accrued these 10 pounds be neither genetic nor environmental. This phe-
to begin with, despite intensive efforts to main- nomenon was observed in the context of obesity
tain the weight (likely far more effort than the produced in rodents placed on a high-fat diet
original exerted to prevent the original weight (Surwit etal., 1988). While rodents generally gain
gain). Similar rapid gain after substantial weight substantial weight when given access to a high-fat
loss has been widely exemplified by many celebri- diet, the mechanisms mediating this phenome-
ties (e.g., Oprah Winfrey [Shugart,2011]). These non are unclear. It is plausible that at least part
observations clearly indicate that a slow accrual of the mechanism has to do with activation of
of adiposity due to a slight mismatching between the PPAR system, which in turn may promote
caloric intake and caloric expenditure may hypothalamic beta oxidation (and reduce hypo-
account for the original increase in adiposity but thalamic glucose metabolism), thus promoting
that this accrual entails regulatory adjustments obese phenotypes as described earlier. Possibly
that effectively defend the new body weight. of greater interest is the basis of the individual
differences in response to a high-fat diet. Genetic
O B E S I T Y: N E I T H E R N AT U R E background can profoundly influence response
NOR NURTURE to weight gain on a high-fat diet (Levin et al.,
This review has focused on the genetic basis of 1997; Surwit etal., 1991). However, as in human
energy balance, the study of which in mice has obesity, it appears that no single genetic poly-
led to profound insights on the molecular mech- morphism can account for more than a relatively
anisms involved. Nevertheless, this approach small amount of the variance in diet-induced
clearly has major limitations in that, despite obesity (Fawcett etal.,2009).
extensive progress in the past 20years, we seem However, a relatively underappreciated phe-
hardly closer to understanding human obesity nomenon is the surprising degree of variability in
(with the exception of vary rare individuals whose weight gain on a high-fat diet in genetically homo-
obesity is caused by single-gene mutations) than geneous inbred C57Bl/6J mice (Koza etal., 2006).
Malignant Brain Cancer Management with Metabolic Therapy 581

Indeed, the coefficient of variation of genetically Andrews, Z.B., etal., UCP2 mediates ghrelins action
homogeneous mice on essentially identical diets on NPY/AgRP neurons by lowering free radicals.
(Koza etal., 2006)is about the same as the coeffi- Nature, 2008. 454(7206):846851.
cient of variance of human body mass index. This Backhed, F., etal., The gut microbiota as an environ-
observation leads to the rather surprising con- mental factor that regulates fat storage. Proc Natl
clusion that neither heredity nor environment Acad Sci USA, 2004. 101(44):1571815723.
is chiefly responsible for individual variance in Balthasar, N., et al., Leptin receptor signaling in
adiposity. We have obtained preliminary evi- POMC neurons is required for normal body
dence supporting that variance of weight gain on weight homeostasis. Neuron, 2004. 42(6):983991.
a high-fat in genetically similar C57Bl/6J mice, as Bergen, H.T., et al., Hyperphagia and weight gain
well as across different mouse strains, correlates after gold-thioglucose: Relation to hypotha-
with hypothalamic expression of the same small lamic neuropeptide Y and proopiomelanocortin.
set of genes (unpublished data). Variation in the Endocrinology, 1998. 139(11):44834488.
expression of these genes probably arose from Berglund, E.D., etal., Direct leptin action on POMC
neurons regulates glucose homeostasis and
stochastic processes that arose during develop-
hepatic insulin sensitivity in mice. J Clin Invest,
ment or aging. Understanding the role of these
2012. 122(3):10001009.
genes in determining body weight may lay the
Borg, W.P., etal., Ventromedial hypothalamic lesions
groundwork for the finally understanding, and
in rats suppress counterregulatory responses to
manipulating, hypothalamic determinants of
hypoglycemia. J Clin Invest, 1994. 93(4):16771682.
energy balance.
Borg, M.A., etal., Local ventromedial hypothalamus
An alternative source of variability could glucose perfusion blocks counterregulation dur-
be the population of microbes in the intesti- ing systemic hypoglycemia in awake rats. J Clin
nal tract. This hypothesis was initially most Invest, 1997. 99(2):361365.
prominently supported by the study of Backhed Borg, M.A., et al., Chronic hypoglycemia and dia-
etal. who reported that germ-free mice exhibit betes impair counterregulation induced by
relatively reduced adiposity and that introduc- localized 2-deoxy-glucose perfusion of the ven-
tion of microbiota (derived from the cecum of tromedial hypothalamus in rats. Diabetes, 1999.
conventionally raised mice) increased adiposity 48(3):584587.
by 60% (Backhed et al., 2004). This increased Borg, M.A., et al., Local lactate perfusion of the
adiposity was attributed to the likely effect of ventromedial hypothalamus suppresses hypo-
bacteria to enhanced absorbance of monosac- glycemic counterregulation. Diabetes, 2003.
charides from the gut, leading to enhanced 52(3):663666.
lipogenesis (Backhed etal., 2004). Other expla- Brabant, G., et al., Serum leptin levels following
nations of this effect have included direct hypothalamic surgery. Horm Metab Res, 1996.
gut-brain nutrient sensing, possibly modulated 28(12):728731.
by intestinal bacteria (Moran & Shanahan, Bray, G.A., Commentary on classics of obesity.
2014). On the other hand, not all studies 4. Hypothalamic obesity. Obes Res, 1993.
have supported this hypothesis. For example, 1(4):325328.
germ-free mice were reported to weigh the Brehm, B.J., et al., A randomized trial com-
same as conventional mice and actually gain paring a very low carbohydrate diet and a
more weight on a high-fat diet than conven- calorie-restricted low fat diet on body weight and
tional mice (Fleissner et al., 2010). Similarly, cardiovascular risk factors in healthy women. J
germ-free rats were reported to weigh the Clin Endocrinol Metab, 2003. 88(4):16171623.
same as conventional rats (Swartz etal., 2013). Bruning, J.C., et al., Role of brain insulin recep-
Nevertheless, the gut microbiome continues to tor in control of body weight and reproduction.
constitute an intriguing potential mechanism Science, 2000. 289:21222125.
to explain variance of adiposity independent of Bultman, S.J., E.J. Michaud, and R.P. Woychik,
heredity or environment. Molecular characterization of the mouse agouti
locus. Cell, 1992. 71(7):1195204.
References
Bunyan, J., E.A. Murrell, and P.P. Shah, The induc-
Ahima, R.S. and J.S. Flier, Leptin. Annu Rev Physiol, tion of obesity in rodents by means of monoso-
2000. 62:413437. dium glutamate. Br J Nutr, 1976. 35(1):2539.
Ahima, R.S., et al., Role of leptin in the neuro- Challis, B.G., et al., A missense mutation disrupt-
endocrine response to fasting. Nature, 1996. ing a dibasic prohormone processing site in
382(6588):250252. pro-opiomelanocortin (POMC) increases
582 Part IV:Homeostatic Therapies

susceptibility to early-onset obesity through a Erickson, J.C., G. Hollopeter, and R.D. Palmiter,
novel molecular mechanism. Hum Mol Genet, Attenuation of the obesity syndrome of ob/ob
2002. 11(17):19972004. mice by the loss of neuropeptide Y. Science, 1996.
Chen, H., et al., Evidence that the diabetes gene 274(5293):17041707.
encodes the leptin receptor: identification of a Even, P. and S. Nicolaidis, Short-term control of
mutation in the leptin receptor gene in db/db feeding: Limitation of the glucostatic theory.
mice. Cell, 1996. 84(3):491495. Brain Res Bull, 1986. 17(5):621626.
Clark, J.T., et al., Neuropeptide Y and human pan- Fawcett, G.L., etal., Fine-mapping of obesity-related
creatic polypeptide stimulate feeding behavior in quantitative trait loci in an F(9/10) advanced
rats. Endocrinology, 1984. 115(1):427429. intercross line. Obesity (Silver Spring), 2009.
Clement, K., etal., A mutation in the human leptin 18(7):13831392.
receptor gene causes obesity and pituitary dys- Fleissner, C.K., et al., Absence of intestinal micro-
function. Nature, 1998. 392(6674):398401. biota does not protect mice from diet-induced
Coleman, D.L. and K.P. Hummel, Effects of parabio- obesity. Br J Nutr, 2010. 104(6):919929.
sis of normal with genetically diabetic mice. Am Fontaine, K.R., etal., Years of life lost due to obesity.
J Physiol, 1969. 217(5):12981304. JAMA, 2003. 289(2):187193.
Coleman, D.L. and K.P. Hummel, Influence of Frederich, R.C., etal., Leptin levels reflect body lipid con-
genetic background on the expression of muta- tent in mice:Evidence for diet-induced resistance to
tions at the diabetes locus in the mouse. II. leptin action. Nat Med, 1995. 1(12):13111314.
Studies on background modifiers. In:Shafrir E, Hahn, T.M., etal., Coexpression of AGRP and NPY
ed. Contemporary topics in the study of diabe- in fasting-activated hypothalamic neurons.
tes and metabolic endocrinology, pp. 182187. Nature Neurosci, 1998. 1(4):271272.
NewYork, Academic Press, 1975:810. Halaas, J.L., et al., Weight-reducing effects of the
Coleman, D.L., Effects of parabiosis of obese with plasma protein encoded by the obese gene.
diabetes and normal mice. Diabetologia, 1973. Science, 1995. 269(5223):543546.
9(4):294298. Hervey, G.R., The effects of lesions in the hypo-
Coleman, D.L. and K.P. Hummel, The influence of thalamus in parabiotic rats. J Physiol, 1959.
genetic background on the expression of the 145(2):336352.
obese (Ob) gene in the mouse. Diabetologia, Hetherington, A.W. and S.W. Ranson, Hypothalamic
1973. 9(4):287293. lesions and adiposity in the rat. Anatomical
Daviglus, M.L., et al., Body mass index in middle Record, 1940. 78:149172.
age and health-related quality of life in older Heymsfield, S.B., et al., Recombinant leptin for
age: The Chicago heart association detection weight loss in obese and lean adults:Arandom-
project in industry study. Arch Intern Med, 2003. ized, controlled, dose-escalation trial. JAMA,
163(20):24482455. 1999. 282(16):15681575.
Davis, N.J., etal., Comparative study of the effects of a Hinney, A., C.I. Vogel, and J. Hebebrand, From mono-
1-year dietary intervention of a low-carbohydrate genic to polygenic obesity:Recent advances. Eur
diet versus a low-fat diet on weight and glycemic Child Adolesc Psychiatry, 2010. 19(3):297310.
control in type 2 diabetes. Diabetes Care, 2009. Hjelmborg, J.B., etal., Genetic influences on growth
32(7):11471152. traits of BMI:Alongitudinal study of adult twins.
Debons, A.F., et al., Diabetes-induced resistance Obesity (Silver Spring), 2008. 16(4):847852.
of ventromedial hypothalamus to damage by Hummel, K.P., M.M. Dickie, and D.L. Coleman,
gold thioglucose: reversal by adrenalectomy. Diabetes, a new mutation in the mouse. Science,
Endocrinology, 1974. 95(6):16361641. 1966. 153(3740):11271128.
Diano, S., etal., Peroxisome proliferation-associated Huszar, D., et al., Targeted disruption of the
control of reactive oxygen species sets melano- melanocortin-4 receptor results in obesity in
cortin tone and feeding in diet-induced obesity. mice. Cell, 1997. 88(1):131141.
Nat Med, 2011. 17(9):11211127. Jackson, R.S., et al., Proopiomelanocortin products
Edmond, J., et al., Capacity for substrate utiliza- and human early-onset obesity. J Clin Endocrinol
tion in oxidative metabolism by neurons, astro- Metab, 1999. 84(2):81920.
cytes, and oligodendrocytes from developing James, W.P., WHO recognition of the global obe-
brain in primary culture. J Neurosci Res, 1987. sity epidemic. Int J Obes (Lond), 2008. 32 Suppl
18(4):551561. 7:S120S126.
Erickson, J.C., K.E. Clegg, and R.D. Palmiter, Kennedy, A.R., et al., A high-fat, ketogenic diet
Sensitivity to leptin and susceptibility to seizures induces a unique metabolic state in mice.
of mice lacking neuropeptide Y. Nature, 1996. Am J Physiol Endocrinol Metab, 2007.
381(6581):415421. 292(6):E1724E1739.
Malignant Brain Cancer Management with Metabolic Therapy 583

Kernie, S.G., D.J. Liebl, and L.F. Parada, BDNF regu- Majdic, G., et al., Knockout mice lacking steroido-
lates eating behavior and locomotor activity in genic factor 1 are a novel genetic model of
mice. EMBO J, 2000. 19(6):1290300. hypothalamic obesity. Endocrinology, 2002.
King, B.M., The rise, fall, and resurrection of the 143(2):607614.
ventromedial hypothalamus in the regulation Matsuo, T. and A. Shino, Induction of diabetic alter-
of feeding behavior and body weight. Physiol ations by goldthioglucose-obesity in KK,ICR and
Behav, 2006. 87(2):221244. C57BL mice. Diabetologia, 1972. 8(6):391397.
Kojima, M., et al., Ghrelin is a growth- Mayer, J., Glucostatic mechanism of regulation of
hormone-releasing acylated peptide from stom- food intake. N Engl J Med, 1953. 249:1316.
ach. Nature, 1999. 402(6762):656660. Miller, M.W., et al., Cloning of the mouse agouti
Komeda, K., M. Yokote, and Y. Oki, Diabetic syn- gene predicts a secreted protein ubiquitously
drome in the Chinese hamster induced with expressed in mice carrying the lethal yellow
monosodium glutamate. Experientia, 1980. mutation. Genes Dev, 1993. 7(3):454467.
36(2):232234. Minokoshi, Y., M.S. Haque, and T. Shimazu,
Koza, R.A., etal., Changes in gene expression fore- Microinjection of leptin into the ventromedial
shadow diet-induced obesity in genetically iden- hypothalamus increases glucose uptake in periph-
tical mice. PLoS Genet, 2006. 2(5):e81. eral tissues in rats. Diabetes, 1999. 48(2):287291.
Kuhajda, F.P., etal., Synthesis and antitumor activity Mizuno, T.M. and C.V. Mobbs, Hypothalamic
of an inhibitor of fatty acid synthase. Proc Natl agouti-related protein messenger ribonucleic
Acad Sci USA, 2000. 97(7):34503454. acid is inhibited by leptin and stimulated by fast-
Lapchak, P.A. and F. Hefti, BDNF and NGF treatment ing. Endocrinology, 1999. 140(2):814817.
in lesioned rats: Effects on cholinergic function Mizuno, T.M., etal., Obese gene expression:reduc-
and weight gain. Neuroreport, 1992. 3(5):405408. tion by fasting and stimulation by insulin and
Lee, G.H., et al., Abnormal splicing of the leptin glucose in lean mice, and persistent elevation
receptor in diabetic mice. Nature, 1996. in acquired (diet-induced) and genetic (yellow
379(6566):632635. agouti) obesity. Proc Natl Acad Sci USA, 1996.
Levin, B.E., etal., Selective breeding for diet-induced 93(8):34343438.
obesity and resistance in SpragueDawley rats. Mizuno, T.M., et al., Rapid communication:
Am J Physiol, 1997. 273(2 Pt 2):R725R730. Hypothalamic pro-opiomelanocortin mRNA
Lewis, M., The lac repressor. C R Biol, 2005. is reduced by fasting and in ob/ob and db/db
328(6):521548. mice, but is stimulated by leptin. Diabetes, 1998.
Liao, G.Y., etal., Dendritically targeted BDNF mRNA 47(2):294297.
is essential for energy balance and response to Mizuno, T.M., et al., Transgenic neuronal expres-
leptin. Nat Med, 2012. 18(4):564571. sion of proopiomelanocortin attenuates
Lobo, M.K., et al., Cell type-specific loss of BDNF fasting-induced hyperphagia and reverses meta-
signaling mimics optogenetic control of cocaine bolic impairments in leptin-deficient obese mice.
reward. Science, 2010. 330(6002):385390. Diabetes, 2003. 52(11):26752683.
Loftus, T.M., et al., Reduced food intake and body Mobbs, C.V., et al., Mining microarrays for meta-
weight in mice treated with fatty acid synthase bolic meaning: Nutritional regulation of hypo-
inhibitors. Science, 2000. 288(5475):23792381. thalamic gene expression. Neurochem Res, 2004.
Lopez, M., etal., Peripheral tissue-brain interactions 29(6):10931103.
in the regulation of food intake. Proc Nutr Soc, Mobbs, C.V., et al., Low-carbohydrate diets cause
2007. 66(1):131155. obesity, low-carbohydrate diets reverse obe-
Lu, D., etal., Agouti protein is an antagonist of the sity:Ametabolic mechanism resolving the para-
melanocyte-stimulating-hormone receptor. dox. Appetite, 2007. 48(2):135138.
Nature, 1994. 371(6500):799802. Montague, C.T., et al., Congenital leptin deficiency
Ludwig, D.S., etal., Melanin-concentrating hormone is associated with severe early-onset obesity in
overexpression in transgenic mice leads to obe- humans. Nature, 1997. 387(6636):903908.
sity and insulin resistance. J Clin Invest, 2001. Moran, C.P. and F. Shanahan, Gut microbiota and
107(3):379386. obesity: role in aetiology and potential thera-
Luquet, S., etal., NPY/AgRP neurons are essential for peutic target. Best Pract Res Clin Gastroenterol,
feeding in adult mice but can be ablated in neo- 2014. 28(4):585597.
nates. Science, 2005. 310(5748):683685. Morgan, T.H., The origin of five mutations in eye
Maes, H.H., M.C. Neale, and L.J. Eaves, Genetic color in drosophila and their modes of inheri-
and environmental factors in relative body tance. Science, 1911. 33(849):534537.
weight and human adiposity. Behav Genet, 1997. Morgan, T.H., The origin of nine wing mutations in
27(4):325351. Drosophila. Science, 1911. 33(848):496499.
584 Part IV:Homeostatic Therapies

Morgan, W.C., The relation of the lethal yellow (Ay) Poplawski, M.M., J.W. Mastaitis, and C.V. Mobbs,
gene to pulmonary tumor formation and obesity Naloxone, but not valsartan, preserves responses
in an inbred strain of mice. J Natl Cancer Inst, to hypoglycemia after antecedent hypoglyce-
1950. 11(2):263268. mia:Role of metabolic reprogramming in coun-
Mori, A. and J.H. Perry, Electron microscopic obser- terregulatory failure. Diabetes, 2011. 60(1):3946.
vations of early changes in the ventromedial Powley, T.L. and J.C. Prechtl, Gold thioglucose selec-
hypothalamus of the mouse following gold- tively damages dorsal vagal nuclei. Brain Res,
thioglucose. Folia Psychiatr Neurol Jpn, 1967. 1986. 367(12):192200.
21(4):291306. Pramyothin, P. and L. Khaodhiar, Metabolic
Mozaffarian, D., etal., Changes in diet and lifestyle syndrome with the atypical antipsychotics.
and long-term weight gain in women and men. N Curr Opin Endocrinol Diabetes Obes, 2010.
Engl J Med, 2011. 364(25):23922404. 17(5):460466.
Muoio, D.M., etal., Peroxisome proliferator-activated Qian, S., et al., Neither agouti-related protein nor
receptor-alpha regulates fatty acid utilization in neuropeptide Y is critically required for the reg-
primary human skeletal muscle cells. Diabetes, ulation of energy homeostasis in mice. Mol Cell
2002. 51(4):901909. Biol, 2002. 22(14):50275035.
Myers, M.G., Jr. and D.P. Olson, Central nervous Qiu, J., et al., Transgenic mice overexpressing
system control of metabolism. Nature, 2012. leptin accumulate adipose mass at an older,
491(7424):357363. but not younger, age. Endocrinology, 2001.
Obici, S., etal., Inhibition of hypothalamic carnitine 142(1):348358.
palmitoyltransferase-1 decreases food intake and Qu, D., etal., A role for melanin-concentrating hor-
glucose production. Nat Med, 2003. 9(6):756761. mone in the central regulation of feeding behav-
Ollmann, M.M., et al., Antagonism of cen- iour. Nature, 1996. 380(6571):243247.
tral melanocortin receptors in vitro and in Rios, M., etal., Conditional deletion of brain-derived
vivo by agouti-related protein. Science, 1997. neurotrophic factor in the postnatal brain leads
278(5335):135138. to obesity and hyperactivity. Mol Endocrinol,
Olshansky, S.J., et al., A potential decline in life 2001. 15(10):17481757.
expectancy in the United States in the 21st cen- Roberts, J.L. and E. Herbert, Characterization
tury. N Engl J Med, 2005. 352(11):11381145. of a common precursor to corticotropin and
Oomura, Y., etal., Glucose and osmosensitive neu- beta-lipotropin:Identification of beta-lipotropin
rones of the rat hypothalamus. Nature, 1969. peptides and their arrangement relative to cor-
222(190):282284. ticotropin in the precursor synthesized in a
Oster, G., etal., Lifetime health and economic ben- cell-free system. Proc Natl Acad Sci USA, 1977.
efits of weight loss among obese persons. Am J 74(12):53005304.
Public Health, 1999. 89(10):15361542. Roberts, J.L. and E. Herbert, Characterization
Overweight, obesity, and health risk. National of a common precursor to corticotropin and
Task Force on the Prevention and Treatment of beta-lipotropin: Cell-free synthesis of the pre-
Obesity. Arch Intern Med, 2000. 160(7):898904. cursor and identification of corticotropin pep-
Owen, J.A., Jr., W. Parson, and K.R. Crispell, tides in the molecule. Proc Natl Acad Sci USA,
Dietary dilution studies in gold thioglucose 1977. 74(11):48264830.
induced obesity in the mouse. Metabolism, 1953. Roth, C., et al., Hyperphagia in children with cra-
2(4):362366. niopharyngioma is associated with hyperlepti-
Peeters, A., etal., Obesity in adulthood and its conse- naemia and a failure in the downregulation of
quences for life expectancy:Alife-table analysis. appetite. Eur J Endocrinol, 1998. 138(1):8991.
Ann Intern Med, 2003. 138(1):2432. Ruderman, N.B., et al., Malonyl-CoA, fuel sensing,
Pijl, H., Obesity: Evolution of a symptom of afflu- and insulin resistance. Am J Physiol, 1999. 276(1
ence. Neth J Med, 2011. 69(4):159166. Pt 1):E1E18.
Poggioli, R., A.V. Vergoni, and A. Bertolini, ACTH- Ryan, K.K., etal., A role for central nervous system
(124) and alpha-MSH antagonize feeding PPAR-gamma in the regulation of energy bal-
behavior stimulated by kappa opiate agonists. ance. Nat Med, 2011. 17(5):623626.
Peptides, 1986. 7(5):843848. Sanacora, G., etal., Increased hypothalamic content
Poplawski, M.M., et al., Hypothalamic responses of preproneuropeptide Y messenger ribonucleic
to fasting indicate metabolic reprogramming acid in genetically obese Zucker rats and its regu-
away from glycolysis toward lipid oxidation. lation by food deprivation. Endocrinology, 1990.
Endocrinology, 2010. 151(11):52065217. 127(2):730737.
Malignant Brain Cancer Management with Metabolic Therapy 585

Schwartz, E. and C.V. Mobbs, Hypothalamic BDNF Swartz, T.D., etal., Preserved adiposity in the Fischer
and obesity: Found in translation. Nat Med, 344 rat devoid of gut microbiota. FASEB J, 2013.
2012. 18(4):496497. 27(4):17011710.
Schwartz, M.W., etal., Specificity of leptin action on Takaya, K., etal., Molecular cloning of rat leptin recep-
elevated blood glucose levels and hypothalamic tor isoform complementary DNAsidentification
neuropeptide Y gene expression in ob/ob mice. of a missense mutation in Zucker fatty (fa/
Diabetes, 1996. 45(4):531535. fa) rats. Biochem Biophys Res Commun, 1996.
Schwartz, M.W., et al., Leptin increases hypotha- 225(1):7583.
lamic pro-opiomelanocortin mRNA expression Tartaglia, L.A., et al., Identification and expression
in the rostral arcuate nucleus. Diabetes, 1997. cloning of a leptin receptor, OB-R. Cell, 1995.
46(12):21192123. 83(7):12631271.
Schwartz, M.W., et al., Is the energy homeostasis Thompson, D., etal., Lifetime health and economic
system inherently biased toward weight gain? consequences of obesity. Arch Intern Med, 1999.
Diabetes, 2003. 52(2):232238. 159(18):21772183.
Sherwood, N.E., etal., Enhancing long-term weight Thornton, J.E., et al., Regulation of hypothalamic
loss maintenance:2year results from the Keep It proopiomelanocortin mRNA by leptin in ob/ob
Off randomized controlled trial. Prev Med, 2013. mice. Endocrinology, 1997. 138(11):50635066.
56(34):171177. Tsujii, S. and G.A. Bray, Acetylation alters the feed-
Shimada, M., et al., Mice lacking melanin- ing response to MSH and beta-endorphin. Brain
concentrating hormone are hypophagic and Res Bull, 1989. 23(3):165169.
lean. Nature, 1998. 396(6712):670674. Unger, T.J., etal., Selective deletion of BDNF in the
Shugart, H.A., Shifting the balance: The contempo- ventromedial and dorsomedial hypothalamus of
rary narrative of obesity. Health Commun, 2011. adult mice results in hyperphagic behavior and
26(1):3747. obesity. J Neurosci, 2007. 27(52):1426514274.
Smart, J.L., V. Tolle, and M.J. Low, Glucocorticoids Wang, C., et al., Brain-derived neurotrophic fac-
exacerbate obesity and insulin resistance in tor (BDNF) in the hypothalamic ventromedial
neuron-specific proopiomelanocortin-deficient nucleus increases energy expenditure. Brain Res,
mice. J Clin Invest, 2006. 116(2):495505. 2010. 1336:6677.
Sternson, S.M., G.M. Shepherd, and J.M. Friedman, Woods, S.C. and L.J. Nolan, The insulin story: A25-year
Topographic mapping of VMH arcuate nucleus perspective. Appetite, 1997. 28(3):281282.
microcircuits and their reorganization by fast- Xu, B., etal., Brain-derived neurotrophic factor regu-
ing. Nat Neurosci, 2005. 8(10):13561363. lates energy balance downstream of melanocortin-4
Sun, Y., etal., Characterization of adult ghrelin and receptor. Nat Neurosci, 2003. 6(7):736742.
ghrelin receptor knockout mice under positive Yeo, G.S., et al., A frameshift mutation in MC4R
and negative energy balance. Endocrinology, associated with dominantly inherited human
2008. 149(2):843850. obesity. Nat Genet, 1998. 20(2):111112.
Surwit, R.S., etal., Diet-induced type II diabetes in Yi, C.X., T. Scherer, and M.H. Tschop, Cajal revis-
C57BL/6J mice. Diabetes, 1988. 37(9):11631167. ited:Does the VMH make us fat? Nat Neurosci,
Surwit, R.S., etal., Control of expression of insulin 2011. 14(7):806808.
resistance and hyperglycemia by different genetic Zhang, Y., et al., Positional cloning of the mouse
factors in diabetic C57BL/6J mice. Diabetes, obese gene and its human homologue. Nature,
1991. 40(1):8287. 1994. 372(6505):425432.
31
Autism Spectrum Disorder and Homeostasis
S U S A N A . M A S I N O , J E S S I C A A . F O R T I N , M I C H E L L E I . M U R P H Y,
L I S A S A A , A N D D AV I D N . R U S K I N

OVERVIEW was one of several differential diagnoses based


on social deficits, impaired communication,
Diverse Symptoms and Disrupted and restricted and repetitive behaviors and
Homeostasis interests. With the revision, autistic disorder
Autism spectrum disorder (ASD) affects chil- and the other possible diagnoses with these core
dren and adults worldwide. Approximately symptoms are all grouped under ASD; thus pre-
1 in 68 children (diagnosed at age 8)have ASD, vious differential diagnoses no longer exist in
with a higher prevalence in males (1 in 42)than DSM V:Aspergers syndrome was removed, and
females (1 in 189; Centers for Disease Control pervasive developmental disorder falls under
and Prevention, 2014). Prevalence in adults the umbrella ofASD.
was recently estimated to be 1 in 100 (Brugha The common core features of ASD accord-
et al., 2011). Current overall ASD rates repre- ing to DSM V are social communication deficits
sent a roughly 30-fold increase from the classic and fixated interests and repetitive behaviors.
survey in 1966 (Lotter, 1966), which reported The second major change, which has been less
1 in ~2,200. This dramatic change in preva- controversial, is bringing together social deficits
lence over time may be influenced by diagnosis and language problems into the single category
parameters and changes in disorder classifica- of social communication problems. (The other
tion, but evidence is mounting that suggests it major symptom category, restricted and repeti-
may partly reflect a real increase due to a variety tive behavior, remains intact.) This change is
of environmental factors. Risk factors for ASD based on the opinion that language difficulties
are known to include genetic and environmen- in people with ASD are limited to the social
tal components; even the genetic component is domain. Examples include difficulties in recip-
complex and extremely heterogeneous (Persico rocal conversation, delays in language devel-
and Napolioni, 2013), thus making the gene-by- opment, adherence to specific nonfunctional
environment interaction contributing to ASD a routines, and intense preoccupations. Often
staggering matrix of unknown and uncontrolla- these behavioral impairments lead to decreased
ble variables in the human population. social support and a lack of meaningful relation-
For the majority of children with ASD, espe- ships with peers (Kim and Lord, 2012). People
cially those diagnosed well after age 2, many of with ASD experience multiple behavioral and
the core symptoms are difficult to remediate. physiological symptoms, which range from mild
With the cause(s) and underlying mechanism(s) to severe.
unknown, finding successful treatments can The great heterogeneity of symptoms and
seem like a circular problem. Commonly severity among individuals with ASD makes it
used tools for the diagnosis of ASD are the difficult to identify specific causal mechanisms
Diagnostic and Statistical Manual of Mental and etiologies that apply to all or even most
Disorders (DSM) of the American Psychiatric patients with the disorder. Known genetic factors
Association (2013) and the International account for ~10% to 20% of ASD cases, leaving
Statistical Classification of Diseases code of the a large fraction of patients with uncertain eti-
World Health Organization (2010). Two major ologies (Betancur, 2011). ASD is recognized to
changes occurred in the recently adopted ver- be highly heritable, however, and a recent pop-
sion V of the DSM. Previously, autistic disorder ulation study involving over 2 million people
Autism Spectrum Disorders and Homeostasis 587

established that heritability provides about 50% COMORBIDITIES


of the risk (Sandin et al., 2014). Most of these ASD is often accompanied by comorbid
identified genetic factors are common, however, physiological and psychological conditions,
and/or common to other disorders and comor- including epilepsy, aggression, anxiety, gastro-
bidities; in most cases it appears to be the com- intestinal problems, altered sensory experiences,
plex interactions among these genes and with the motor impairments, and sleep disturbances
environment that produces an ASD diagnosis. (Argyropoulos et al., 2012). Diverse and com-
Differences and similarities in ASD due to gen- mon comorbidities suggest that multiple aspects
der, environmental factors, prenatal influences, of dysregulation can converge on a common
genetics, and physiology may eventually reveal behavioral phenotype, and thus some aspects
mechanisms that lead to the disorder and thus of dysregulation may be shared widely among
potential interventions and therapeutic targets. patients with ASD regardless of the underlying
Given this complexityunlikely to be resolved cause of the disorder. Recently, in an attempt
into a common mechanism or therapeutic to understand the interactions between ASD
targeta focus on restoring homeostasis could and comorbidities and among common comor-
promote broad-based improvement regardless of bidities, patients with ASD were diagnosed into
the underlying cause(s) ofASD. several comorbid clusters based on electronic
Depending on severity, an ASD diagnosis records: (a) a seizure cluster; (b) a psychiatric
can be devastating and result in lifelong dis- cluster; (c)a cluster that included gastrointestinal
ability. With a disorder so common and per- disorders, auditory disorders, and infection, and
sistent, and increasing in prevalence, there are (d)an unresolved cluster containing most (~90%)
major economic implications. Expenditures patients (Doshi-Velez et al., 2014). Nevertheless,
for ASD sufferers have been estimated to be at this classification scheme underscores the broad
least two-fold to six-fold over controls (Barrett symptomatology beyond the behavioral criteria
etal., in press; Peacock etal., 2012; Wang etal., that define an ASD diagnosis. Here we outline
2012). Total costs increase with severity (Lavelle common comorbidities and focus on opportuni-
etal., 2014), as do out-of-pocket costs (Raz etal., ties for neural, immune, and metabolic homeo-
2013). Annual cost per ASD patient has been stasis that may in turn address multiple aspects
estimated at $35,000 (Horlin et al., 2014), and ofASD.
total cost per patient over a lifetime as $2mil-
lion to $3million (Buescher etal., 2014; Ganz, Neural
2007); total annual expenditure in the United ASD is typically thought of as a developmental
States is roughly $8 billion (Trasande and Liu, neurological disorder and is diagnosed based
2011). Around the world, practical, effective, on behavioralthough substantial evidence is
and economically viable therapies are needed. mounting for addressing comorbid metabolic
Alongside new therapies, prevention by avoid- and immune dysfunction, discussed in more
ing environmental risk factors may become detail later. The most common comorbidities are
an important strategy in addressing rising in the neural realm and range from seizure dis-
incidence. orders, sensory-motor abnormalities, psychiatric
Herein we present a viewpoint that frames conditions, and disrupted sleep. A high comor-
ASD treatment in the context that core ASD bidity of epilepsy and abnormal EEG, motor
symptoms are often difficult to treat and are often delay and dysfunction, and sensory sensitivity all
accompanied by neural, immune, and/or meta- point to abnormal neural activity. Hyperactivity
bolic dysregulationall with diverse underlying and aggression are also common, and a variety of
cause(s) that may change over time even within psychoactive drugs are prescribed with the hope
individuals. Targeting systemic dysregulation that core symptoms might be alleviated along
with strategies that restore homeostasis may with these comorbid symptoms.
be an effective approach. Therapies shown to Epilepsy is a frequent comorbidity, with
improve behavioral symptoms may be effective recent estimates ranging from 7% to 21% of chil-
because they restore homeostasis at least partially dren diagnosed with ASD also having a diag-
and thus may help reveal key underlying factors nosis of epilepsy (Amiet et al., 2013; Carlsson
in ASD. In keeping with this perspective, we pro- et al., 2013; Cuccaro et al., 2012; Doshi-Velez
vide examples of homeostatic dysregulation in et al., 2014; Kantzer et al., 2013); this range of
ASD, current and potential treatment strategies, incidence is undoubtedly due to sampling from
and future research. different populations. An abstract presented at
588 Part IV:Homeostatic Therapies

the International Meeting for Autism Research mutations in genes encoding ion channels, such
in 2014 (Supekar et al., 2014) presented a much as the sodium channel genes SCN1A and SCN2A,
higher rate based on analyzing clinical data of have been identified in epilepsy and also in ASD
over 1 million patients at Stanford University (Betancur, 2011). Additional gene mutations, such
Medical Center: epilepsy was diagnosed in 41% as in protocadherin 19 and those associated with
of ASD patients up to18 years old. Although metabolic disorders and intellectual disabilities,
the rate remained high, there was a decrease in have also been identified in patients with comor-
comorbid seizures in adults with ASD (~24% bid ASD/epilepsy (Betancur and Coleman,2012).
in ASD patients between the ages of 18 and 35 There is a rich array of mouse models with
and ~15% in those over 35). Conversely, a recent varying relationships between ASD and seizures.
analysis screened children diagnosed with epi- Some mouse ASD models are frankly epilep-
lepsy for comorbid behavioral disorders and tic (Peagarikano etal., 2011; Zhao etal., 2010);
found that 21% of epileptic children also met some have heightened seizure susceptibility but
the diagnostic criteria for autism (Reilly et al., no spontaneous seizures (Musumeci etal., 2000;
2014). In a cluster analysis of pediatric/adolescent Pineda et al., 2013; Qiu et al., 2009; Sala et al.,
ASD aimed at comorbid epilepsy specifically, 2011); some have little or no change in seizure
patients fell into five clusters, one of which (5% susceptibility (Radyushkin et al., 2009; Ruskin
of patients) had a high (29%) epilepsy rate, early et al., 2013). Conversely, some mouse models of
ASD diagnosis, sensory abnormalities, and high epilepsy also have autistic symptoms (Greco etal.,
repetitive behaviors, whereas two other clusters 2013; Meidenbauer et al., 2011). Interestingly,
of high-functioning individuals (38% of patients) mice in these latter models often become epi-
had the lowest (8%) epilepsy rates (Cuccaro leptic after autistic symptoms are evident and so
etal.,2012). are time-variant for the comorbidity. Many ASD
Ultimately, estimates of the comorbid- models remain uncharacterized or incompletely
ity between epilepsy and ASD are necessarily characterized for seizures or seizure suscepti-
conservativeASD might not be diagnosed once bility; conversely, many mice used as models of
a child has a primary diagnosis of epilepsy, and, in epilepsy or with a lower seizure threshold have
children diagnosed with ASD, seizures could be never been tested for symptoms of autism. As a
occurring without clear convulsions or impair- whole, these animal models represent an expand-
ment of consciousness. Febrile seizures and sei- ing research landscape in which core symptoms
zures due to acute trauma, infection, or metabolic of ASD can be precipitated by single-gene muta-
illness are not diagnosed as epilepsy because they tions, complex gene interactions, and/or environ-
have a known provocation (Tuchman and Rapin, mental factors. Perhaps the best animal models to
2002). However, seizures due to these events may test treatment efficacy combine a complex genetic
impact brain homeostasis at a subclinical level predisposition with epigenetic prenatal and post-
and cause ongoing microseizures undetectable natal environmental factors.
behaviorally or with a scalp EEG (Stead et al., Diverse additional comorbidities involving
2010). For example, febrile seizures increase the the nervous system are also often associated with
risk of future seizures (Berg, 2008; Mohebbi ASD. Arecent review concluded that 30% to 60%
et al., 2004), suggesting the initial seizures had of autistic children use at least one psychotropic
lasting effects on neural activity and/or seizure medication (Siegel, 2012), and it was estimated
threshold. These seizure-inducing events may previously that up to 70% of autistic children
push a vulnerable central nervous system out of with intellectual disabilities were prescribed at
homeostasis and into a ASD diagnosis. least one psychotropic medication (Tsakanikos
Seizures represent dysregulated neuronal et al., 2006). The range of coexisting neuropsy-
activity, and diverse seizure types are associated chiatric diagnoses includes seizure disorders,
with ASD, including complex partial, atypical as noted previously, as well as attention deficit/
absence, myoclonic, and tonic-clonic (Tuchman hyperactivity disorder (60% have poor attention
and Rapin, 2002). Because the rate of epilepsy and concentration), anxiety disorder (up to 74%
in children in the general population is only 2% have anxiety or significant fears), affective dis-
to 3% (Amiet et al., 2013; Kohane et al., 2012; orders, obsessive compulsive disorder (37% have
Mouridsen et al., 2011), there is strong indica- obsessive phenomena), and Tourettes disorder
tion that common underlying factors influence (Tsai, 1999). Up to 43% of people with ASD also
this comorbidity. Regarding excessive excitability, have a history of self-injury (Tsai,1999).
Autism Spectrum Disorders and Homeostasis 589

People with ASD experience a range of (Horbinski and Chu, 2005), and mitochondrial
intellectual, sensory, and motor impairments. dysfunction is linked directly to a number of
With the recent inclusion of Aspergers syn- neurological conditions (Garcia-Escudero et al.,
drome within the diagnosis of ASD (disputed 2013; Maruszak and Zekanowski, 2011; Pathak
by some:Tsai and Ghaziuddin, 2014), IQ scores etal., 2013; Zhu and Chu, 2010). In addition, as
range greatly, but prior to the latest revision of mitochondrial-based energy production is criti-
the DSM, ~50% of autistic children were char- cally important for neurodevelopment, mito-
acterized as having an intellectual disability chondrial dysfunction might contribute to the
(IQ<70; Charman et al., 2011). Sensory abnor- developmental pathology of ASD (Haas,2010).
malities include severe reactions to loud or unu- In a comprehensive literature review exam-
sual noises, extreme disturbances with certain ining mitochondrial dysfunction, children
tactile sensations, and resistance to wearing with ASD had a 5.0% prevalence rate while
footwear (Kim and Lord, 2012). Children with the general population had a rate of approxi-
ASD also experience motor impairments (Hilton mately 0.01%, making it the most common
et al., 2014), with up to one-third experiencing metabolic abnormality associated with autism
delays in reaching motor milestones (Weissman (Frye and Rossignol, 2011; Rossignol and Frye,
etal.,2008). 2012). However, this prevalence rate of mito-
Sleep disorders are also common in children chondrial disorders could even be underesti-
with ASD and contribute to a range of other mated due to differences in clinical techniques
impairments. Sleep abnormalities include late (Giulivi et al., 2010). In a study comparing 80
sleep onset, sleep maintenance discontinuity, ASD and 73 control children, the children
and early morning awakening (Kohane et al., with ASD had decreased antioxidant abilities
2012; Polimeni etal., 2005; Sivertsen etal., 2012). and increased oxidative stress (James et al.,
When children with ASD experience psychiatric, 2006). Furthermore, children with ASD showed
sensorimotor, and sleep disordersin addition lower serum carnitine and pyruvate levels and
to cognitive and behavioral abnormalities asso- increased ammonia and alanine levels, further
ciated with ASDit further decreases quality of suggesting a mitochondrial defect (Oliveira
life and increases their need for therapy. etal., 2005). We currently lack a detailed mech-
anistic understanding of the role of mitochon-
Metabolic dria in ASD; however, in addition to regulating
Metabolic abnormalities are prevalent in the cellular proliferation and differentiation, these
autistic population. Gastrointestinal problems organelles are also involved in other aspects
are also quite common (Doshi-Velez etal., 2014; of brain function such as neuroinflammation,
Ibrahim etal., 2009; Wang etal., 2011)and might modulating GABA activity, and maintaining
relate to a disordered gut microbial community calcium homeostasisall implicated in ASD
(De Angelis et al., 2013; Williams et al., 2011; (Breitenkamp etal., 2014; Depino, 2013; Fatemi
Williams et al., 2012). There are also distur- etal., 2009a; Fatemi etal., 2009b).
bances in specific metabolites (El-Ansary et al., It is now well established that metabolic
2010; James et al., 2006; Page and Coleman, abnormalities are common with chronic neu-
2000; Spilioti et al., 2013) and enzymes (Marie rological conditions, particularly at the cellular
etal., 2002; Muratore etal., 2013; Sivendran etal., level, but translational research leveraging this
2004), often indicating impaired redox homeo- information into new therapies is lacking and
stasis and reduced methylation capacity. Some of is becoming increasingly urgent. ASD is not
these may be causative for rather than comorbid unique among neurological disorders as many
with behavioral/cognitive autistic symptoms. have comorbid metabolic dysfunction, yet in
Mitochondrial dysfunction is among the ASD more widespread and systemic metabolic
most common metabolic comorbidities (Adams changes are more prevalent; a growing number
et al., 2011b; El-Ansary et al., 2010; Oliveira of reviews conclude that mitochondrial dysfunc-
etal., 2005), and multiple lines of evidence from tion can lead to autistic symptoms (Dhillon etal.,
clinical, genetic, and biochemical studiesin 2011; Frye and Rossignol, 2011; Giulivi et al.,
humans and animal modelssupport a role for 2010; Hagberg et al., 2014; Legido et al., 2013;
mitochondrial dysfunction in ASD. The mito- Rossignol and Frye, 2012; Villafuerte, 2011). This
chondrion generates cell adenosine triphosphate remains an unexplored area of research whereby
(ATP) and integrates multiple signaling cascades improving mitochondrial function could help
590 Part IV:Homeostatic Therapies

restore metabolic homeostasis and reduce diverse (Atladttir etal., 2009). Additionally, about 12%
symptoms and comorbidities. of mothers of children with ASD have antifetal
brain antibodies in their serum, which is sig-
Immunological nificantly higher than in mothers of typical chil-
Many of the organ systems in ASD patients dren (Careaga etal., 2010). The role of maternal
show inflammation-related changes in immune- immune and metabolic environment in utero
related proteins and ribonucleic acids (Depino, has received increasing attention and is one area
2013). In the brain and cerebrospinal fluid, an where there is a particularly high correlation
inflammatory-like state was quantified by ele- between clinical evidence and behavioral and
vated cytokines and activated microglia and physiological outcomes in experimental models
astrocytes in postmortem ASD brains; notably, ofASD.
the age range of the subjects was 5 to 44 years
old, suggesting that this immune-activated state ETIOLOGY OFAUTISM
starts early and can be lifelong (Vargas et al., SPECTRUM DISORDERS
2005). Elevated cytokine levels were also found Data about ASD etiology has increased
in living ASD children aged 3 to 10years (Vargas dramaticallyyet, paradoxically, for most
etal., 2005). These findings were again supported patients their particular etiology remains murky
by additional studies on the same postmortem or completely unknown. We do not attempt
brains, as well as new autistic brain samples (Chez a thorough review of this complex topic here,
etal., 2007; Morgan etal., 2010). Microarray stud- and where possible we relate to our overarching
ies have found that in ASD brains there is a dys- theme of homeostasis.
regulation of immune-related genes (cytokines
and chemokines; Lintas etal.,2012). Genetic
It has been suggested that the inflammatory- In recent decades the known genetic basis for
like state in the central nervous system may ASD has improved as most known medical
be related to abnormalities in the peripheral conditions associated with ASD have had their
immune system, but the connection between genetic underpinnings discovered. Thus the field
peripheral immune abnormalities and the has progressed over two decades from known
altered behavior in ASD is not well understood medical conditions being associated with ~10%
(Careaga et al., 2010). Compared to controls, of ASD cases (Rutter et al., 1994) to known
as well as nonautistic children with develop- genetic conditions being associated with up to
mental disabilities, autistic children exhibited 20% of ASD cases (Betancur, 2011). This is still a
elevated cytokines and chemokines (including relatively low number, and genetics are undeni-
interleukin-1, IL-6, IL-8, and IL-12p40) in blood ably important:twin studies have always shown
plasma. Additionally, these immune factors were that ASD has a very high heritability. Studies of
positively associated with impaired communi- monozygotic twins indicate heritability rates of
cation and behavioral abnormalities (Ashwood >70%, and these might be underestimates as even
et al., 2011a, 2011b). Furthermore, other stud- monozygotic twins are not necessarily geneti-
ies found that in vitro blood mononuclear cells cally identical due to the possibility of different
show altered cytokine responses (Enstrom etal., within-pair copy number variations and different
2010; Jyonouchi et al., 2005; Patterson, 2009). X chromosome inactivation patterns (Posthuma
Genetic susceptibility to environmental factors and Polderman,2013).
could bias the subject toward both brain and The genetic basis for ASD is clearly com-
immune-related abnormalities simultaneously; plex, and disorders in over 100 specific genes
ASD-associated gene candidates regulate both have been associated with ASD (Betancur, 2011).
brain and immune system development/function These genes code for a wide array of protein
(Careaga etal., 2010). Despite the need for more productsenzymes, ion channels, transcription
research, it is well known that elevated peripheral factors, transmitter and growth factor recep-
cytokines can cause dramatic changes in behav- tors, cell adhesion molecules, and so onand the
ior (Patterson,2009). genetically based ASD cases have a wide range of
A large epidemiologic study found that incidence, down to published single case studies.
some autoimmune diseases (rheumatoid arthri- There are also over 40 genomic disorders (i.e., copy
tis, celiac disease, and type 1 diabetes) are number variations in which multigene sequences
more common in mothers of autistic children are deleted or duplicated) that are associated with
Autism Spectrum Disorders and Homeostasis 591

ASD (Betancur, 2011). In addition to all this het- MIA-induced effects on offspring behavior are
erogeneity, there is also varying penetrance of the lacking in animals lacking IL6, can be prevented
genetic/genomic disorder to a diagnosed autistic by maternal injection of anti-IL6 antibodies and
phenotype, ranging from high (e.g., fragile X can be replicated by maternal IL6 injection in lieu
syndrome, tuberous sclerosis) to low (Duchenne of poly-(I:C) injection (Smith et al., 2007); thus
muscular dystrophy, noonan syndrome). Even a augmented IL6 levels during gestation seem suffi-
complete understanding of genetic contributions cient. These data seem to rule out a prior hypoth-
is unlikely to lead to broadly effective therapies esis that detrimental effects on offspring were due
for ASD in the near future. to diminished IL6 due to antipyretic treatment
during maternal viral infectioninduced fever
Environmental (Torres, 2003), as IL6 is necessary for several
The role of the environment in ASD includes aspects of nervous tissue development. It seems
many factors, including the prenatal environ- very likely that, as with so many signaling agents,
ment. The perinatal environment and outcomes there is a balanced and optimal IL6 level during
are also factors: perinatal distress and/or low gestation (Marz et al., 1999), as well as an opti-
birth weight associates with ASD diagnosis mal balance between IL6 and other interleukins
(Froehlich-Santino et al., 2014; Gardener et al., (Meyer etal.,2008).
2011; Losh et al., 2012). Child vaccination has Regarding factors from the outside environ-
repeatedly been shown to have no association ment, one multiyear study found an associa-
with ASD (Taylor et al., 2014). However, mater- tion between incidence of ASD and proximity
nal immune activation (MIA)due to bacteria of mothers (particularly in the second and third
or virusesoccurring during gestation can lead trimesters) to agricultural pesticide use (Shelton
to modified behavior in the offspring even into etal., 2014), and children with ASD have signifi-
adulthood. The influence of outside environmen- cantly higher levels of heavy metals than con-
tal factors is challenging to dissect but may reveal trols (Adams et al., 2007) with exposure level
important contributing factors and vulnerabili- and symptom severity correlating (Adams etal.,
ties as discussedlater. 2013). Many other outside factors (e.g., dietary
MIA-induced inflammation triggers an supplements, anticonvulsants, air pollution,
increase of proinflammatory factors including drinking-water source, electromagnetic radia-
interleukins and activates immune cells in the tion, among others) have been postulated or are
decidua, bathing the fetus in proinflammatory being investigated. With so many factors, and a
compounds and antibodies and increasing the disorder with such complex etiology, causation
likelihood of the child developing ASD (Patterson, from the environment will be difficult to ascer-
2011b). For example, an epidemiological study of tain. Where possible, it would seem prudent to
over 10,000 ASD cases found that a viral infec- limit exposure to suspicious environmental fac-
tion in the mother during the first trimester, and tors, particularly in individuals who may be at
any infection (viral or bacterial) during the sec- higher risk such as those with a family history of
ond trimester, was associated with an ASD diag- ASD, prenatal or perinatal risk factor(s), or expo-
nosis in the child (Atladttir etal., 2010). Similar sure to a combination of environmental risk fac-
findings have been reported in more recent stud- tors that may be additive or synergistic.
ies (Atladttir etal., 2012; Lee etal., 2015; Zerbo
et al., in press). This clinical finding has been Epigenetic
replicated in rodents and recently in primates Epigenetic modification of DNA is a means by
(Bauman etal., 2014; Hsiao etal., 2012; Malkova which gene expression can be regulated with-
et al., 2012; Smith et al., 2007). Rodent models out any change in DNA sequence. This regu-
have been developed using, for instance, synthetic lation takes the form of DNA methylation,
agents that induce an immune response such as histone methylation, or histone acetylation.
poly-(I:C) (evokes antiviral-type response) and There is increasing interest in a possible epige-
lipopolysaccharide (evokes antibacterial-type netic component of ASD etiology, comporting
response). Offspring demonstrate the core symp- with changes in methylation capacity in autistic
toms of autism (Malkova etal., 2012; Smith etal., patients (Adams etal., 2011b; James etal., 2006;
2007), some with symptoms only in males (Xuan Muratore etal., 2013). Several studies have found
and Hampson, 2014), and also mitochondrial a significantly altered gene methylation pattern,
dysfunction (Giulivi et al., 2013). In rodents, or methylome, in ASD patients compared to
592 Part IV:Homeostatic Therapies

controls (Berko etal., 2014; Nardone etal., 2014), Pharmacological Approaches


even when the control subjects are the monozy- Pharmacological attempts at treatment have been
gotic twins of the patients (Wong et al., 2014). used with varying degrees of success. None of
As one example, changes in brain-derived neu- these were developed for ASD per se, and many
rotrophic factor (BDNF) have been associated deal only with comorbidities. In many cases, sei-
with ASD (Kasarpalkar etal., 2014), and changes zures can be controlled with anticonvulsant drugs
in BDNF gene expression and DNA methylation (although roughly one-third of ASD/epilepsy
have been associated with early life adversity patients have drug-refractory epilepsy: Sansa
in general (Jirtle and Skinner, 2007). New evi- etal., 2011), but even when these agents have been
dence suggests altered BDNF methylation status prescribed also to help irritability and aggression,
is similar in blood and brainthus offering the meta-analysis shows they are ineffective against
potential for a much-needed early biomarker comorbidities other than epilepsy (Hirota etal.,
(Kundakovic etal., in press). 2013). Hyperactivity, impulsiveness, and atten-
Epigenetic dysregulation correlates with tion problems can be controlled with stimulants
symptom severity (Wong etal., 2014); immune- acting on dopamine or norepinephrine systems
and synapse-related genes are heavily involved (Baribeau and Anagnostou, 2014; Cortese etal.,
(in accord with other evidence; Nardone et al., 2012; McCracken et al., 2014; Pearson et al.,
2014). Methylation problems appear in much 2013), though side effects can be worse in ASD
the same set of genes implicated in ASD through sufferers than in controls (Doyle and McDougle,
frank genetic dysfunction (Berko et al., 2014), 2012). Atypical neuroleptics can be helpful with
and epigenetics is a means by which a pre- or per- irritability and aggression, but again, adverse
inatal influence can cause long-term altered gene side effects can be prominent (Baribeau and
expression because changes in DNA methylation Anagnostou, 2014; Elbe and Lalani, 2012; Moyal
can be stable or very long-lasting (Tordjman etal., etal., 2014; Politte and McDougle, 2014). Anxiety
2014). Furthermore, the methylome is heritable, may be alleviated by serotonergic drugs (Vasa
such that the methylation status of a parent dur- etal.,2014).
ing gamete generation is passed on to offspring. There has been less success in pharmacologi-
This is one possible explanation for the finding cally ameliorating the core autistic symptoms,
that paternal age is an ASD risk factor (Hultman with most attention given to repetitive and ste-
etal., 2011), even though fathers obviously can- reotyped behavior. After years of use, the con-
not contribute to the gestational environment. sensus is that serotonin reuptake inhibitors are
Many nongenetic parental risk factors could be poorly effective against repetitive behavior, espe-
similary passed on (Mouridsen et al., 2007), as cially in children (Baribeau and Anagnostou,
could epigenetic changes associated with mater- 2014; Doyle and McDougle, 2012; Williams etal.,
nal aging, also a risk factor (Berko etal.,2014). 2013); however, atypical antipsychotics have
emerged as useful in limiting these symptoms
H O M E O S TAT I C T R E AT M E N T S (Ching and Pringsheim, 2012; Dove etal., 2012),
FORA SPECTRUM and, as noted, adverse side effects of antipsychot-
OFSYMPTOMS ics can be dose-limiting. For social behavior,
A perspective that most cases of ASD are related some individual studies have noted improve-
to disrupted neural, metabolic, and/or immuno- ment possibly relating to successful alleviation
logical homeostasisrather than highly selective of comorbidities (e.g., Pearson et al., 2013), but
pharmacological targetsfocuses treatments there is poor meta-analytical evidence of any
more on broad-based strategies to alleviate multi- drug treatment significantly promoting prosocial
ple symptoms. Certainly ongoing research efforts behavior (Farmer etal., 2013). However, experi-
need to focus simultaneously on both mechanis- mental clinical research is in progress with alter-
tic underpinnings and symptom relief to bet- native agents such as oxytocin, D-cycloserine,
ter understand and treat ASD: therapies shown memantine, and bumetanide (Baribeau and
to help with symptoms of ASD could also help Anagnostou, 2014; Doyle and McDougle, 2012);
reveal underlying mechanisms. Here we outline some of these may offer more broad homeo-
some evidence as examples, focusing on treat- static effects. For pharmacological treatments
ments that are established as safeand perhaps of either core symptoms or comorbidities, there
with multiple health benefitsand offer sugges- have been calls for more placebo-controlled ran-
tions for further research into related treatment domized trials, larger sample sizes, and inclu-
opportunities. sion of quality-of-life measures (Mohiuddin and
Autism Spectrum Disorders and Homeostasis 593

Ghaziuddin, 2013; Politte and McDougle, 2014; SYMPTOMS OF AUTISM


Vasa etal.,2014).
Here we take special notice that evidence of Seizures
Repetitive
behaviors
purinergic dysfunction in ASD: purine abnor-
malities have been associated with autism but
until recently were not linked to the core symp- Disrupted
toms of the disorder. We previously published sleep Anxiety
Decreased
a hypothesis, a review, and initial data relating Seizure-reducer repetitive behaviors
ASD and the purine adenosine (Masino et al.,
2011a; Masino etal., 2011c). Adenosine is an ubiq- Sleep-promoter Decreased anxiety
uitous purine nucleoside that links metabolism
to neuronal function (Dunwiddie and Masino,
2001; Fredholm, 2011). Its recognized roles have Adenosine
evolved significantly from initially being termed
a retaliatory metabolite (Newby etal., 1985) FIGURE 31.1: The relationship between adenosine
i.e. a neuromodulator participating in cell energy and symptoms of autism. Adenosine has multiple
cycles, with a predominant function to inhibit impacts on physiology and behavior. Increased aden-
synaptic transmission and limit energy demand osine would be predicted to ameliorate multiple core
during conditions with a net energy loss due to and comorbid symptoms of autism.
dephosphorylation of ATP. Since this initial and Adapted from Masino etal. (2011c).
still valid characterization as a distress signal
(Fredholm, 2007), it has been recognized that
adenosine is a powerful endogenous antiseizure a metabolic treatment that increases adenosine
molecule (Dunwiddie, 1999; Fedele et al., 2006; based on recent in vivo and in vitro data (see later
Kawamura et al., 2014), effective in stopping all discussion).
types of seizures, and ever-present as a dynamic Another purine, the cellular fuel molecule
regulator of neuronal activity based on small and adenine nucleotide ATP, is a topic of recent
physiological changes in the internal and external rodent research and a pending clinical trial with
neuronal milieu. Regarding ASD, diverse stimuli the ATP receptor blocker suramin. This drug has
that have been reported to ameliorate symptoms a century-long history of use in humans against
of ASD would all be predicted to increase aden- sleeping sickness; thus it is well characterized for
osine; in parallel, autistic symptoms would be side effects, may be appropriate for short-term
predicted to be reduced by increased adenosine use, and reversed numerous behavioral, bio-
(Masino etal., 2011a; Masino etal., 2009; Masino chemical, neuroanatomical, and metabolic dys-
etal., 2011c). Figure 31.1 is a schematic illustrat- functions in a mouse model of ASD (Naviaux
ing some aspects of the relationship between et al., 2014; Naviaux et al., 2013). These effects
symptoms of autism and effects of adenosine. were interpreted as due to reduced inflamma-
Directly targeting adenosine receptors has been tion, which would be of major homeostatic ben-
perennially challenging due to cardiovascu- efit in ASD. Some questions remain at this early
lar and other side effects. However, decades of stage: Is the effect generalizable to other mod-
research have yielded some new developments els of ASD? Is the effect truly mediated by ATP
(Korboukh etal., 2012; Luongo etal., 2012). For receptors, and which subtype(s) as suramin is not
example, Ken Jacobson has identified detailed a highly selective drug (Hohenegger etal., 1998)?
structural specificity for new agonists (Tosh etal., How does the drug affect behavior when it does
2012), which would have multiple clinical uses. not seem to reach portions of the brain beyond
Previous strategies identified adenosine amine the brainstem (Naviaux et al., 2014), and is the
congeners as potential clinically-useful receptor mechanistic effect peripheral, possibly on the gut
agonists (Von Lubitz et al., 1999). A compound microbiome or enteric nervous system activity?
extracted from an herb, N6-(4-hydroxybenzyl) Nevertheless, this is an exciting development and
adenine riboside, designated T111, has shown a new target.
dual effects of increasing adenosine signal- Overall, the pharmacological approaches
ing via actions at adenosine A2a receptors and available and used to date would not be expected
the ENT1 adenosine transporter (Huang et al., to have a broad homeostasis-restoring benefit for
2011). In addition to these and potential other persons with ASD. Ultimately, a less selective drug
pharmacological options, the ketogenic diet is might be a better approach to autismone that
594 Part IV:Homeostatic Therapies

has multiple effects, even some not well under- subpopulations of ASD to their optimal target
stood. Often these less selective or dirty drugs diets and supplements would be of great use. As
have fewer side effects and good safety profiles, the microbiome is apparently abnormal in ASD,
are less likely to produce tolerance, and might diets that specifically modulate the intestinal
represent a homeostatic approachironically flora could be of use. Regarding supplements, a
based on poor pharmacology. Such a drug meta-analysis concluded that there is little evi-
may already be approved by the Food and Drug dence for effectiveness of vitamin B6/magne-
Administration, and a screen that could deline- sium supplementation (Nye and Brice, 2005);
ate efficacy in autism would be a valuable tool. To randomized, placebo-controlled studies have
date, the pharmacological treatment of ASD and found no effect of omega-3 fatty acids, dimeth-
its associated comorbidities has identified some ylglycine, or digestive enzymes (James et al.,
effective drugs and potential therapeutic tar- 2011; Kern etal., 2001; Munasinghe etal., 2010),
gets, but much further study and advancement is but a vitamin/mineral supplement was signifi-
needed to develop specific treatments with lim- cantly helpful with some symptoms (Adams
ited side effects to pharmacologically treatASD. etal., 2011a). Many other supplements have been
tried in studies that were open label/not placebo
Metabolic and Dietary Approaches controlled, and those with suggested beneficial
As noted, metabolic approaches have an enor- effects should advance into well-controlled stud-
mous potential due to the prevalence of metabolic ies given clear positive effects of placebo (e.g.,
dysfunction in ASD and the potential to treat Kern et al., 2001). Individualized dietary treat-
core symptoms via re-establishing metabolic ment remains a promising new area that could
function. Such an approach could have a wide but yield new therapies with few side effects.
concerted influence in ASD and improve multi- One dietary approach that has been tried
ple aspects of overall health. with success in ASD patients and in animal
models is the ketogenic diet (KD; Arvio et al.,
Diet 2010; Evangeliou etal., 2003; Ruskin etal., 2013;
Diets remain popular ASD treatments:one study Spilioti etal., 2013). This high-fat, very low car-
found 17% of children and adolescents with ASD bohydrate diet is a remarkably effective nonphar-
had taken some type of specially formulated vita- macological treatment for patients with epilepsy,
min or supplement, and 16% were on a modi- even in cases poorly responsive to anticonvul-
fied diet (Witwer and Lecavalier, 2005). Dietary sant drugs (Kinsman et al., 1992; Neal et al.,
approaches for this disorder have remained 2009; Suo et al., 2013). Furthermore, there are
popular for many reasonssome parents favor many decades of clinical experience with KDs.
approaches other than medication, and there is a Strikingly, some refractory patients (consist-
high comorbidity of gastrointestinal symptoms, ently at least 10%15% across studies) become
some of which may be alleviated or reduced with seizure-free, and at least 50% achieve over 50%
diet. In cases with known metabolic abnormali- seizure reduction (Hallbk etal., 2015; Hassan
ties, multiple reports suggest removing or adding et al., 1999; Kang et al., 2007; Neal et al., 2009;
specific dietary constituents can prevent or allevi- Suo etal., 2013). Asubset of these can be weaned
ate autistic symptoms (Anonymous, 1968; Hooft off the KD and remain seizure-free (Martinez
etal., 1968; James etal., 2004; Page and Moseley, et al., 2007; Patel et al., 2010), suggesting that
2002; Page etal., 1997; Spilioti etal.,2013). KDs engage long-lasting, possibly epigenetic,
Many diets and dietary supplements have anticonvulsant mechanisms (Boison etal., 2013;
been tried to treat ASD. In randomized, con- Kobow etal., 2013). KDs limit glucose availability
trolled trials, the gluten-free, casein-free diet, and so recruit ketone bodies (made in the liver
perhaps the most well-known ASD treatment from -oxidation of fat) to be used as a substi-
to the public, has been split between find- tute cellular fuel, a major switch in body/brain
ings of effectiveness and ineffectiveness; fail- metabolism. Clearly, KDs would be useful in the
ure in a placebo-controlled trial certainly treatment of epilepsy when comorbid with ASD.
diminished enthusiasm in the field (Elder But based on a hypothesized link between autism
etal., 2006; Knivsberg etal., 2002; Seung etal., and adenosine (see previous discussion) and evi-
2007; Whiteley et al., 2010). Other diets have dence that KD feeding augments brain adenosine
not been in randomized, controlled trials. For levels (Kawamura et al., 2014; Kawamura et al.,
dietary manipulations, studies to better link 2010; Masino and Geiger, 2008; Masino et al.,
Autism Spectrum Disorders and Homeostasis 595

2011b), KDs should improve autistic symptoms. three-chamber test in which test mice have the
Indeed, this is what is found in those few clini- option of spending time in a chamber alone or
cal studies that addressed this issue (Arvio etal., in a chamber with a novel mouse (and in which
2010; Evangeliou etal., 2003; Spilioti etal., 2013). normal strains of mice prefer to spend time
In the largest of these, 18 of 30 ASD patients that with a novel mouse), BTBR mice fed a control diet
maintained KD treatment had improvements in were not social, whereas BTBR mice maintained
autistic symptoms ranging from minor to sub- on a KD were social and preferentially spent time
stantial (Evangeliou et al., 2003). However, this in the chamber with a novel mouse (Figure 31.2;
study used a nonstandard KD protocol and has Ruskin etal., 2013). When sociability was meas-
not yet been replicated. Notably, given the asso- ured another way (i.e., in time spent in direct
ciation of mitochondrial defects in ASD, it is par- social contact with the novel mouse), KD-fed
ticularly relevant that KDs are known to promote BTBR mice were significantly more social than
mitochondrial biogenesis and aid energy bal- their control-fed counterparts (Figure 31.3). KD
ance (ATP production) (Bough etal., 2006; Noh feeding also improved social transmission of a
etal., 2004; Srivastava etal., 2013; Sullivan etal., food preference and significantly reduced repeti-
2004)and are used to treat patients with electron tive behavior (Ruskin etal., 2013). These findings
transfer chain defects (Kang etal.,2007). whereby the KD improved symptoms of ASD
We tested the KD/ASD interaction in a mouse have recently been confirmed in another rodent
model of ASD, the BTBR mouse, an inbred autism model, gestational valproic acid exposure
strain in which all mice show low sociability (Ahn etal.,2014).
in the home cage and in testing environments KDs are challenging for some patients
(Babineau et al., 2013; Moy et al., 2007). In the because they are necessarily quite restrictive

500 baseline sociability social novelty


(phase 1) (phase 2) (phase 3)

control diet ***


400
ketogenic diet
**
Time in chamber (s)

300

200

100

0
for stranger mouse
center
to be empty

for stranger mouse


center
to be empty

stranger mouse
center
empty

stranger mouse
center
empty

novel mouse
center
familiar mouse

novel mouse
center
familiar mouse

FIGURE 31.2: Sociability and preference for social novelty in BTBR mice and dietary effects. Data are from
three-chamber testing. In baseline (in the absence of other mice), test mice did not prefer one side over the other,
as expected. With a stranger mouse in one chamber (phase 2), BTBR mice on control diet did not significantly pre-
fer that side, but BTBR mice fed the ketogenic diet (KD) did prefer to spend time in the stranger mouse chamber.
With a novel stranger mouse placed in the opposite chamber (phase 3), BTBR mice fed the KD (but not those fed
the control diet) significantly preferred to spend time with the novel mouse over the familiar mouse. Outlier data
were identified with Grubbs test and discarded. Statistics are paired t tests of side times, corrected for multiple
comparisons (**p < 0.01, ***p < 0.001). No other paired side-by-side comparisons were significant.
Data adapted from Ruskin etal. (2013).
596 Part IV:Homeostatic Therapies

regarding food types. Some investigators easily absorbed and metabolized to ketone
have supplemented KDs with medium-chain bodies, but at the end of -oxidative metabo-
triglycerides (MCTs) so that carbohydrate lism a three-carbon chain compound is left
restriction need not be so strict. More so than over, which can be converted to succinyl-CoA.
the long-chain triglycerides in typical KDs, Succinyl-CoA is a tricarboxylic acid cycle
MCTs are easily absorbed and metabolized intermediatethus dietary supplementation
to ketone bodies. MCT-containing KDs have with an odd-chain MCT could boost the tricar-
been used clinically with success in epilepsy boxylic acid cycle (termed anaplerosis) and so
(Ebus et al., 2014; Huttenlocher et al., 1971; aid in faulty energy metabolism. Patients with
Neal et al., 2009) and were used in the larg- epilepsy due to glucose transporter deficiency
est clinical ASD/KD study to date (Evangeliou (i.e., GLUT1 deficiency, a condition treated
et al., 2003). Alternatively, MCTs, which are successfully with the classical ketogenic
flavorless, colorless oils, can simply be added diet: Klepper and Leiendecker, 2013) showed
to a normal diet, and MCTs used in this man- improvement with seizures and neuropsycho-
ner have recently been shown to be anticon- logical performance when triheptanoin sup-
vulsant in rodent models (Socaa et al., 2015; plemented their regular diet (Pascual et al.,
Wla et al., 2015). A specific subtype of MCT 2014). Notably, in a mouse model of Rett syn-
is one in which the fatty acids have backbones drome, an ASD-related genetic disorder that
composed of an odd number of carbons, as until recently was subsumed under autism in
opposed to the even-chain carbon fatty acids the DSM, a triheptanoin-supplemented normal
typically found in nature. These fats, par- diet improved a large number of metabolic and
ticularly triheptanoin (the triglyceride of the physiological dysfunctions, delayed death, and
odd-chain fatty acid heptanoic acid), are also enhanced behaviors including social behaviors
(Park et al., 2014). It is unclear if odd-chain
MCTs truly have different effects than the
300 control diet normal even-chain. However, MCTs, odd- or
ketogenic diet even-chain, would be an easily studied (and
***
250
harmless) food supplement to study clinically
*** in ASD patients. KDs, MCT/KDs, and MCTs
appear to be dietary treatments with signifi-
200
cant effects on metabolism and neural func-
Contact time (s)

tion and indeed may represent approaches that


150 normalize several aspects of neural and meta-
bolic function. Clearly, they deserve careful
100 clinicalstudy.
For existing diet-based therapies, parents
50 report marked improvements with various
dietary changes; some may be due to a placebo
0 effect or simply because the diet may be health-
phase 2 phase 3 ier overall. Given the number of variables in a
diet, combined with the etiological heteroge-
FIGURE 31.3: Dietary effects on sociability in BTBR neity in ASD, there is of course the possibility
mice measured with social contact in three-chamber that particular diets or supplements might ben-
testing. Social contact is defined here as physical con- efit individuals with particular etiologies (e.g.,
tact of the test mouse mouth, nose, or forepaws with the Pietropaolo etal., 2014). As noted, this area could
stranger mouse or the small cage keeping the stranger benefit from significantly more research to define
mouse in place. Feeding a ketogenic diet significantly and discriminate beneficial dietary approaches
increased time in social contact with the stranger within subgroups of the population with ASD.
mouse (phase 2) or the total social contact with both Therefore, the search for safe and effective die-
stranger mice (phase 3). Outlier data were identified tary treatments of ASD is ongoing, could impact
with Grubbs test and discarded. Statistics are unpaired multiple aspects of homeostasis, and also has the
t tests between diets, corrected for multiple compari- potential to improve overall health. More detailed
sons (***p < 0.001). information on dietary manipulations with spe-
Data adapted from Ruskin etal. (2013) (phase 3)and unpub- cific nutrients can be found in Chapter16 of this
lished data (phase2). volume.
Autism Spectrum Disorders and Homeostasis 597

Exercise a disordered microbiome, oral treatment with


Exercise has perhaps been inadequately stud- Bacteroides fragilis (a prominent component of
ied and used as a targeted therapy in ASD. a healthy microbiome) normalized the microbi-
Physical activity is, like diet, a holistic treat- ome and some bacterial metabolites and normal-
ment with simultaneous effects on multiple ized elevated stereotyped behavior and anxiety
systems. It avoids the side effects of medica- but did not affect impaired sociability (all tested
tions and the complications of diets, is a boon several weeks after treatment; Hsiao etal., 2013).
to overall health, and, depending on how and Interestingly, when vitamin/mineral supple-
when it is presented, may be viewed by pediatric ments improved autistic symptoms in children,
patientsand even adultsas fun. In children those children who were most likely to benefit
with ASD, exercise can reduce stereotypy and had low starting levels of vitamin K and biotin,
self-stimulatory behaviors (Anderson-Hanley vitamins largely produced by healthy gut flora
et al., 2011; Neely et al., 2015; Watters and (Adams etal., 2011a). Thus, in some cases, dietary
Watters, 1980), improve cognitive and executive supplementation could be compensating for an
function (Anderson-Hanley et al., 2011), and abnormal microbiome. Further animal experi-
help with strength, agility, and running speed ments in the vein of Hsiao et al. (2013) could
(Hilton etal., 2014; Oriel etal., 2011). All of these determine the extent to which the microbiome
studies were performed after acute exercise. influences behavior and cognition. It remains
Another study found that chronic (14 weeks) to be seen if full fecal transplantation from a
exercise significantly reduced stereotypic behav- microbiomically normal donor would lead to a
ior, and this reduction remained for one month more complete normalization of the ASD hosts
after exercise cessation (Bahrami et al., 2012). behavior.
The lasting effects of chronic exercise could be
due to multiple factors, including epigenetic Behavioral Approaches
effects, but the precise pathways by which exer- As noted, a range of clear physiological changes
cise has these acute and chronic effects is unclear. can be found in patients with ASD, and early
In accord with the adenosine/ASD hypothesis intervention with cognitive and behavioral
(Masino and Geiger, 2008; Masino et al., 2009; therapies is the most effective treatment to date.
Masino et al., 2011a), acute exercise elevates There is also evidence that some children can
adenosine and its major metabolite inosine in escape the diagnosis of autism and improve
brain (Dworak et al., 2007); chronic exercise their behavior over time such that they no longer
downregulates adenosine receptor expression in meet the diagnostic criteria (Fein et al., 2014).
the brain, consistent with a homeostatic receptor Various cognitive and behavioral treatment
response to chronically high activation by aden- options are available, and translation of clinic
osine (Clark etal., 2014; Costa etal., 2012); and and university-based programs into the commu-
chronic exercise reduces the painful response nity has produced promising results, with behav-
to tactile sensitivity in neuropathic pain in an ior skills training focusing on childcaregiver
adenosine receptor-dependent manner, sug- joint attention, reciprocal imitation training, and
gesting enhanced adenosinergic transmission symbolic play, and social skills training on prag-
(Martins etal.,2013). matic communication, language use, and social
perception and interaction (Adams et al., 2012;
Microbiome Corbett etal., 2014; Goods etal., 2013; Ingersoll
Because an aberrant microbiome is related to et al., 2012; Landa et al., 2011; Warreyn et al.,
cognitive and behavioral problems (see previous 2014). While these programs and treatments
discussion)and possibly causative through as have experienced some success in ameliorating
yet undetermined pathwaysit is possible that the core symptoms of autism in children, further
normalizing the gut microbial community could research is needed into the empirical techniques
then normalize these problems. Normal intes- that cause these improvements, early ASD detec-
tinal flora can be reconstituted, for instance by tion and intervention, adolescent and adult ther-
fecal transplantation, which has seen extraor- apies, and cultural/social barriers influencing
dinary success in treating Clostridium difficile program implementation.
infection (Landy et al., 2011) and is in rand- Behavioral/cognitive therapy is one
omized controlled trials for other bowel prob- approach that directly targets the core symp-
lems and metabolic syndrome (Singh etal., 2014). toms of autism, and perhaps because it is
In the MIA mouse model of ASD, which includes a holistic approachengaging all systems
598 Part IV:Homeostatic Therapies

simultaneouslyit is an ideal example of a home- date. Combining MIA with a polygenic model
ostatic therapy. Unfortunately it is also a domain of increased risk for ASD could be a particularly
in which it is difficult to access why it works at a exciting and relevant animalmodel.
mechanistic level to then develop a better dose. Awareness of the prenatal period and the
However, because no other therapy can replace specific impact of MIA-induced increased risk
behavioral experience with social situations and for ASD is coming to the forefront in parallel
reciprocal communication, in the absence of a with information about the metabolic and physi-
true magic pill this modality would seem to be ological prenatal environment. For example,
an enduring and perhaps permanent therapeutic mothers with gestational diabetes, high blood
option for ASDand perhaps an underexplored pressure, obesity, and other conditions are more
and/or underutilized therapy for other neuro- likely to have a child with ASD or other neu-
logical disorders. rodevelopmental disorders (Polo-Kantola et al.,
2014; Sullivan et al., 2014; Xu et al., 2014). To
Early Intervention and Prevention our knowledge there are no validated animal
Early behavioral and anatomical markers and models in which metabolic dysregulation during
manifestations of ASD have recently been gestationor even in generalincreases risks for
explored with the goal of providing early inter- ASD; however, animal models available currently
vention and diagnosis (Barton et al., 2012). may exhibit behavioral characteristics of ASD
Similar to the findings of Courchesne etal. (2011) and simply need to be characterized as such to
in deceased adult ASD brains, studies in young add a new cadre of research tools forASD.
children have found increased whole-brain vol-
ume especially in the white and grey matter of CONCLUSIONS AND FUTURE
the cerebral cortex, but there remains a great deal DIRECTIONS
of heterogeneity and research needed to identify In sum, ASD spans a wide range of symptoms
more specific brain regions implicated in ASD and language and cognitive abilities throughout
(Amaral etal., 2008; Barton etal., 2012). Parents the lifespan. While diagnosis is based on deficits
of children later diagnosed with ASD have also in social communication and interactions, as
reported early symptoms including decreased well as restricted, repetitive behaviors and inter-
responsiveness to parent voices, blunted recipro- ests, ASD is often accompanied by comorbidities
cal caregiver attention, increased irritability and such as seizures, anxiety, depression, and gastro-
regulatory difficulties, and abnormal sensory intestinal issues. ASD has a complex and in the
reactivity (Barton etal., 2012). Even after an ASD vast majority of cases poorly understood under-
diagnosis, intense and early behavioral interven- lying etiology. To date, rational treatment strate-
tion, rather than medication, can lead to a com- gies for the core symptoms are lacking, and with
plete loss of ASD symptomology in some patients so little known about the cause(s) of most cases
(Fein et al., 2014), although subtle differences of autism, designing effective therapies remains
remain compared to controls (Troyb etal., 2014; extremely challenging. Clinical and experimen-
Tyson etal.,2014). tal evidence suggests that abnormal neuronal
Certainly preventing maternal infection excitability, metabolism, and/or immune func-
during pregnancy as much as possible is a prac- tion are common features of ASD, and there-
tical and effective strategy to reduce the risk fore a focus on restoring homeostasis may offer
for ASD. Paul Patterson (2011a) strongly pro- the most broad-based solution for reducing core
moted this approach and a better public aware- behavioral symptoms and possibly comorbidi-
ness of the risks for multiple conditions in his ties. An increased attention to restoring or main-
book Infectious Behavior. As a simple strategy taining homeostasis should perhaps extend to the
beyond prevention, could targeted treatment gestational period, as adverse immune or meta-
with anti-inflammatory agents help reduce ASD bolic conditions during fetal development may
riskparticularly in a population with increased increase the risk of an autism diagnosis. Overall,
genetic predisposition? This is a strategy that strategies that promote neural, metabolic, and
could easily be tested in validated animal models. immune homeostasis, combined with proven
For example, the MIA experimental model seems behavioral interventions, may offer the best avail-
quite robusteffective in primates and rodents, able treatment and long-term outcomes forASD.
with timing similar to the human conditionand At this time, translational ASD research
is perhaps one of our better models of ASD to should embrace therapeutic opportunities to
Autism Spectrum Disorders and Homeostasis 599

improve or support bioenergetics and network the prenatal valproic acid model of autism spec-
homeostasis in the nervous system and support trum disorder. Dev. Neurosci. 36, 371380.
health and homeostasis of overall physiology. Amaral, D.G., Schumann, C.M., and Nordahl, C.W.
This could be via explicit systemic approaches (2008). Neuroanatomy of autism. Trends
(such as exercise or diet), increasing endog- Neurosci. 31, 137145.
enous homeostatic molecules (such as adeno- American Psychiatric Association (2013). Diagnostic
sine, BDNF), anti-inflammatory strategies and Statistical Manual of Mental Disorders V,
(exercise, dietary changes, supplements, or 4th ed. (Washington DC:American Psychiatric
pharmaceuticals), maternal health interven- Association).
tion and disease prevention, and other targeted Amiet, C., Gourfinkel-An, I., Laurent, C., Bodeau, N.,
general improvements in neural, metabolic, Genin, B., Leguern, E., Tordjman, S., and Cohen, D.
or immunological function. Individual symp- (2013). Does epilepsy in multiplex autism pedigrees
toms and practicality of a treatment approach define a different subgroup in terms of clinical
can be taken into consideration in the clinic. characteristics and genetic risk? Mol. Autism4,47.
Anderson-Hanley, C., Tureck, K., and Schneiderman,
Alongside early intervention, a synergis-
R.L. (2011). Autism and exergaming: Effects on
tic approach that combines supporting and
repetitive behaviors and cognition. Psychol. Res.
restoring normal physiology of these complex
Behav. Manag. 4, 129137.
systems along with behavioral/cognitive ther-
Anonymous. (1968). Dietary treatment of phenylke-
apy may offer the best chance for lasting and
tonuria. Br. Med. J. 4,135.
significant improvement.
Argyropoulos, A., Gilby, K.L., and Hill-Yardin, E.L.
(2012). Studying autism in rodent models:
References Reconciling endophenotypes with comorbidi-
Adams, C., Lockton, E., Freed, J., Gaile, J., Earl, G., ties. Front. Hum. Neurosci. 7,417.
McBean, K., Nash, M., Green, J., Vail, A., and Arvio, M., Kuisma, L., and Pontinen, M. (2010). [The
Law, J. (2012). The Social Communication modified Atkins diet brought back the joy of life
Intervention Project: A randomized controlled to a developmentally severely disabled youth].
trial of the effectiveness of speech and language Duodecim. 126, 557560.
therapy for school-age children who have prag- Ashwood, P., Krakowiak, P., Hertz-Picciotto, I.,
matic and social communication problems Hansen, R., Pessah, I., and Van de Water, J.
with or without autism spectrum disorder. Int. (2011a). Elevated plasma cytokines in autism
J.Lang. Commun. Disord. 47, 233244. spectrum disorders provide evidence of immune
Adams, J.B., Audhya, T., McDonough-Means, S., dysfunction and are associated with impaired
Rubin, R.A., Quig, D., Geis, E., Gehn, E., Loresto, behavioral outcome. Brain Behav. Immun.
M., Mitchell, J., Atwood, S., etal. (2011a). Effect 25,4045.
of a vitamin/mineral supplement on children Ashwood, P., Krakowiak, P., Hertz-Picciotto, I.,
and adults with autism. BMC Pediatr. 11,111. Hansen, R., Pessah, I.N., and Van de Water, J.
Adams, J.B., Audhya, T., McDonough-Means, S., (2011b). Associations of impaired behaviors with
Rubin, R.A., Quig, D., Geis, E., Gehn, E., Loresto, elevated plasma chemokines in autism spectrum
M., Mitchell, J., Atwood, S., et al. (2011b). disorders. J. Neuroimmunol. 232, 196199.
Nutritional and metabolic status of children with Atladttir, H.., Henriksen, T.B., Schendel, D.E.,
autism vs. neurotypical children, and the asso- and Parner, E.T. (2012). Autism after infec-
ciation with autism severity. Nutr. Metab.8,34. tion, febrile episodes, and antibiotic use during
Adams, J.B., Audhya, T., McDonough-Means, S., pregnancy: An exploratory study. Pediatrics
Rubin, R.A., Quig, D., Geis, E., Gehn, E., 130,e1447.
Loresto, M., Mitchell, J., Atwood, S., etal. (2013). Atladttir, H.., Pedersen, M.G., Thorsen, P.,
Toxicological status of children with autism vs. Mortensen, P.B., Deleuran, B., Eaton, W.W., and
neurotypical children and the association with Parner, E.T. (2009). Association of family history
autism severity. Biol. Trace Elem. Res. 151, 171180. of autoimmune diseases and autism spectrum
Adams, J.B., Romdalvik, J., Ramanujam, V.M., and disorders. Pediatrics 124, 687694.
Legator, M.S. (2007). Mercury, lead, and zinc in Atladttir, H.., Thorsen, P., stergaard, L.,
baby teeth of children with autism versus con- Schendel, D.E., Lemcke, S., Abdallah, M., and
trols. J. Toxicol. Environ. Health 70, 10461051. Parner, E.T. (2010). Maternal infection requir-
Ahn, Y., Narous, M., Tobias, R., Rho, J.M., and ing hospitalization during pregnancy and autism
Mychasiuk, R. (2014). The ketogenic diet modi- spectrum disorders. J. Autism Dev. Disord. 40,
fies social and metabolic alterations identified in 14231430.
600 Part IV:Homeostatic Therapies

Babineau, B.A., Yang, M., Berman, R.F., and mechanism of the ketogenic diet. Ann. Neurol.
Crawley, J.N. (2013). Low home cage social 60, 223235.
behaviors in BTBR T+tf/J mice during juvenile Breitenkamp, A.F., Matthes, J., Nass, R.D., Sinzig, J.,
development. Physiol. Behav. 114115,4954. Lehmkuhl, G., Nurnberg, P., and Herzig, S.
Bahrami, F., Movahedi, A., Marandi, S.M., and (2014). Rare mutations of CACNB2 found in
Abedi, A. (2012). Kata techniques training con- autism spectrum disease-affected families
sistently decreases stereotypy in children with alter calcium channel function. PLoS One 9,
autism spectrum disorder. Res. Dev. Disabil. 33, e95579.
11831193. Brugha, T.S., McManus, S., Bankart, J., Scott, F.,
Baribeau, D.A., and Anagnostou, E. (2014). An Purdon, S., Smith, J., Bebbington, P., Jenkins, R.,
update on medication management of behav- and Meltzer, H. (2011). Epidemiology of autism
ioral disorders in autism. Curr. Psychiatry Rep. spectrum disorders in adults in the community
16,437. in England. Arch. Gen. Psychiatry 68, 459465.
Barrett, B., Mosweu, I., Jones, C.R., Charman, T., Buescher, A.V., Cidav, Z., Knapp, M., and Mandell,
Baird, G., Simonoff, E., Pickles, A., Happe, F., D.S. (2014). Costs of autism spectrum disorders
and Byford, S. (in press). Comparing service use in the United Kingdom and the United States.
and costs among adolescents with autism spec- JAMA Pediatr. 168, 721728.
trum disorders, special needs and typical devel- Careaga, M., Van de Water, J., and Ashwood, P.
opment. Autism. (2010). Immune dysfunction in autism:Apath-
Barton, M.L., Orinstein, A., Troyb, E., and Fein, D. way to treatment. Neurotherapeutics 7,
(2012). Early manifestations of autism spec- 283292.
trum disorders. In:The Neuroscience of Autism Carlsson, L.H., Norrelgen, F., Kjellmer, L.,
Spectrum Disorders, J.D. Buxbaum, and P.R. Westerlund, J., Gillberg, C., and Fernell, E.
Hof, eds. (Chicago:Elsevier), pp.3969. (2013). Coexisting disorders and problems in
Bauman, M.D., Iosif, A.-M., Smith, S.E.P., Bregere, C., preschool children with autism spectrum disor-
Amaral, D.G., and Patterson, P.H. (2014). ders. ScientificWorldJournal 2013, 213979.
Activation of the maternal immune system dur- Centers for Disease Control and Prevention (2014).
ing pregnancy alters behavioral development of Prevalence of autism spectrum disorder among
Rhesus monkey offspring. Biol. Psychiatry 75, children aged 8yearsautism and developmen-
332341. tal disabilities monitoring network, 11 sites,
Berg, A.T. (2008). Risk of recurrence after a first United States, 2010. MMWR Surveill. Summ.
unprovoked seizure. Epilepsia 49 Suppl. 1,1318. 63(2),121.
Berko, E.R., Suzuki, M., Beren, F., Lemetre, C., Charman, T., Pickles, A., Simonoff, E., Chandler,
Alaimo, C.M., Calder, R.B., Ballaban-Gil, K., S., Loucas, T., and Baird, G. (2011). IQ in chil-
Gounder, B., Kampf, K., Kirschen, J., etal. (2014). dren with autism spectrum disorders:Data from
Mosaic epigenetic dysregulation of ectodermal the Special Needs and Autism Project (SNAP).
cells in autism spectrum disorder. PLoS Genet. Psychol. Med. 41, 619627.
10, e1004402. Chez, M.G., Dowling, T., Patel, P.B., Khanna, P., and
Betancur, C. (2011). Etiological heterogeneity in Kominsky, M. (2007). Elevation of tumor necro-
autism spectrum disorders: More than 100 sis factor-alpha in cerebrospinal fluid of autistic
genetic and genomic disorders and still count- children. Pediatr. Neurol. 36, 361365.
ing. Brain Res. 1380,4277. Ching, H., and Pringsheim, T. (2012). Aripiprazole
Betancur, C., and Coleman, M. (2012). Etiological for autism spectrum disorders (ASD). Cochrane
heterogeneity in autism spectrum disorders:role Database Syst. Rev. 5, CD009043.
of rare variants. In:The Neuroscience of Autism Clark, P.J., Ghasem, P.R., Mika, A., Day, H.E.,
Spectrum Disorders, J.D. Buxbaum, and P.R. Herrera, J.J., Greenwood, B.N., and Fleshner, M.
Hof, eds. (Chicago:Elsevier), pp. 113144. (2014). Wheel running alters patterns of uncon-
Boison, D., Sandau, U., Ruskin, D.N., Kawamura, M., Jr., trollable stress-induced cfos mRNA expression
and Masino, S.A. (2013). Homeostatic control of in rat dorsal striatum direct and indirect path-
brain functionnew approaches to understand ways:Apossible role for plasticity in adenosine
epileptogenesis. Front. Cell. Neurosci. 7,109. receptors. Behav. Brain Res. 272, 252263.
Bough, K.J., Wetherington, J., Hassel, B., Pare, J.F., Corbett, B.A., Swain, D.M., Coke, C., Simon, D.,
Gawryluk, J.W., Greene, J.G., Shaw, R., Smith, Y., Newsom, C., Houchins-Juarez, N., Jenson, A.,
Geiger, J.D., and Dingledine, R.J. (2006). Wang, L., and Song, Y. (2014). Improvement
Mitochondrial biogenesis in the anticonvulsant in social deficits in autism spectrum disorders
Autism Spectrum Disorders and Homeostasis 601

using a theatre-based, peer-mediated interven- Dunwiddie, T.V., and Masino, S.A. (2001). The role
tion. Autism Res. 7,416. and regulation of adenosine in the central ner-
Cortese, S., Castelnau, P., Morcillo, C., Roux, S., and vous system. Annu. Rev. Neurosci. 24,3155.
Bonnet-Brilhault, F. (2012). Psychostimulants Dworak, M., Diel, P., Voss, S., Hollmann, W.,
for ADHD-like symptoms in individuals and Struder, H.K. (2007). Intense exercise
with autism spectrum disorders. Expert Rev. increases adenosine concentrations in rat
Neurother. 12, 461473. brain:Implications for a homeostatic sleep drive.
Costa, M.S., Ardais, A.P., Fioreze, G.T., Mioranzza, S., Neuroscience 150, 789795.
Botton, P.H., Portela, L.V., Souza, D.O., and Ebus, S.C.M., Lambrechts, D.A.J.E., Herraets,
Porciuncula, L.O. (2012). Treadmill running fre- I.J.T., Majoie, M.J.M., de Louw, A.J., Boon, P.J.,
quency on anxiety and hippocampal adenosine Aldenkamp, A.P., and Arends, J.B. (2014). Can
receptors density in adult and middle-aged rats. an early 24-hour EEG predict the response to the
Prog. Neuro-Psychopharmacol. Biol. Psychiatry ketogenic diet? A prospective study in 34 chil-
36, 198204. dren and adults with refractory epilepsy treated
Courchesne, E., Mouton, P.R., Calhoun, M.E., with the ketogenic diet. Seizure 23, 468474.
Semendeferi, K., Ahrens-Barbeau, C., Hallet, M.J., El-Ansary, A., Al-Daihan, S., Al-Dbass, A., and
Barnes, C.C., and Pierce, K. (2011). Neuron num- Al-Ayadhi, L. (2010). Measurement of selected
ber and size in prefrontal cortex of children with ions related to oxidative stress and energy metab-
autism. JAMA 306, 20012010. olism in Saudi autistic children. Clin. Biochem.
Cuccaro, M.L., Tuchman, R.F., Hamilton, K.L., 43,6370.
Wright, H.H., Abramson, R.K., Haines, J.L., Elbe, D., and Lalani, Z. (2012). Review of the phar-
Gilbert, J.R., and Pericak-Vance, M. (2012). macotherapy of irritability of autism. J. Can.
Exploring the relationship between autism Acad. Child Adolesc. Psychiatry 21, 130146.
spectrum disorder and epilepsy using latent Elder, J.H., Shankar, M., Shuster, J., Theriaque, D.,
class cluster analysis. J. Autism Dev. Disord. 42, Burns, S., and Sherrill, L. (2006). The gluten-free,
16301641. casein-free diet in autism: Results of a prelimi-
De Angelis, M., Piccolo, M., Vannini, L., Siragusa, S., nary double blind clinical trial. J. Autism Dev.
De Giacomo, A., Serrazzanetti, I., Cristofori, F., Disord. 36, 413420.
Guerzoni, M.E., Gobbetti, M., and Francavilla, R. Enstrom, A.M., Onore, C.E., Van de Water, J.A.,
(2013). Fecal microbiota and metabolome of chil- and Ashwood, P. (2010). Differential monocyte
dren with autism and pervasive developmental responses to TLR ligands in children with autism
disorder not otherwise specified. PLOS One 8, spectrum disorders. Brain Behav. Immun.
e76993. 24,6471.
Depino, A.M. (2013). Peripheral and central inflam- Evangeliou, A., Vlachonikolis, I., Mihailidou,
mation in autism spectrum disorders. Mol. Cell. H., Spilioti, M., Skarpalezou, A., Makaronas,
Neurosci. 53,6976. N., Prokopiou, A., Christodoulou, P.,
Dhillon, S., Hellings, J.A., and Butler, M.G. (2011). Liapi-Adamidou, G., Helidonis, E., etal. (2003).
Genetics and mitochondrial abnormalities in Application of a ketogenic diet in children with
autism spectrum disorders: A review. Curr. autistic behavior:Pilot study. J. Child Neurol. 18,
Genomics 12, 322332. 113118.
Doshi-Velez, F., Ge, Y., and Kohane, I. (2014). Farmer, C., Thurm, A., and Grant, P. (2013).
Comorbidity clusters in autism spectrum dis- Pharmacotherapy for the core symptoms in
orders: An electronic health record time-series autistic disorder:Current status of the research.
analysis. Pediatrics 133, e54e63. Drugs 73, 303314.
Dove, D., Warren, Z., McPheeters, M.L., Taylor, Fatemi, S.H., Folsom, T.D., Reutiman, T.J., and
J.L., Sathe, N.A., and Veenstra-VanderWeele, J. Thuras, P.D. (2009a). Expression of GABAB
(2012). Medications for adolescents and young receptors is altered in brains of subjects with
adults with autism spectrum disorders: A sys- autism. Cerebellum 8,6469.
tematic review. Pediatrics 130, 717726. Fatemi, S.H., Reutiman, T.J., Folsom, T.D., and
Doyle, C.A., and McDougle, C.J. (2012). Thuras, P.D. (2009b). GABAA receptor down-
Pharmacologic treatments for the behavioral regulation in brains of subjects with autism. J.
symptoms associated with autism spectrum Autism Dev. Disord. 39, 223230.
disorders across the lifespan. Dialogues Clin. Fedele, D.E., Li, T., Lan, J.Q., Fredholm, B.B., and
Neurosci. 14, 263279. Boison, D. (2006). Adenosine A1 receptors are
Dunwiddie, T.V. (1999). Adenosine and suppression crucial in keeping an epileptic focus localized.
of seizures. Adv. Neurol. 79, 10011010. Exp. Neurol. 200, 184190.
602 Part IV:Homeostatic Therapies

Fein, D., Barton, M., Eigsti, I.M., Kelley, E., Naigles, L., Hallbk, T., Sjolander, A., Amark, P., Miranda, M.,
Schultz, R.T., Stevens, M., Helt, M., Orinstein, A., Bjurulf, B., and Dahlin, M. (2015). Effectiveness
Rosenthal, M., et al. (2014). Optimal outcome of the ketogenic diet used to treat resistant child-
in individuals with a history of autism. J. Child hood epilepsy in Scandinavia. Eur. J. Paediatr.
Psychol. Psychiatry 54, 195205. Neurol. 19,2936.
Fredholm, B.B. (2007). Adenosine, an endogenous Hassan, A.M., Keene, D.L., Whiting, S.E., Jacob, P.J.,
distress signal, modulates tissue damage and Champagne, J.R., and Humphreys, P. (1999).
repair. Cell Death Differ. 14, 13151323. Ketogenic diet in the treatment of refractory epi-
Fredholm, B.B. (2011). Physiological and pathophysi- lepsy in childhood. Pediatr. Neurol. 21, 548552.
ological roles of adenosine. Sleep Biol. Rhythms Hilton, C.L., Cumpata, K., Klohr, C., Gaetke, S.,
9 (Suppl.1),2428. Artner, A., Johnson, H., and Dobbs, S. (2014).
Froehlich-Santino, W., Londono Tobon, A., Effects of exergaming on executive function
Cleveland, S., Torres, A., Phillips, J., Cohen, B., and motor skills in children with autism spec-
Torigoe, T., Miller, J., Fedele, A., Collins, J., trum disorder:a pilot study. Am. J.Occup. Ther.
et al. (2014). Prenatal and perinatal risk factors 68,5765.
in a twin study of autism spectrum disorders. J. Hirota, T., Veenstra-Vanderweele, J., Hollander, E.,
Psychiatr. Res. 54, 100108. and Kishi, T. (2013). Antiepileptic medications in
Frye, R.E., and Rossignol, D.A. (2011). Mitochondrial autism spectrum disorder: A systematic review
dysfunction can connect the diverse medical and meta-analysis. J. Autism Dev. Disord. 44,
symptoms associated with autism spectrum dis- 948957.
orders. Pediatr. Res. 69, 41R-47R. Hohenegger, M., Waldhoer, M., Beindl, W., Boing, B.,
Ganz, M.L. (2007). The lifetime distribution of Kreimeyer, A., Nickel, P., Nanoff, C., and
the incremental societal costs of autism. Arch. Freissmuth, M. (1998). Gs-selective G pro-
Pediatr. Adolesc. Med. 161, 343349. tein antagonists. Proc. Natl. Acad. Sci. USA 95,
Garcia-Escudero, V., Martin-Maestro, P., Perry, G., 346351.
and Avila, J. (2013). Deconstructing mitochon- Hooft, C., Van Nevel, C., and De Schaepdryver, A.F.
drial dysfunction in Alzheimer disease. Oxid. (1968). Hyperuricosuric encephalopathy without
Med. Cell. Longev. 2013, 162152. hyperuricaemia. Archiv. Dis. Child. 43, 734737.
Gardener, H., Spiegelman, D., and Buka, S.L. Horbinski, C., and Chu, C.T. (2005). Kinase signal-
(2011). Perinatal and neonatal risk factors ing cascades in the mitochondrion:Amatter of
for autism: A comprehensive meta-analysis. life or death. Free Radic. Biol. Med. 38,211.
Pediatrics 128, 344355. Horlin, C., Falkmer, M., Parsons, R., Albrecht, M.A.,
Giulivi, C., Napoli, E., Schwartzer, J., Careaga, M., and and Falkmer, T. (2014). The cost of autism spec-
Ashwood, P. (2013). Gestational exposure to a viral trum disorders. PLoS One 9, e106552.
mimetic poly(I:C) results in long-lasting changes Hsiao, E.Y., McBride, S.W., Chow, J., Mazmanian, S.K.,
in mitochondrial function by leucocytes in the and Patterson, P.H. (2012). Modeling an autism
adult offspring. Mediators Inflamm. 2013, 609602. risk factor in mice leads to permanent immune
Giulivi, C., Zhang, Y.F., Omanska-Klusek, A., dysregulation. Proc. Natl. Acad. Sci. USA 109,
Ross-Inta, C., Wong, S., Hertz-Picciotto, I., 1277612781.
Tassone, F., and Pessah, I.N. (2010). Hsiao, E.Y., McBride, S.W., Hsien, S., Sharon, G.,
Mitochondrial dysfunction in autism. JAMA Hyde, E.R., McCue, T., Codelli, J.A., Chow, J.,
304, 23892396. Reisman, S.E., Petrosino, J.F., et al. (2013).
Goods, K.S., Ishijima, E., Chang, Y.C., and Kasari, Microbiota modulate behavioral and physiologi-
C. (2013). Preschool based JASPER intervention cal abnormalities associated with neurodevelop-
in minimally verbal children with autism:Pilot mental disorders. Cell 155, 14511463.
RCT. J. Autism Dev. Disord. 43, 10501056. Huang, N.K., Lin, J.H., Lin, J.T., Lin, C.I., Liu, E.M.,
Greco, B., Manago, F., Tucci, V., Kao, H.T., Valtorta, Lin, C.J., Chen, W.P., Shen, Y.C., Chen, H.M.,
F., and Benfenati, F. (2013). Autism-related Chen, J.B., etal. (2011). A new drug design tar-
behavioral abnormalities in synapsin knockout geting the adenosinergic system for Huntingtons
mice. Behav. Brain Res. 251,6574. disease. PLoS One 6, e20934.
Haas, R.H. (2010). Autism and mitochondrial dis- Hultman, C.M., Sandin, S., Levine, S.Z., Lichtenstein,
ease. Dev. Disabil. Res. Rev. 16, 144153. P., and Reichenberg, A. (2011). Advancing pater-
Hagberg, H., Mallard, C., Rousset, C.I., and nal age and risk of autism: New evidence from
Thornton, C. (2014). Mitochondria: Hub of a population-based study and a meta-analysis
injury responses in the developing brain. Lancet of epidemiological studies. Mol. Psychiatry 16,
Neurol. 13, 217232. 12031212.
Autism Spectrum Disorders and Homeostasis 603

Huttenlocher, P.R., Wilbourn, A.J., and Signore, J.M. diet sensitizes glucose control of hippocampal
(1971). Medium-chain triglycerides as a therapy excitability. J. Lipid Res. 55, 22542260.
for intractable childhood epilepsy. Neurology 21, Kawamura, M., Jr., Ruskin, D.N., and Masino, S.A.
10971103. (2010). Metabolic autocrine regulation of neu-
Ibrahim, S.H., Voigt, R.G., Katusic, S.K., Weaver, A.L., rons involves cooperation among pannexin
and Barbaresi, W.J. (2009). Incidence of gas- hemichannels, adenosine receptors and KATP
trointestinal symptoms in children with channels. J. Neurosci. 30, 38863895.
autism: A population-based study. Pediatrics Kern, J.K., Miller, V.S., Cauller, L., Kendall, R.,
124, 680686. Mehta, J., and Dodd, M. (2001). Effectiveness
Ingersoll, B., Meyer, K., Bonter, N., and Jelinek, S. of N,N-dimethylglycine in autism and perva-
(2012). A comparison of developmental social- sive developmental disorder. J. Child Neurol. 16,
pragmatic and naturalistic behavioral interven- 169173.
tions on language use and social engagement in Kim, S.H., and Lord, C. (2012). The behavioral mani-
children with autism. J. Speech Lang. Hear. Res. festations of autism spectrum disorders. In:The
55, 13011313. Neuroscience of Autism Spectrum Disorders J.D.
James, S., Montgomery, P., and Williams, K. (2011). Buxbaum, and P.R. Hof, eds. (Chicago:Elsevier),
Omega-3 fatty acids supplementation for autism pp.2539.
spectrum disorders (ASD). Cochrane Database Kinsman, S.L., Vining, E.P., Quaskey, S.A.,
Syst. Rev., CD007992. Mellits, D., and Freeman, J.M. (1992). Efficacy
James, S.J., Cutler, P., Melnyk, S., Jernigan, S., Janak, L., of the ketogenic diet for intractable seizure
Gaylor, D.W., and Neubrander, J.A. (2004). disorders: Review of 58 cases. Epilepsia 33,
Metabolic biomarkers of increased oxida- 11321136.
tive stress and impaired methylation capacity Klepper, J., and Leiendecker, B. (2013). Glut1 defi-
in children with autism. Am. J. Clin. Nutr. 80, ciency syndrome and novel ketogenic diets. J.
16111617. Child Neurol. 28, 10451048.
James, S.J., Melnyk, S., Jernigan, S., Cleves, M.A., Knivsberg, A.M., Reichelt, K.L., Hoien, T., and
Halsted, C.H., Wong, D.H., Cutler, P., Bock, K., Nodland, M. (2002). A randomised, controlled
Boris, M., Bradstreet, J.J., etal. (2006). Metabolic study of dietary intervention in autistic syn-
endophenotype and related genotypes are asso- dromes. Nutr. Neurosci. 5, 251261.
ciated with oxidative stress in children with Kobow, K., Kaspi, A., Harikrishnan, K.N., Kiese, K.,
autism. Am. J. Med. Genet. B Neuropsychiatr. Ziemann, M., Khurana, I., Fritzsche, I., Hauke, J.,
Genet. 141, 947956. Hahnen, E., Coras, R., et al. (2013). Deep
Jirtle, R.L., and Skinner, M.K. (2007). Environmental sequencing reveals increased DNA methylation
epigenomics and disease susceptibility. Nat. Rev. in chronic rat epilepsy. Acta Neuropathol. 126,
Genet. 8, 253262. 741756.
Jyonouchi, H., Geng, L., Ruby, A., and Kohane, I.S., McMurry, A., Weber, G., MacFadden, D.,
Zimmerman-Bier, B. (2005). Dysregulated Rappaport, L., Kunkel, L., Bickel, J., Wattanasin, N.,
innate immune responses in young children Spence, S., Murphy, S., et al. (2012). The
with autism spectrum disorders:Their relation- co-morbidity burden of children and young
ship to gastrointestinal symptoms and dietary adults with autism spectrum disorders. PLoS
intervention. Neuropsychobiology 51,7785. One 7, e33224.
Kang, H.C., Lee, Y.M., Kim, H.D., Lee, J.S., and Korboukh, I., Hull-Ryde, E.A., Rittiner, J.E.,
Slama, A. (2007). Safe and effective use of the Randhawa, A.S., Coleman, J., Fitzpatrick, B.J.,
ketogenic diet in children with epilepsy and Setola, V., Janzen, W.P., Frye, S.V., Zylka, M.J.,
mitochondrial respiratory chain complex etal. (2012). Orally active adenosine A1 receptor
defects. Epilepsia 48,8288. agonists with antinociceptive effects in mice. J.
Kantzer, A.K., Fernell, E., Gillberg, C., and Med. Chem. 55, 64676477.
Miniscalco, C. (2013). Autism in community Kundakovic, M., Gudsnuk, K., Herbstman, J.B.,
pre-schoolers:Developmental profiles. Res. Dev. Tang, D., Perera, F.P., and Champagne, F.A.
Disabil. 34, 29002908. (in press). DNA methylation of BDNF as a bio-
Kasarpalkar, N.J., Kothari, S.T., and Dave, U.P. marker of early-life adversity. Proc. Natl. Acad.
(2014). Brain-derived neurotrophic factor in Sci.USA.
children with autism spectrum disorder. Ann. Landa, R.J., Holman, K.C., ONeill, A.H., and Stuart,
Neurosci. 21, 129133. E.A. (2011). Intervention targeting development
Kawamura, M., Jr., Ruskin, D.N., Geiger, J.D., of socially synchronous engagement in toddlers
Boison, D., and Masino, S.A. (2014). Ketogenic with autism spectrum disorder: A randomized
604 Part IV:Homeostatic Therapies

controlled trial. J. Child Psychol. Psychiatry A.R.S. (2013). High-intensity swimming exer-
52,1321. cise reduces neuropathic pain in an animal
Landy, J., Al-Hassi, H.O., McLaughlin, S.D., Walker, model of complex regional pain syndrome type
A.W., Ciclitira, P.J., Nicholls, R.J., Clark, S.K., and I:Evidence for a role of the adenosinergic system.
Hart, A.L. (2011). Faecal transplantation therapy Neuroscience 234,6976.
for gastrointestinal disease. Aliment. Pharmacol. Maruszak, A., and Zekanowski, C. (2011).
Ther. 34, 409415. Mitochondrial dysfunction and Alzheimers
Lavelle, T.A., Weinstein, M.C., Newhouse, J.P., disease. Prog. Neuropsychopharmacol. Biol.
Munir, K., Kuhlthau, K.A., and Prosser, L.A. Psychiatry 35, 320330.
(2014). Economic burden of childhood autism Marz, P., Heese, K., Dimitriades-Schmutz, B.,
spectrum disorders. Pediatrics 133, e520e529. Rose-John, S., and Otten, U. (1999). Role of
Lee, B.K., Magnusson, C., Gardner, R.M., interleukin-6 and soluble IL-6 receptor in
Blomstrom, S., Newschaffer, C.J., Burstyn, I., region-specific induction of astrocytic differen-
Karlsson, H., and Dalman, C. (2015). Maternal tiation and neurotrophin expression. Glia 26,
hospitalization with infection during pregnancy 191200.
and risk of autism spectrum disorders. Brain Masino, S.A., and Geiger, J.D. (2008). Are purines
Behav. Immun., 44, 100105. mediators of the anticonvulsant/neuroprotective
Legido, A., Jethva, R., and Goldenthal, M.J. (2013). effects of ketogenic diets? Trends Neurosci. 31,
Mitochondrial dysfunction in autism. Semin. 273278.
Pediatr. Neurol. 20, 163175. Masino, S.A., Kawamura, M., Jr., Plotkin, L.M.,
Lintas, C., Sacco, R., and Persico, A.M. (2012). Svedova, J., DiMario, F.J., Jr., and Eigsti, I.-E.
Genome-wide expression studies in autism spec- (2011a). The relationship between the neuro-
trum disorder, Rett syndrome, and Down syn- modulator adenosine and behavioral symptoms
drome. Neurobiol. Dis. 45,5768. of autism. Neurosci. Lett. 500,15.
Losh, M., Esserman, D., Anckarsater, H., Sullivan, P.F., Masino, S.A., Kawamura, M., Jr., Wasser, C.D.,
and Lichtenstein, P. (2012). Lower birth weight Pomeroy, L.T., and Ruskin, D.N. (2009).
indicates higher risk of autistic traits in discordant Adenosine, ketogenic diet and epilepsy: The
twin pairs. Psychol. Med. 42, 10911102. emerging therapeutic relationship between
Lotter, V. (1966). Epidemiology of autistic conditions metabolism and brain activity. Curr.
in young children. Soc. Psychiatry 1, 124137. Neuropharmacol. 7, 257268.
Luongo, L., Petrelli, R., Gatta, L., Giordano, C., Masino, S.A., Li, T., Theofilas, P., Sandau, U., Ruskin,
Guida, F., Vita, P., Franchetti, P., Grifantini, D.N., Fredholm, B.B., Geiger, J.D., Aronica, E.,
M., de Novellis, V., Cappellacci, L., etal. (2012). and Boison, D. (2011b). A ketogenic diet sup-
5-Chloro-5-deoxy-()-ENBA, a potent and presses seizures in mice through adenosine A1
selective adenosine A1 receptor agonist, allevi- receptors. J. Clin. Invest. 121, 26792683.
ates neuropathic pain in mice through functional Masino, S.A., Svedova, J., Kawamura, M., Jr., DiMario,
glial and microglial changes without affecting F.D., Jr., and Eigsti, I.-M. (2011c). Adenosine and
motor or cardiovascular functions. Molecules 17, autismRecent research and a new perspective.
1371213726. In: AutismA Neurodevelopmental Journey
Malkova, N.V., Yu, C.Z., Hsiao, E.Y., Moore, M.J., from Genes to Behaviour, V. Eapen, ed. (Rijeka,
and Patterson, P.H. (2012). Maternal immune Croatia:InTech), pp. 103122.
activation yields offspring displaying mouse McCracken, J.T., Badashova, K.K., Posey, D.J.,
versions of the three core symptoms of autism. Aman, M.G., Scahill, L., Tierney, E., Arnold,
Brain Behav. Immun. 26, 607616. L.E., Vitiello, B., Whelan, F., Chuang, S.Z., etal.
Marie, S., Race, V., Nassogne, M.C., Vincent, M.F., (2014). Positive effects of methylphenidate on
and Van den Berghe, G. (2002). Mutation of a hyperactivity are moderated by monoaminergic
nuclear respiratory factor 2 binding site in the gene variants in children with autism spectrum
5 untranslated region of the ADSL gene in three disorders. Pharmacogen. J. 14, 295302.
patients with adenylosuccinate lyase deficiency. Meidenbauer, J.J., Mantis, J.G., and Seyfried, T.N.
Am. J.Hum. Genet. 71,1421. (2011). The EL mouse:Anatural model of autism
Martinez, C.C., Pyzik, P.L., and Kossoff, E.H. (2007). and epilepsy. Epilepsia 52, 347357.
Discontinuing the ketogenic diet in seizure-free Meyer, U., Murray, P.J., Urwyler, A., Yee, B.K.,
children: recurrence and risk factors. Epilepsia Schedlowski, M., and Feldon, J. (2008). Adult
48, 187190. behavioral and pharmacological dysfunctions
Martins, D.F., Mazzardo-Martins, L., Soldi, F., following disruption of the fetal brain balance
Stramosk, J., Piovezan, A.P., and Santos, between pro-inflammatory and IL-10-mediated
Autism Spectrum Disorders and Homeostasis 605

anti-inflammatory signaling. Mol. Psychiatry 13, multiple dysregulated biological pathways.


208221. Transl. Psychiatry 4,e433.
Mohebbi, M.R., Navipour, R., Seyedkazemi, M., Naviaux, J.C., Schuchbauer, M.A., Li, K., Risbrough,
Zamanian, H., and Khamseh, F. (2004). V.B., Powell, S.B., and Naviaux, R.K. (2014).
Adult-onset epilepsy and history of childhood Reversal of autism-like behaviors and metabo-
febrile seizures: A retrospective study. Neurol. lism in adult mice with single-dose antipuriner-
India 52, 463465. gic therapy. Transl. Psychiatry 4,e400.
Mohiuddin, S., and Ghaziuddin, M. (2013). Naviaux, R.K., Zolkipli, Z., Wang, L., Nakayama,
Psychopharmacology of autism spectrum disor- T., Naviaux, J.C., Le, T.P., Schuchbauer, M.A.,
ders:Aselective review. Autism 17, 645654. Rogac, M., Tang, Q., Dugan, L.L., et al. (2013).
Morgan, J.T., Chana, G., Pardo, C.A., Achim, C., Antipurinergic therapy corrects the autism-like
Semendeferi, K., Buckwalter, J., Courchesne, E., features in the poly(IC) mouse model. PLoS One
and Everall, I.P. (2010). Microglial activation 8, e57380.
and increased microglial density observed in the Neal, E.G., Chaffe, H., Schwartz, R.H., Lawson, M.S.,
dorsolateral prefrontal cortex in autism. Biol. Edwards, N., Fitzsimmons, G., Whitney, A., and
Psychiatry 68, 368376. Cross, J.H. (2009). A randomized trial of classical
Mouridsen, S.E., Rich, B., and Isager, T. (2011). A and medium-chain triglyceride ketogenic diets
longitudinal study of epilepsy and other central in the treatment of childhood epilepsy. Epilepsia
nervous system diseases in individuals with and 50, 11091117.
without a history of infantile autism. Brain Dev. Neely, L., Rispoli, M., Gerow, S., and Ninci, J. (2015).
33, 361366. Effects of antecedent exercise on academic
Mouridsen, S.E., Rich, B., Isager, T., and Nedergaard, engagement and stereotypy during instruction.
N.J. (2007). Autoimmune diseases in parents of Behav. Modif. 39,:98116.
children with infantile autism: A case-control Newby, A.C., Worku, Y., and Holmquist, C.A. (1985).
study. Dev. Med. Child Neurol. 49, 429432. Adenosine formation:Evidence for a direct bio-
Moy, S.S., Nadler, J.J., Young, N.B., Perez, A., chemical link with energy metabolism. Adv.
Holloway, L.P., Barbaro, R.P., Barbaro, J.R., Myocardiol. 6, 273284.
Wilson, L.M., Threadgill, D.W., Lauder, J.M., Noh, H.S., Lee, H.P., Kim, D.W., Kang, S.S., Cho,
etal. (2007). Mouse behavioral tasks relevant to G.J., Rho, J.M., and Choi, W.S. (2004). A cDNA
autism:Phenotypes of 10 inbred strains. Behav. microarray analysis of gene expression profiles
Brain Res. 176,420. in rat hippocampus following a ketogenic diet.
Moyal, W.N., Lord, C., and Walkup, J.T. (2014). Mol. Brain Res. 129,8087.
Quality of life in children and adolescents with Nye, C., and Brice, A. (2005). Combined vitamin
autism spectrum disorders: What is known B6-magnesium treatment in autism spec-
about the effects of pharmacotherapy? Paediatr. trum disorder. Cochrane Database Syst. Rev.,
Drugs 16, 123128. CD003497.
Munasinghe, S.A., Oliff, C., Finn, J., and Wray, J.A. Oliveira, G., Diogo, L., Grazina, M., Garcia, P.,
(2010). Digestive enzyme supplementation for Ataide, A., Marques, C., Miguel, T., Borges, L.,
autism spectrum disorders:Adouble-blind ran- Vicente, A.M., and Oliveira, C.R. (2005).
domized controlled trial. J. Autism Dev. Disord. Mitochondrial dysfunction in autism spectrum
40, 11311138. disorders:Apopulation-based study. Dev. Med.
Muratore, C.R., Hodgson, N.W., Trivedi, M.S., Child Neurol. 47, 185189.
Abdolmaleky, H.M., Persico, A.M., Lintas, Oriel, K.N., George, C.L., Peckus, R., and Semon,
C., De La Monte, S., and Deth, R.C. (2013). A. (2011). The effects of aerobic exercise on
Age-dependent decrease and alternative splic- academic engagement in young children with
ing of methionine synthase mRNA in human autism spectrum disorder. Pediatr. Phys. Ther.
cerebral cortex and an accelerated decrease in 23, 187193.
autism. PLoS One 8, e56927. Page, T., and Coleman, M. (2000). Purine metabo-
Musumeci, S.A., Bosco, P., Calabrese, G., Bakker, C., lism abnormalities in a hyperuricosuric subclass
De Sarro, G.B., Elia, M., Ferri, R., and Oostra, of autism. Biochim. Biophys. Acta 1500, 291296.
B.A. (2000). Audiogenic seizures susceptibil- Page, T., and Moseley, C. (2002). Metabolic
ity in transgenic mice with fragile X syndrome. treatment of hyperuricosuric autism. Prog.
Epilepsia 41,1923. Neuro-Psychopharmacol. Biol. Psychiatry 26,
Nardone, S., Sams, D.S., Reuveni, E., Getselter, D., 397400.
Oron, O., Karpuj, M., and Elliott, E. (2014). DNA Page, T., Yu, A., Fontanesi, J., and Nyhan, W.L.
methylation analysis of the autistic brain reveals (1997). Developmental disorder associated with
606 Part IV:Homeostatic Therapies

increased cellular nucleotidase activity. Proc. Pietropaolo, S., Goubran, M.G., Joffre, C., Aubert, A.,
Natl. Acad. Sci. USA 94, 1160111606. Lemaire-Mayo, V., Crusio, W.E., and Laye, S.
Park, M.J., Aja, S., Li, Q., Degano, A.L., Penati, J., (2014). Dietary supplementation of omega-3 fatty
Zhuo, J., Roe, C.R., and Ronnett, G.V. (2014). acids rescues fragile X phenotypes in Fmr1-Ko
Anaplerotic triheptanoin diet enhances mito- mice. Psychoneuroendocrinology 49, 119129.
chondrial substrate use to remodel the metabo- Pineda, E., Shin, D., You, S.J., Auvin, S., Sankar, R.,
lome and improve lifespan, motor function, and and Mazarati, A. (2013). Maternal immune acti-
sociability in MeCP2-null mice. PLoS One 9, vation promotes hippocampal kindling epilepto-
e109527. genesis in mice. Ann. Neurol. 74,1119.
Pascual, J.M., Liu, P., Mao, D., Kelly, D.I., Polimeni, M.A., Richdale, A.L., and Francis, A.J.
Hernandez, A., Sheng, M., Good, L.B., Ma, Q., (2005). A survey of sleep problems in autism,
Marin-Valencia, I., Zhang, X., et al. (2014). Aspergers disorder and typically developing
Triheptanoin for glucose transporter type Idefi- children. J. Intellect. Disabil. Res. 49, 260268.
ciency (G1D):modulation of human ictogenesis, Politte, L.C., and McDougle, C.J. (2014). Atypical
cerebral metabolic rate, and cognitive indices by antipsychotics in the treatment of children
a food supplement. JAMA Neurol. 71, 12551265. and adolescents with pervasive developmen-
Patel, A., Pyzik, P.L., Turner, Z., Rubenstein, J.E., tal disorders. Psychopharmacology (Berl.) 231,
and Kossoff, E.H. (2010). Long-term outcomes 10231036.
of children treated with the ketogenic diet in the Polo-Kantola, P., Lampi, K.M., Hinkka-Yli-Salomaki,
past. Epilepsia 51, 12771282. S., Gissler, M., Brown, A.S., and Sourander, A.
Pathak, D., Berthet, A., and Nakamura, K. (2013). (2014). Obstetric risk factors and autism spec-
Energy failure: Does it contribute to neurode- trum disorders in Finland. J. Pediatr. 164,
generation? Ann. Neurol. 74, 506516. 358365.
Patterson, P.H. (2009). Immune involvement in Posthuma, D., and Polderman, T.J. (2013). What have
schizophrenia and autism: Etiology, pathol- we learned from recent twin studies about the
ogy and animal models. Behav. Brain Res. 204, etiology of neurodevelopmental disorders? Curr.
313321. Opin. Neurol. 26, 111121.
Patterson, P.H. (2011a). Infectious Behavior: Qiu, L.F., Lu, T.J., Hu, X.L., Yi, Y.H., Liao, W.P., and
Brain-Immune Connections in Autism, Xiong, Z.Q. (2009). Limbic epileptogenesis in
Schizophrenia, and Depression (Cambridge, a mouse model of fragile X syndrome. Cereb.
MA: MIT Press). Cortex 19, 15041514.
Patterson, P.H. (2011b). Maternal infection and Radyushkin, K., Hammerschmidt, K., Boretius, S.,
immune involvement in autism. Trends Mol. Varoqueaux, F., El-Kordi, A., Ronnenberg, A.,
Med. 17, 389394. Winter, D., Frahm, J., Fischer, J., Brose, N., etal.
Peacock, G., Amendah, D., Ouyang, L., and Grosse, (2009). Neuroligin-3-deficient mice: Model
S.D. (2012). Autism spectrum disorders and health of a monogenic heritable form of autism with
care expenditures: The effects of co-occurring an olfactory deficit. Genes Brain Behav. 8,
conditions. J. Dev. Behav. Pediatr. 33,28. 416425.
Pearson, D.A., Santos, C.W., Aman, M.G., Arnold, L.E., Raz, R., Lerner-Geva, L., Leon, O., Chodick, G., and
Casat, C.D., Mansour, R., Lane, D.M., Loveland, Gabis, L.V. (2013). A survey of out-of-pocket
K.A., Bukstein, O.G., Jerger, S.W., et al. (2013). expenditures for children with autism spectrum
Effects of extended release methylpheni- disorder in Israel. J. Autism Dev. Disord. 43,
date treatment on ratings of attention-deficit/ 22952302.
hyperactivity disorder (ADHD) and associated Reilly, C., Atkinson, P., Das, K.B., Chin, R.F.,
behavior in children with autism spectrum dis- Aylett, S.E., Burch, V., Gillberg, C., Scott, R.C.,
orders and ADHD symptoms. J. Child Adolesc. and Neville, B.G. (2014). Neurobehavioral
Psychopharmacol. 23, 337351. comorbidities in children with active epi-
Peagarikano, O., Abrahams, B.S., Herman, E.I., lepsy:Apopulation-based study. Pediatrics 133,
Winden, K.D., Gdalyahu, A., Dong, H., e1586e1593.
Sonnenblick, L.I., Gruver, R., Almajano, J., Rossignol, D.A., and Frye, R.E. (2012). Mitochondrial
Bragin, A., et al. (2011). Absence of CNTNAP2 dysfunction in autism spectrum disor-
leads to epilepsy, neuronal migration abnormali- ders: A systematic review and meta-analysis.
ties, and core autism-related deficits. Cell 147, Mol. Psychiatry 17, 290314.
235246. Ruskin, D.N., Svedova, J., Cote, J.L., Sandau, U.,
Persico, A.M., and Napolioni, V. (2013). Autism Rho, J.M., Kawamura, M., Jr., Boison, D., and
genetics. Behav. Brain Res. 251, 95112. Masino, S.A. (2013). Ketogenic diet improves
Autism Spectrum Disorders and Homeostasis 607

core symptoms of autism in BTBR mice. PLoS Socaa, K., Nieoczym, D., Pierg, M., and Wla, P.
One 8, e65021. (2015). Role of the adenosine system and glucose
Rutter, M., Bailey, A., Bolton, P., and Le Couteur, restriction in the acute anticonvulsant effect of
A. (1994). Autism and known medical condi- caprylic acid in the 6Hz psychomotor seizure
tions: Myth and substance. J. Child Psychol. test in mice. Prog Neuropsychopharmacol Biol.
Psychiatry 35, 311322. Psychiatry 57C,4451.
Sala, M., Braida, D., Lentini, D., Busnelli, M., Spilioti, M., Evangeliou, A.E., Tramma, D.,
Bulgheroni, E., Capurro, V., Finardi, A., Theodoridou, Z., Metaxas, S., Michailidi,
Donzelli, A., Pattini, L., Rubino, T., etal. (2011). E., Bonti, E., Frysira, H., Haidopolou, A.,
Pharmacologic rescue of impaired cognitive flex- Asprangathou, D., et al. (2013). Evidence for
ibility, social deficits, increased aggression, and treatable inborn errors of metabolism in a cohort
seizure susceptibility in oxytocin receptor null of 187 Greek patients with autism spectrum dis-
mice:Aneurobehavioral model of autism. Biol. order (ASD). Front. Hum. Neurosci. 7,858.
Psychiatry 69, 875882. Srivastava, S., Baxa, U., Niu, G., Chen, X., and Veech,
Sandin, S., Lichtenstein, P., Kuja-Halkola, R., R.L. (2013). A ketogenic diet increases brown
Larsson, H., Hultman, C.M., and Reichenberg, adipose tissue mitochondrial proteins and UCP1
A. (2014). The familial risk of autism. JAMA 311, levels in mice. IUBMB Life 65,5866.
17701777. Stead, M., Bower, M., Brinkmann, B.H., Lee, K.,
Sansa, G., Carlson, C., Doyle, W., Weiner, H.L., Marsh, W.R., Meyer, F.B., Litt, B., Van Gompel,
Bluvstein, J., Barr, W., and Devinsky, O. (2011). J., and Worrell, G.A. (2010). Microseizures and
Medically refractory epilepsy in autism. Epilepsia the spatiotemporal scales of human partial epi-
52, 10711075. lepsy. Brain 133, 27892797.
Seung, H., Rogalski, Y., Shankar, M., and Elder, Sullivan, E.L., Nousen, E.K., and Chamlou, K.A.
J. (2007). The gluten-and casein-free diet and (2014). Maternal high fat diet consumption dur-
autism: Communication outcomes from a pre- ing the perinatal period programs offspring
liminary double-blind clinical trial. J. Med. behavior. Physiol. Behav. 123, 236242.
Speech-Lang. Pathol. 15, 337345. Sullivan, P.G., Rippy, N.A., Dorenbos, K.,
Shelton, J.F., Geraghty, E.M., Tancredi, D.J., Delwiche, Concepcion, R.C., Agarwal, A.K., and Rho, J.M.
L.D., Schmidt, R.J., Ritz, B., Hansen, R.L., and (2004). The ketogenic diet increases mitochon-
Hertz-Picciotto, I. (2014). Neurodevelopmental drial uncoupling protein levels and activity.
disorders and prenatal residential proximity to Ann. Neurol. 55, 576580.
agricultural pesticides: The CHARGE study. Suo, C., Liao, J., Lu, X., Fang, K., Hu, Y., Chen, L., Cao,
Environ. Health Perspect. 122, 11031109. D., Huang, T., Li, B., and Li, C. (2013). Efficacy
Siegel, M. (2012). Psychopharmacology of autism and safety of the ketogenic diet in Chinese chil-
spectrum disorder:Evidence and practice. Child dren. Seizure 22, 174178.
Adolesc. Psychiatr. Clin. N.Am. 21, 957973. Supekar, K., Iyer, T.P., Odriozola, P., and Menon, V.
Singh, R., Nieuwdorp, M., Ten Berge, I.J., Bemelman, (2014). The influence of gender and age on preva-
F.J., and Geerlings, S.E. (2014). The potential lence rates of comorbid disorders in autism. Paper
beneficial role of faecal microbiota transplanta- 16709 presented at the International Meeting for
tion in diseases other than Clostridium difficile Autism Research (Atlanta, GA,May).
infection. Clin. Microbiol. Infect., 20, 11191125. Taylor, L.E., Swerdfeger, A.L., and Eslick, G.D. (2014).
Sivendran, S., Patterson, D., Spiegel, E., McGown, I., Vaccines are not associated with autism: An
Cowley, D., and Colman, R.F. (2004). Two novel evidence-based meta-analysis of case-control
mutant human adenylosuccinate lyases (ASLs) and cohort studies. Vaccine 32, 36233629.
associated with autism and characterization of Tordjman, S., Somogyi, E., Coulon, N., Kermarrec, S.,
the equivalent mutant Bacillus subtilis ASL. J. Cohen, D., Bronsard, G., Bonnot, O.,
Biol. Chem. 279, 5378953797. Weismann-Arcache, C., Botbol, M., Lauth, B.,
Sivertsen, B., Posserud, M.B., Gillberg, C., et al. (2014). Gene environment interactions
Lundervold, A.J., and Hysing, M. (2012). Sleep in autism spectrum disorders:Role of epigenetic
problems in children with autism spectrum mechanisms. Front. Psychiatry5,53.
problems: A longitudinal population-based Torres, A.R. (2003). Is fever suppression involved in
study. Autism 16, 139150. the etiology of autism and neurodevelopmental
Smith, S.E.P., Li, J., Garbett, K., Mirnics, K., and disorders? BMC Pediatr.3,9.
Patterson, P.H. (2007). Maternal immune acti- Tosh, D.K., Paoletta, S., Deflorian, F., Phan, K., Moss,
vation alters fetal brain development through S.M., Gao, Z.G., Jiang, X., and Jacobson, K.A.
interleukin-6. J. Neurosci. 27, 1069510702. (2012). Structural sweet spot for A1 adenosine
608 Part IV:Homeostatic Therapies

receptor activation by truncated (N)-methanocarba Wang, L.W., Tancredi, D.J., and Thomas, D.W. (2011).
nucleosides: Receptor docking and potent anticon- The prevalence of gastrointestinal problems in
vulsant activity. J. Med. Chem. 55, 80758090. children across the United States with autism
Trasande, L., and Liu, Y. (2011). Reducing the stag- spectrum disorders from families with multiple
gering costs of environmental disease in chil- affected members. J. Dev. Behav. Pediatr. 32,
dren, estimated at $76.6 billion in 2008. Health 351360.
Aff. (Millwood) 30, 863870. Warreyn, P., van der Paelt, S., and Roeyers, H.
Troyb, E., Orinstein, A., Tyson, K., Helt, M., Eigsti, I.M., (2014). Social-communicative abilities as treat-
Stevens, M., and Fein, D. (2014). Academic abili- ment goals for preschool children with autism
ties in children and adolescents with a history of spectrum disorder: The importance of imita-
autism spectrum disorders who have achieved tion, joint attention, and play. Dev. Med. Child
optimal outcomes. Autism 18, 233243. Neurol. 56, 712716.
Tsai, L.Y. (1999). Psychopharmacology in autism. Watters, R.G., and Watters, W.E. (1980). Decreasing
Psychosom. Med. 61, 651665. self-stimulatory behavior with physical exercise
Tsai, L.Y., and Ghaziuddin, M. (2014). DSM-5 ASD in a group of autistic boys. J. Autism Dev. Disord.
moves forward into the past. J. Autism Dev. 10, 379387.
Disord. 44, 321330. Weissman, J.R., Kelley, R.I., Bauman, M.L., Cohen,
Tsakanikos, E., Costello, H., Holt, G., Bouras, N., B.H., Murray, K.F., Mitchell, R.L., Kern, R.L., and
Sturmey, P., and Newton, T. (2006). Natowicz, M.R. (2008). Mitochondrial disease
Psychopathology in adults with autism and in autism spectrum disorder patients: A cohort
intellectual disability. J. Autism Dev. Disord. 36, analysis. PLoS One 3,e3815.
11231129. Whiteley, P., Haracopos, D., Knivsberg, A.M.,
Tuchman, R., and Rapin, I. (2002). Epilepsy in Reichelt, K.L., Parlar, S., Jacobsen, J., Seim, A.,
autism. Lancet Neurol. 1, 352358. Pedersen, L., Schondel, M., and Shattock, P.
Tyson, K., Kelley, E., Fein, D., Orinstein, A., Troyb, E., (2010). The ScanBrit randomised, controlled,
Barton, M., Eigsti, I.M., Naigles, L., Schultz, R.T., single-blind study of a gluten- and casein-free
Stevens, M., et al. (2014). Language and ver- dietary intervention for children with autism
bal memory in individuals with a history of spectrum disorders. Nutr. Neurosci. 13, 87100.
autism spectrum disorders who have achieved Williams, B.L., Hornig, M., Buie, T., Bauman, M.L.,
optimal outcomes. J. Autism Dev. Disord. 44, Paik, M.C., Wick, I., Bennett, A., Jabado, O.,
648663. Hirschberg, D.L., and Lipkin, W.I. (2011).
Vargas, D.L., Nascimbene, C., Krishnan, C., Impaired carbohydrate digestion and transport
Zimmerman, A.W., and Pardo, C.A. (2005). and mucosal dysbiosis in the intestines of chil-
Neuroglial activation and neuroinflammation in dren with autism and gastrointestinal distur-
the brain of patients with autism. Ann. Neurol. bances. PLoS One 6, e24585.
57,6781. Williams, B.L., Hornig, M., Parekh, T., and Lipkin,
Vasa, R.A., Carroll, L.M., Nozzolillo, A.A., Mahajan, R., W.I. (2012). Application of novel PCR-based
Mazurek, M.O., Bennett, A.E., Wink, L.K., and methods for detection, quantitation, and phy-
Bernal, M.P. (2014). A systematic review of treat- logenetic characterization of Sutterella species
ments for anxiety in youth with autism spectrum in intestinal biopsy samples from children with
disorders. J. Autism Dev. Disord. 44, 32153229. autism and gastrointestinal disturbances. mBio
Villafuerte, S. (2011). Suggestive evidence on the 3, e00261e00211.
genetic link between mitochondria dysfunction Williams, K., Brignell, A., Randall, M., Silove, N., and
and autism. Acta Psychiatr. Scand. 123,95. Hazell, P. (2013). Selective serotonin reuptake
Von Lubitz, D.K., Lin, R.C., Bischofberger, N., inhibitors (SSRIs) for autism spectrum disorders
Beenhakker, M., Boyd, M., Lipartowska, R., and (ASD). Cochrane Database Syst. Rev. 8, CD004677.
Jacobson, K.A. (1999). Protection against isch- Witwer, A., and Lecavalier, L. (2005). Treatment
emic damage by adenosine amine congener, a incidence and patterns in children and adoles-
potent and selective adenosine A1 receptor ago- cents with autism spectrum disorders. J. Child
nist. Eur. J.Pharmacol. 369, 313317. Adolesc. Psychopharmacol. 15, 671681.
Wang, J., Zhou, X., Xia, W., Sun, C.H., Wu, L.J., Wang, Wla, P., Socaa, K., Nieoczym, D., arnowski, T.,
J.L., and Tomoda, A. (2012). Parent-reported arnowska, I., Czuczwar, S.J., and Gasior, M.
health care expenditures associated with autism (2015). Acute anticonvulsant effects of capric acid in
spectrum disorders in Heilongjiang province, seizure tests in mice. Prog. Neuropsychopharmacol.
China. BMC Health Serv. Res.12,7. Biol. Psychiatry 57C, 110116.
Autism Spectrum Disorders and Homeostasis 609

Wong, C.C., Meaburn, E.L., Ronald, A., Price, T.S., activation on the behavior of mouse offspring.
Jeffries, A.R., Schalkwyk, L.C., Plomin, R., and PLoS One 9, 104433.
Mill, J. (2014). Methylomic analysis of mono- Zerbo, O., Qian, Y., Toshida, C., Grether, J.K.,
zygotic twins discordant for autism spectrum Van Der Water, J., and Croen, L.A. (in press).
disorder and related behavioural traits. Mol. Maternal infection during pregnancy and autism
Psychiatry 19, 495503. spectrum disorders. J. Autism Dev. Disord.
World Health Organization (2010). International Zhao, Y., Fung, C., Shin, D., Shin, B.C., Thamotharan, S.,
Statistical Classification of Diseases and Related Sankar, R., Ehninger, D., Silva, A., and Devaskar,
Health Problems (Geneva: World Health S.U. (2010). Neuronal glucose transporter iso-
Organization). form 3 deficient mice demonstrate features of
Xu, G., Jing, J., Bowers, K., Liu, B., and Bao, W. autism spectrum disorders. Mol. Psychiatry 15,
(2014). Maternal diabetes and the risk of autism 286299.
spectrum disorders in the offspring: A system- Zhu, J., and Chu, C.T. (2010). Mitochondrial dys-
atic review and meta-analysis. J. Autism Dev. function in Parkinsons disease. J. Alzheimers
Disord. 44, 766775. Dis. 20 (Suppl.2), S325S334.
Xuan, I.C.Y., and Hampson, D.R. (2014).
Gender-dependent effects of maternal immune
INDEX

A1 receptor (A1R), 491. See also Adenosine receptor Acupuncture, 341349


caffeine on, 461,463 for addiction, 525526
A1 receptor (A1R) agonists, on chronic pain, 343,343f on adenosine
A 2A receptor (A 2AR), 491. See also Adenosine receptor analgesic actions of, 342343,343f
acupuncture activation of, 342347, 343f, 344f, 346f release of, in humans, 345346, 345f,346f
(See also Acupuncture) adenosine increase in,347
in Alzheimers disease, 464465 adenosine signaling in analgesia of, 343345,344f
caffeine and, 348, 461, 463465 ATP, ADP, and AMP increases from,347
in epilepsy, 404,404f biological mechanisms of,434
on glutamate,408 caffeine on,348
in Parkinsons disease, 491492 clinical benefits of, 341342
in seizure control,404 with deoxycoformycin,345
A 2A receptor (A 2AR) antagonists history of,341
in neuroprotection, 422,492 principles of,525
in Parkinsons disease,492 purinergic-mediated signaling by, other, 346347
Ab initio drugs, 231,237 for traumatic brain injury, 434435
Acamprosate,522 understanding, challenges and progress in, 341342
Acetoacetate (ACA), 250f,253 Adalimumab,166
on GABAergic neuron firing,254 Adaptive immunity, in epilepsy, 159160
on glutamate release, presynaptic,254 Addiction, 509527. See also specific substances
on reactive oxygen species levels,256 alternative therapies for, 522527
Acetylcholine acupuncture, 525526
in addiction, 516517 biologic compounds,522
in central fatigue,306 exercise, 523524
on neurogenesis, hippocampal, 205206 herbal remedies, 526527
in Parkinsons disease,490 mindfulness training and meditation, 524525
in REMS sleep, 315f, 316317 animal modeling of, 510511
on sleep and cognition, 319320 approved therapies for, 522523
after traumatic brain injury,431 compulsivity in,511
Acetylcholine receptor type ligand-gated ion channels, diagnosis of,509
51. See also specifictypes drug classification in,509
Acetylcholinesterase, after traumatic brain injury,431 endocrine function in, 518520
Acetyl-coenzyme A(acetyl-CoA) neurosteroids,520
carnitine on availability of, 272273 sex steroids, 518520
in ketogenesis, 250f, 252253 energy balance and metabolic disturbance in,
ketogenic diets on, 257258 520521
metabolites of,271 with epilepsy,512
Acetyl-coenzyme A(acetyl-CoA)/CoA, carnitine on, homeostatic control systems overview in,511
271, 272,273f inflammatory immune responsesin
Acetyl-L-carnitine,274 cytokines, 517518
Action potentials, transmembrane separation of histamines,518
ionsin,98 microglia,517
612 Index

Addiction (Cont.) TRPV1 activation on release of,342


mesocorticolimbic reward circuitry in, 509510 Adenosine 5-monophosphate (AMP), in addiction,512
N-acetylcysteine for,523 Adenosine deaminase (ADA),33
neuromodulator homeostasis in, 511514 Adenosine kinase (ADK), 32, 33, 404405,404f
adenosine, 511512 in Alzheimers seizures,465
dopamine,511 on DNA methylation,466
endocannabinoids, 513514 in epilepsy, upregulation of, 77f,86
serotonin, 512513 on memory, 466467
neurotransmitter and receptor homeostasis in, Adenosine kinase (ADK) inhibitors, therapeutic,38
514517 Adenosine receptor, 342. See also
cholinergic, 516517 A1 receptor (A1R); A 2A receptor(A 2AR)
GABA, 515516 actions of,461
glutamate, 514515 acupuncture on, 342347, 343f, 344f, 346f (See also
neurotrophic factors in,521 Acupuncture)
BDNF, 521522 in addiction, 511512
GDNF,522 caffeine on, 348,461
prevalence of,509 Alzheimers disease and, 462465
stages of,509 function of, 33,33f
Adenosine ADO, 3138,422 methylxanthines on, 318,411
acupuncture on, 343345,344f in Parkinsons disease, 491492
analgesic actions of, 342343,343f seizures and,465
increase in,347 signalingof,34
release of, in humans, 345346, 345f,346f subtypes and distribution of, 3334,33f
in addiction, 511512 Adenosine receptor blockers, on sleep-wake
in ADK/ADO system,461 transition,318
in Alzheimers disease, 452467 (See also Adenosine signaling
Alzheimers disease, adenosinein) in acupuncture analgesia, 343345,344f
astrocyte-released, 77f,8081 exercise and sleep on,411
in astrocytes Adenosine ADO therapy,3738
formation and regulation of, 33f,35 for epilepsy, 408,409
release of, 77,77f augmentation of, 407, 408,409
from ATP,3435 neuroprotective effects of, 38,422
in autism, 593,593f Adenosine triphosphate (ATP). See ATP (adenosine
on bioenergetics,3435 triphosphate)
on blood-brain barrier,35 Adiposity. See also Obesity and diabetes
brain levels of,3133 regulation of, 570571
in central fatigue, 306308,307f ADK/ADO,461
effect inversionin,38 ADO,461
in epilepsy, 404405, 404f,408 on DNA methylation,466
epigenetics of, 406407, 407f,409 ADO deficiency, in Alzheimers seizures,465
homeostasis of, 404405,404f Adrenergic neurotransmission. See also specifictypes
exercise on, 303304 in Parkinsons disease,490
formation, metabolism, and transport of, 3233,32f Adsorptive-mediated transcytosis,147
functions of,461 Aerobic fermentation, of cancer cells (Warburg effect),
on GABAergic transmission,36 262, 555556,555f
on glutamatergic transmission,3637 Age, on epilepsy, 411412
for memory improvement, 465467 Aging
neuromodulation and homeostatic regulation by, in anxiety and stress disorders, 544545
33,33f carnitine and,273
as neuromodulator,3537 melatonin in,277
neurovascular couplingof,35 Aglycemia,102
on pain transmission, 342343,343f ATP production in,101
in Parkinsons disease, 491492 Agouti mouse,573
receptors for,461 Agouti-related peptide (AgRP), 576577
on seizures, 254,465 AIT-082,381
on sleep, 81, 305, 307,308 Albumin
on sleep-wake transition,318 blood-brain barrier disruption on,405
in synaptic plasticity,3637 on brain, 148,149
synthesisof,31 Alcohol
Index 613

on BDNF levels,522 nerve growth factor on,376


brain targets of,239 neural stem/progenitor cells in, 214215
on endocannabinoids,514 neurofibrillary tangles in, 214, 261, 452453
GDNF and,522 neurotrophic factors in,373
histaminergic drugs on responses to,518 pathogenesis of,384
on sleep,321 reactive astrocytosis in, 47,454
Alcohol addiction (alcohol use disorder), 509527. See seizures in,465
also Addiction symptoms in,452
alternative therapies for, 523527 synaptic scaling in,118
acupuncture,525 Tau protein in,454
daidzin and daidzein,526 tumor necrosis factor in,455
exercise, 523524 in twins, onset of,456
kudzu, 526527 vitamin B12 deficiency in,458
mindfulness training,525 Alzheimers disease, adenosine in, 452467
approved treatments for, 522523 amyloid plaques in, glial activation in,456
BDNF and,522 epigenetic changes and memory loss in, 456460
cholinergic homeostasis in, 516517 chromatin in,457
ghrelin on alcohol craving in,521 DNA methylation and methionine
pregnenolone sulfate and,520 cycle in,458
receptorsin DNA methylation and susceptibility genes in,
GABA, 515516 457458
muscarinic,516 epigenetic functions in,456
nicotinic acetylcholine,516 histone modifications in, 459460
serotonin homeostasis in, 512513 as homeostatic regulator, 461467
Allopregnanolone,520 ADK/ADO system in,461
Allostasis, 353,535 caffeine adenosine receptor targeting in,
Allostatic load,353 462465
Alpha 2-adrenergic (-2) receptors, in basal caffeine as cognitive normalizer in, 461462
ganglia,490 receptors for,461
-alpha-melanin-stimulating hormone (-MSH),574 seizures and,465
-linolenic acid (ALA), 284285 inflammation and, 454456
-synuclein,486 symptoms and lesions in, 453454
Alzheimers disease Amantadine hydrochloride (AMH), for traumatic
adenosine signalingin,35 brain injury,430
amyloid-beta peptide (A) in, 118, 214, 261, Amino acids. See also specifictypes
379,453 exercise and,306
p75NTR and,379 Aminos
BDNF in,376 blood-brain barrier on acid transport of,
blood-brain barrier dysfunction in,149 145146,146f
caffeine on risk of, 461462 Amitriptyline, on REMS sleep,431
clinical presentation and characteristics of, 214, Ammonia, in central fatigue,306
261,453 AMP-activated kinase (AMPK), ketogenic diets on,
DNA methylationin 260,263f
chromatin and,457 AMPA receptor,489
hypomethylation in, 458,460 cocaine, heroin, and methamphetamine on,
energy demands in,272t 514515
epidemiology of,214 scaling of,110
epilepsy in,465 Amphetamine. See also Methamphetamine
folate in, CSF,458 addiction to ( See also Addiction)
GSK3-signalingin,60 serotonin homeostasis in, 512513
on hippocampus, 214215,455 on BDNF levels,522
history of,452 histaminergic drugs on responses to,518
homeostatically controlled mechanismsin,35 on sleep, 317318
homocysteine in,458 Amygdala
inflammation in, 454456 in addiction, 509510
ketogenic diets on, 261262 in anxiety disorders,537
lesions in, 453454 in emotional processing, 536,537f
seizures, memory loss, and,456 in post-traumatic stress disorder,537
microglial cells in,455 in social anxiety disorder,537
614 Index

Amyloid-beta peptide(A) cholecystokinin in, 539540


in Alzheimers disease, 118, 214, 261, 379,453 classical treatmentof
p75NTR and,379 antidepressants,541
progression and plaque formation in,454 benzodiazepines, 540541
caffeine on,462 SSRIs and SNRIs, 540541
glial activation by,454 classification of, DSM-IV,538
p75NTR and,379 clinical presentationof
on promoter hypermethylation, 457458 in adults,543
Amyloid plaques in children, 543544
formation of,454 comorbidities of,545
glial activation in,456 CRH and glucocorticoids in,539
Amyotrophic lateral sclerosis(ALS) in elderly, 544545
branched chain amino acids for,281 emotional homeostasisin
energy demands in,272t dysfunctions of,538
ketogenic diets on, 263264 neurobiology of,535
neurotrophic factors in,373 neurochemical basis of, 535538
Anandamide,513 endocannabinoid system in,540
Anaplerosis, ketogenic diets on,259 future of,545
Anoxia, ATP production in,101 GABA and benzodiazepine systems in,539
Anterior cingulate cortex, in anxiety disorders, homeostatic therapies for, 540542
537,537f deep brain stimulation,543
exposure therapy on,543 deep transcranial magnetic stimulation,543
Antibody-associated encephalitides, adaptive immu- GABA agonist drugs,541
nity in,160 neural circuitries for, 536537,537f
Anticonvulsants. See also specificagents neuropeptide Y in,540
for seizures in autism,592 norepinephrine system in,539
for traumatic brain injury,432 prevalence of, by age,544
Antidepressants. See also specificagents serotonin system in,539
for anxiety and stress disorders,541 sex differences in,543
on neurogenesis APP C-terminal peptide (AICD),460
dentate granule cell,208 APP promoter, 457458
hippocampal, 213214 Aquaporin
for traumatic brain injury,431 in cytotoxic edema, 1314,13f
Antiepileptic drugs (AEDs), 407. See also specificagents functions of,104
adverse effects of,334 in potassium homeostasis,1011
elimination of, gender on,412 in vasogenic edema,14
failures of,401 Aquaporin 4 (AQP4)
multiple ion channel actions of,236 brain injury on,403
neuronal targets of,401 in cytotoxic edema, 1314,13f
target-centered rational strategy failure for,236 in epileptogenesis, 11,403
for traumatic brain injury,432 expression of,104
Antiglial fibrillary acidic protein autoantibody,424 functions of,104
Antihistamines, on sleep,318 in potassium homeostasis,1011
Anti-inflammatory therapy. See also specifictypes in vasogenic edema,14
for epilepsy, 409410 Arachidonoylethanolamide,513
for traumatic brain injury,432 2-Arachidonoylglycerol (2-AG), 513514
Antioxidants Arachnoid epithelium,3
ketogenic diets on, 255257 anatomy of,143
melatonin as,278 physiology of, 144145
sleep and,323 Arc, in synaptic scaling,180
Antipsychotics Arcuate hypothalamus, in obesity, 575, 576577
atypical ( See also specific agents) Aripiprazole, targets for,236
for seizures in autism,592 Arousal, adenosineon,36
on sleep,317 Aspartate
for schizophrenia, targets for,236 functions of,420
Anxiety and stress disorders, 535545 traumatic brain injury on, 420421
animal model translational studies on, 538539 Associative Hebbian plasticity, 124125
BDNF in,376 Associative learning, hippocampal long-term
in children and adolescents,544 potentiation in,124
Index 615

Associative synaptic plasticity,124 [K+]0 elevation in, on glycogen metabolism,103


Astrocyte,7590 [K+]0 in, neuronal activityon,98
adenosinein lactate astrocyte to axon transfer in, 102103
from ATP released, 77f,8081 in neurotransmitter homeostasis, 100101,101f
formation and regulation by, 33f,35 synaptic activityin,98
setpoints for tone of, 33f,35 Astrocyte neuron lactate shuttle hypothesis,103
ATP release in,7980 Astrocytic network,75
in blood-brain barrier, 144,144f Astrocytic transporters, 12,13f
in brain development,4 Astrocytosis, reactive. See Reactive astrocytosis
calcium excitability in,7576 Astroglial cells. See Astrocyte
cell volume homeostasis microdynamics of, 1217 Astrogliosis,47
(See also Cell volume homeostasis micrody- reactive ( See Reactive astrocytosis)
namics, astroglial) seizures from,465
channels expressedby,75 sleep and, 318,323
cytoarchitecture and distribution of, 3,4,5f on synaptic transmission and circuit
discoveryof,3 excitability,4748
distribution of,493 in temporal lobe epilepsies,47
D-serine release in, 77f, 7879,404 Atkins diet, modified, 251252,251f
in epilepsy, 8486,404 ATP (adenosine triphosphate)
ADK upregulation in, 77f,86 acupuncture on,347
glutamine synthetase downregulation in,8586 adenosine in,593
reactive astrocytosis in,8485 as adenosine source,3435
in epilepsy, and sleep,8184 alternate substrates for,570
on brain activity at circuit level,81 astrocyte release of, 77, 77f,7980
in inflammatory response and sleep behavior,84 adenosine from, 77f,8081
on sleep homeostasis, 77f, 8182,83f in brain homeostasis,322
sleep loss, depression, and,8384 brain levelsof,34
sleep loss, memory consolidation, and,8283 exercise and, 302, 303304,304f
functions of, 46, 5f, 75, 98,493 ketogenic diet on,259
GABA releasein,79 mitochondrial
glutamate releasein,78 in epilepsy,405
homeostasis regulation in, 36,5f sleep and,322
of calcium and sodium,1718 neuronal functionsof,34
human vs. rodent, quality of,105 requirements for,101
in Parkinsons disease, inflammatory reaction in, sleep on,322
493494 ATP13A2 (PARK9) Parkinsons disease, 484485
potassium permeability of,7,98 ATP-binding cassette proteins, in epilepsy,409
receptors expressedby,75 ATP-citrate lyase (ACL),257
signals of, physiologic stimuli for,76 Atypical antipsychotics. See also specificagents
SNARE complex in, 77,77f for seizures in autism,592
structureof,3 on sleep,317
swelling and cell volume recovery in,13f Atypical neuroleptics. See also specificagents
on transmitters modulating synaptic and neuronal for autism,592
activity, 7778,77f Autism spectrum disorder (ASD), 586599
types of,34 adenosine in, 593,593f
Astrocyteneuron interactions,98105 carnitine and,274
action potentialsin,98 comorbidities of, 587590
blood vessel diameter in, 104105 epilepsy, 163165, 164f, 587588
in CSF circulation and brain extracellular space general,587
cleansing,104 immunologic,590
energy metabolism in, 101102,102f metabolic, 588590
glycogen and memory in, 103104 neural, 587588
human vs. rodent,105 sleep disorders,589
interdependenceof,98 core features of, DSMV,586
in ion homeostasis, 98100 (See also specificions) energy demands in,272t
[K+]0, 9899,99f epidemiology of,586
pH0,99100 etiology of, 163164, 590592
pH changes and regulation, environmental,591
activity-induced,99100 epigenetic, 591592
616 Index

Autism spectrum disorder (ASD) (Cont.) hypothalamic, in obesity,579


genetic, 586587, 590591 -amyloid protein. See Amyloid-beta peptide(A)
expenditures on,587 -hydroxybutyrate (BHB), 250f,253
future directions in, 597598 on glutamate release, presynaptic,254
homeostasis disruption in, 586587 on histone deacetylases, 256257,261
ketogenic diets on,264 on reactive oxygen species levels,256
maternal immune activation in, 164, 164f,591 -lactam antibiotics, for epilepsy, 407408
prevention of,598 Bevacizumab
melatonin and,278 on brain tumor energy metabolism,558
mitochondrial metabolism in,274 for gliobastoma multiforme, 553554
prevention of,597 BHB dehydrogenase (BDH1),253
probiotics and,284 Bicarbonate equilibrium potential, 5253,52f
purinergic dysfunction in,593 subcellular heterogeneity of, 52,53f
risk factors for,586 Bienenstock, Cooper, and Munro (BCM) rule,133
symptoms of, heterogeneity of, 586587 Bioenergetics, adenosine on,3435
treatments for, homeostatic, 592598 Bioenergetics in epilepsy, mitochondrial,406
behavioral approaches,597 ketogenic diets on, 254255,255f
diet, 594596, 595f,596f ketogenic diets on bioenergics reserve in, 258259
early interventions,597 Biologics. See also specificagents
exercise,597 for addiction,522
microbiome,597 for traumatic brain injury, 435436
pharmacological, 592594,593f Bipolar disorder
principles,592 adenosine signaling in,3738
Autoimmune disease. See also specifictypes anxiety disorders with,545
with autism,590 energy demands in,272t
in traumatic brain injury, 424425 GSK3-signalingin,60
Autonomic nervous system homeostatically controlled mechanismsin,35
meditation on, 357358 Blood-brain barrier (BBB), 3, 143150
in Parkinsons disease,491 adenosineon,35
Axonal stretch/strain disruption, from traumatic brain anatomy of, 143144,144f
injury,420 development of,144
Axon guidance molecules,373 in disease, 148150
epilepsy, 149, 405,409
Bacteroides fragilis, for autism,597 general mechanisms, 148149
Basal ganglia, 299. See also neurological,149
specific parts and neurotransmitters neuronal network, 149150
in motor function,488 drug delivery in,148
in Parkinsons disease,488 glucose and amino acid transport in, 145146,146f
signal processing pathways in,488 inflammation on,156
Basement membrane, in blood-brain barrier,143 ion transport in, 146147,146f
Basic FGF,379 leukocyte migration across,145
BCL-2-associated agonist of cell death (BAD),254 neuronal and vascular functions of,148
BDNF-TrkB signaling, 376, 377, 405, 410,578 neurovascular unit in, 3, 143144,144f
BDNF val66met,427 peptides and proteins in, 147148
Benzodiazepine physiology of, 144145,146f
addiction to ( See also Addiction) traumatic brain injury on, 47, 421422
GABA receptors in,515 Blood-CSF barrier,143
for anxiety and stress disorders, 540541 Blood pressure, meditation on,355
Benzodiazepine-receptor agonists, on anxiety, 539,540 Blood vessel diameter, astrocyteneuron interactions
Benzodiazepine receptor, in anxiety,539 in, 104105
Benzodiazepine system, in anxiety and stress Body weight
disorders,539 BDNF on, 577578
Bergmann glia,4 excess ( See Obesity and diabetes)
bestrophin-1,16 gut microbiota on,581
[Ca 2+]i on,1718 regulation of, 570571
Beta-cell lymphoma 2 (BCL-2), in traumatic brain sleep on,322
injury recovery,423 Botanicals, 333338
Beta-oxidation for addiction, 526527
carnitine palmitoyltransferase 1A on,578 Cannabis sativa,335
Index 617

CNS disorder treatment improvement and,336 risk of, 461462


curcumin, 334335 on A-induced decline,462
evidence-based medicine for, 336337 on BACE1 expression,462
herbal extracted compounds, 335336 cognitive effects of, 461462
history and current use of,333 complex actions of,348
Huperzine A(Huperzia serrata), 335336 on epilepsy,411
mixtures of,337f prevalence of use of,348
mixtures of active compounds in, 334335 on sleep, 321322
mixtures of herbs in,334 Calcium excitability, in astrocytes,7576
newly discovered targets in, products for, 337338 Calcium, intracellular ([Ca 2+]i)
Brain. See also specifictopics in astrocyte homeostasis regulation,1718
function and complexity of, 231232 on cerebral vascular tone,17
integrative activity of,239 on neuronal excitability,17
metabolism of,232 Calcium sliding threshold,136
after traumatic brain injury, atrophy of, 425426 Caloric restriction, 252, 559560,559f
Brain cancer. See also specifictypes Calpain,61
blood-brain barrier dysfunction in,149 Canakinumab,166
epigenetics in,194 Cancer. See also Brain cancer; specifictypes
ketogenic diets on,262 energy demands in,272t
malignant, 553561 (See also epigenetics in,194
Glioblastoma multiforme) somatic mutations in,553
Brain-derived neurotrophic factor (BDNF), 373, Cannabinoid addiction. See
376377 Addiction; Marijuana addiction
in addiction, 521522 Cannabis sativa addiction, 335, 509527
on body weight, 577578 endocannabinoid homeostasis in, 513514
in epilepsy,405 mindfulness training on,525
in neuroprotection,422 Capsaicin, on adenosine concentrations,342
in Parkinsons disease, 376,495 Carbamazepine. See also Antiepileptic drugs(AEDs)
therapeutic,377 targets and mechanisms of action of,407
after traumatic brain injury,425 for traumatic brain injury,432
in depression,427 Carbonic anhydrase, on bicarbonate in chloride chan-
Brain trauma. See Traumatic brain injury(TBI) nels, 5253,53f
Branched chain amino acids (BCAAs), 278283 L-Carnitine, 271274,272f
carnitine on,273 on acetyl-CoA availability, 272273
in central fatigue, 306,307f acetyl-L-carnitine and,274
for CNS disease, 280281 on acyl-Co-A/CoA, 271, 272,273f
CNS functions of, 279280,280f aging and,273
epilepsy and, 281282,282f autism and,274
on glutamate homeostasis, 279280, 280f, 281,282f dietary sources of,271
ketogenic diet and, 282283 functions of, 271272
metabolism of, 278279,279f homeostatic mechanisms on,271
Brevican,373 on long-chain fatty acid transport, 271,272f
Buddhist meditation, 360. See also Meditation; on metabolism
Mindfulness-based stress reduction(MBSR) carbohydrate and protein, 271272
Buprenorphine, for opioid addiction,523 lipid, 271,272f
Bursts of spikes, in heterosynaptic plasticity, 131, valproate and,274
132,132f Carnitine deficiency syndromes,273
Carnitine esters, 271274,272f
[Ca 2+] Carnitine palmitoyltransferase 1A (Cpt1a)
in heterosynaptic plasticity,131 on beta-oxidation,578
in neuronal hyperactivity,195 malonyl-CoA on,578
[Ca 2+]0, blood-brain barrier on,147 Cation channels. See also specifictypes
Caffeine in astrocyte volume regulation,1617
on acupuncture effects,348 CB1 cannabinoid receptor, 513514,541
on adenosine receptors, 318, 411, 461, 463465 CB2 cannabinoid receptor,541
adenosine receptor targeting of, and Alzheimers CBD
disease, 462465 for anxiety and stress disorders, 541,542
on Alzheimers disease effects of,335
as cognitive normalizer, 461462 CCR2,455
618 Index

CD4+T lymphocytes, in seizure models,161 Cholecystokinin (CKK), in anxiety and stress disor-
CD8+T lymphocytes ders, 539540
in Rasmussens encephalitis,160 Cholinergic receptors and neurotransmission. See also
in seizure models,161 specifictypes
Ceftriaxone, for epilepsy, 407408 in addiction, 516517
Celecoxib in Parkinsons disease, 490491
on epilepsy from brain insults,409 Cholinergics. See also specificagents
on major depressive disorder,162 for traumatic brain injury,431
Cell adhesion molecules,373 Chondroitin sulphate proteoglycan,373
Cell-based replacement therapy,374 Choroid plexus,3
Cell volume homeostasis microdynamics, Choroid plexus epithelium
astroglial,1217 anatomy of,143
cytotoxic edema in, 12,13f physiology of, 144145
fundamentalsof,12 Chromatin, in Alzheimers disease,457
plasma membrane channels in, 1217 (See also Chromatin remodelers,192
Cell volume regulation, astroglial) Chronic intrinsic plasticity,5657
regulatory volume decrease in, 12,13f Chronicpain
vasogenic edemain,12 A1 receptor agonists on, 343,343f
Cell volume regulation, astroglial, 12,13f acupuncture for, 343, 343f (See also Acupuncture)
cation channels in,1617 definition of,348
chloride channels in,1516 prevalence of,348
potassium channelsin,14 Chronic traumatic encephalopathy (CTE),426
principles of plasma membrane channels in, 12,13f CIC-2,15
water channels in,1314 CIC receptor family,1516
Central fatigue, exercise and, 305308,307f Circadian pacemaker,317
Ceramide,379 Citalopram. See also
Cerebellum,299 Selective serotonin reuptake inhibitors (SSRIs)
Cerebral atrophy, after traumatic brain injury, 425426 for anxiety disorders,541
Cerebral blood flow, in exercise, 300301 efficacy of,431
Cerebral hyperperfusion syndrome, blood-brain bar- for traumatic brain injury,431
rier breakdown in,149 Cl-/ HCO3- exchanger,100
Cerebral ischemia, from traumatic brain injury,422 Clozapine, targets for,236
Cerebral metabolic ratio, 302303 Cocaine
Cerebrospinal fluid(CSF) on AMPA receptors, 514515
circulation of, and extracellular space on BDNF levels,522
cleansing,104 estrous cycle on use of,519
production and flow of,104 GDNF and,522
Change, stability and,108 on glutamate,514
Channelopathies,60 on sleep, 317318
ChemBank, 240241 Cocaine addiction, 509527. See also Addiction
ChEMBL,241 acupuncture for,525
Chemokines. See also specifictypes cholinergic homeostasis in, 516517
on adhesion molecules,161 exercise for,524
astrocytes on,421 GABA receptors in,516
in autism,590 mindfulness training for,525
in immunotherapy, depression from,162 N-acetylcysteine for,523
in inflammation, 421,517 nicotinic acetylcholine receptors in,516
in epilepsy, 155, 156,157t serotonin homeostasis in, 512513
in Parkinsons disease,492 Cognition
NF-kB in,517 complexity of, 239,240f
Chloride channels epigenetic chomatin modifications in,195
in astrocyte volume regulation,1516 exercise on,308
inhibition via opening of, 51,52f nootropic drugs on, 239,240f
Chloride equilibrium potentials, subcellular heteroge- pharmacologic support of,231
neity of, 52,53f sleep on, 319320
Chloride transporters, cation. See also specifictypes Cognitive behavioral therapy
functionof,52 for anxiety and stress disorders, 542543
potassium-chloride cotransporter in, 52,53f for autism, 597598
sodium-potassium-chloride cotransporterin,52 Cognitive impairment
Index 619

from epilepsy, inflammation in, 165166,165f ketogenic diets on,277


life expectancy on, increased,237 metabolism disorders of, 274275
from traumatic brain injury,427 neuroprotection by, 274275
Cognitive rehabilitation, for traumatic brain injury, for Parkinsons disease, 274275
433434 CREB-binding protein (CREBBP), 193194
Compassion meditation, 354355. See also Meditation CRF1 antagonists, on anxiety,540
Competition, 127128 Curcumin, 334335
in heterosynaptic plasticity, 129130, 134136 for traumatic brain injury,436
normalization in,127 Cushings disease,575
synaptic proopiomelanocortin in,576
Hebbian plasticity and, 126,127 Cys-loop receptors, 51. See also specifictypes
in synaptic scaling,116 Cytokines. See also specifictypes
Complementary health approaches (CHA), 333. See in addiction, 517518
also specifictypes astryocyte upregulation of, 149150
Compulsivity,511 in autism,590
Control theory, 113,114f blood-brain barrier transport of,146f
proportional-integral-derivative controller in,115 in central fatigue, after exercise,306
synaptic scaling in, 113115,114f classification of,517
Contusions, from traumatic brain injury,420 in depression after traumatic brain injury,427
Cooperativity, rule of,124 exercise on,433
Cortical remapping,180 functions of,517
Cortical reorganization, 180181,181f in inflammation
Corticotropin-releasing factor (CRF) receptor, gender in epilepsy, 155159, 157t, 161, 164, 405406
differences in,543 in posttraumatic epilepsy,426
Corticotropin releasing hormone(CRH) in traumatic brain injury, 421422,
in anxiety and stress disorders,539 426428,432
on sleep and stress responses, 320321 maternal immune activation on, 164165
in stress response,535 meditation on,356
Cortisol neurotrophic,521
in addiction,525 in Parkinsons disease, 492,493
FKBP5 polymorphisms on,540 sleep and, 324,324f
on hippocampus,359 Cytokine storm,164
meditation on, 358359 Cytomegalovirus, human (HCMV)
after traumatic brain injury,429 in brain cancer,557
Cotransporters. See also specifictypes on tumor cell energy metabolism,557
KCC2, 5254, 53f,57,59 Cytotoxic CD8+T lymphocytes, in Rasmussens
Na+/ HCO3-,100 encephalitis,160
in neuronal excitability homeostasis,52 Cytotoxic edema, 12,13f
sodium-potassium-chloride, 6, 52, 53f,59 AQP4 in, 1314,13f
COX-2 after traumatic brain injury,420
in inflammatory cascade, 157t,159
on learning and memory,165 Daidzein, for addiction,526
COX-2 inhibitors Daidzin, for addiction,526
on epilepsy D-amphetamine (AMPH), for traumatic brain
from brain insults,409 injury,430
inflammation in,166 DCBIP, on infarct size in cerebral artery occlusion,
on major depressive disorder,162 15,1819
CpG islands (CGIs), 187188,457 Deep brain stimulation
orphan,188 for anxiety and stress disorders,542
CpG methylation, 187,457 for depression and OCD,543
Craniopharyngioma, hypothalamic,572 Deep transcranial magnetic stimulation, for
C-reactive protein (CRP), meditation on,356 PTSD,543
Creatine, 274277 Dehydroepiandrosterone sulfate (DHEAS), in
biosynthesis and metabolism of, 275,276f addiction,520
blood-brain barrier crossing by,275 9-Tetrahydrocannabinol (THC), 513514
brain trauma and, 275276 for posttraumatic stress disorder,541
distribution of,275 Dentate granule cell neurogenesis, 202, 203f,204
epilepsy and, 276277 antidepressants on,208
for Huntingtons and Parkinsons diseases, 274275 functional significance of, 206208
620 Index

Dentate granule cell neurogenesis (Cont.) DNA demethylation, 188189,188f


in memory and learning,206 DNA hypermethylation, in epilepsy, 406, 407f, 408409
in pattern separation,207 DNA hypomethylation
population and turnover of,207 in Alzheimers disease, 458,460
in psychiatric disorders,208 in traumatic brain injury,409
Dentate gyrus (DG). See also Hippocampus DNA methylation, 187188,188f
neural stem/progenitor cells in, 203f, 204205 adenosine kinase on,466
neurogenesis in, adult, 202204,203f ADO tone on,466
Deoxycoformycin, acupuncture with,345 in Alzheimers disease
Depressant addiction, 509527. See also Addiction chromatin and,457
Depression methionine cycle and,458
adenosine signalingin,35 susceptibility genes and, 457458
anxiety with,545 definition of,457
deep brain stimulation for,543 in epilepsy, 406, 407f, 408409
on hippocampus, size,213 functions and locations of,457
homeostatically controlled mechanismsin,35 in gene silencing, 190,195
inflammationin S-adenosylhomocysteine hydrolase in, 458,465
with epilepsy, 162163,163f S-adenosyl methionine in, 188, 189f, 193,465
after traumatic brain injury, 427428 vitamin B on,458
neural stem/progenitor cells and neurogenesis for, DNA methylome restoration, for epilepsy, 407f,
213214 408409
in Parkinsons disease, 482, 487,488 DNA methyltransferase (DNMT),187
GABAergic neurotransmission in,490 Docosahexaenoic acid (DHA), 284285
glutamatergic neurotransmission in,489 Dopamine(DA)
noradrenergic neurotransmission in,490 in addiction, 511,512
serotonergic neurotransmission in,491 adenosine on systems for,37
sleep and, 321322 in central fatigue,306
sleep loss, astrocytes, and,8384 exercise on, 304305
Descartes, Ren,232 in Parkinsons disease, 427, 481,488
Desipramine, on REMS sleep,431 after traumatic brain injury, in cognitive
Development,brain dysfunction,427
epigenetics in, 192193 traumatic brain injury on,421
learning and, 108109 Dopamine-enhancing agents, on sleep, 317318
Dexamethasone Dopamine-inhibiting agents, on sleep,317
on brain tumor energy metabolism, 557558,558f Dopamine neuron loss, in Parkinsons disease, 481,
for gliobastoma multiforme,553 488489
DHEAS, in addiction,520 in striatum,488
Diabetes, 570581. See also Obesity and diabetes in substantia nigra,488
diabetes (db) gene, 573575 Dopamine quinones, in Parkinsons disease,496
Diaschisis Dopamine receptors,488
definition of,427 Drug discovery. See also specific drugs and disorders
after traumatic brain injury,427 magic shotguns in,236
Diet, 271285. See also multitarget pharmacology in, 235236
Ketogenic diet (KD); specifictypes phenotypic vs. target-based screening in,236
for autism, 594596, 595f,596f rational,235
on brain function,248 target-based, 234235
branched chain amino acids in, 278283 Drugs ofabuse
creatine in, 274277 addiction to, 509527 (See also Addiction)
on energy demands, 271,272t classification of,509
for epilepsy, 410411 D-serine, 77f, 7879,404
L-carnitine and carnitine esters in, 271274,272f Dystrophin-associated protein complex (DAPC)
melatonin in, 277278 AQP4in,10
omega-3 fatty acids in, 284285 AQP4Kir4.1 interactions in,1011
probiotics in,284 Kir4.1in,9
therapies based on,248 Dzogchen meditation, 360. See also Meditation
Diffuse axonal injury (DAI),420
Disease-associated molecular networks, 240241 EAAC-1 (excitatory amino acid carrier-1 transporters)
Disulfiram,522 in GABA reuptake,45
DJ-1 (PARK7) Parkinsons disease,484 in glutamate uptake,4546
Index 621

Ecstasy addiction, 509527. See also Addiction definition of, 187,424


serotonin homeostasis in, 512513 DNA demethylation in, 188189,188f
Ectonucleoside transporters (ENTs), in astrocytes,77f DNA methylation in, 187188,188f
Ectonucleotidases. See also Acupuncture in epilepsy, 195,409
local injection of,347 adenosine in, 406407, 407f,409
Edema functions of,456
cytotoxic, 12,13f histone modification in, posttranslational,
AQP4 in, 1314,13f 189190,189f
after traumatic brain injury,420 in learning, 193194
glibenclamide on,16,19 in memory, 193195
vasogenic,12,14 in neurodevelopmental disorders,233
Eicosapentaenoic acid (EPA), 284285 in neurogenesis and brain development, 192193
EIF4G1 Parkinsons disease,484 in neuronal membrane excitation,195
Emergent properties, 233, 234,242 noncoding RNAs in, 190191,191f
Emotional homeostasis in synaptic plasticity, 193194
anxiety and stress disorders on, 538 (See also in systems pharmacology, 233234,234f
Anxiety and stress disorders) in traumatic brain injury,424
dysfunctions of,538 Epilepsy, 401407. See also
neurobiology of,535 Temporal lobe epilepsy(TLE)
neurochemical basis of, 535538 A1 receptors in, 404,404f
sex differences in,543 addiction with,512
Emotional processing,535 adenosine homeostasis in, 404405,404f
Encephalitides, antibody-associated,160 adenosine signaling in,3738
Endocannabinoid receptors,513 in Alzheimers disease,465
Endocannabinoids AQP4 in, 11,403
in addiction, 513514 astrocytes in, 8486,404
endogenous,513 ADK upregulation in, 77f,86
Endocannabinoid (eCB) system glutamine synthetase downregulation in,8586
in anxiety and stress disorders,540 with autism, 587588
drugs enhancing, for anxiety and stress inflammation in, 163165,164f
disorders,541 BDNF in,376
Endocytic vesicles, on blood-brain barrier,147 blood-brain barrier breakdown in, 149,405
Energy balance. See also Mitochondrial bioenergetics brain injury in,409
in addiction, 520521 branched chain amino acids on, 281282,282f
ventromedial hypothalamus in, 571573 clinical manifestations of, 6263,84
Energy deprivation, ATP in,101 connexinsin,11
Energy metabolism creatine and, 275, 276277
astrocytes in, 101102,102f definition of, 333334
in cancer cells (Warburg effect), 262, 555556,555f DNA hypermethylation in, 406, 407f, 408409
in malignant brain tumors, standard of care on, DNA methylation in,406
557559,558f energy demands in,272t
Environmental factors. See also specifictypes epidemiology of,401
in autism,591 epigenetics in, 195, 233234, 234f,409
in epilepsy, 233234,234f adenosine and, 406407, 407f,409
in Parkinsons disease, 485, 486487 geneenvironment interactions in, 233234,234f
Ephrins,373 glial dysfunction and gliotransmitter homeostasis
Epigenetics, 187195 in, 403404
of Alzheimers disease, 456460 glial fibrillary acidic protein and vimentin in, 47, 85,
chromatin in,457 87, 88f89f
DNA methylation and methionine cycle in,458 gliotransmission in, 8690, 403404
DNA methylation and susceptibility genes in, glutamate release in,8687
457458 NMDA receptor trafficking in, synaptic, 8790,
functions of,456 88f89f
histone modifications in, 459460 glutamate and glutamine homeostasis in,403
in autism, 591592 glutamate-glutamine cyclein,47
in brain development, 192193 growth factors in,405
in brain tumors,194 homeostatically controlled mechanismsin,35
chromatin remodelers in,192 IL-1R1 signaling in, 157158,157t
in cognition,195 IL-1R/TLR signaling pathway in, 405406
622 Index

Epilepsy (Cont.) in adenosine transport,3334


immunomodulatory and inflammatory responses Equilibrium potential,51
in, 405406 ERK, 377378
intrinsic plasticity in, 6061,62f ERK signaling, second messengers on,384
ion and water homeostasis in,403 Estradiol
ketogenic diets on, 248261, 410411 (See also in addiction, 518520
Ketogenic diet (KD), on epilepsy) on dopamine release, cocaine-induced,519
Kir4.1 in,9,403 for traumatic brain injury, 423,435
mitochondrial bioenergetics in,406 Estrogen
multidrug transporter expression in,409 on GABA and GABA receptors, 411412
network homeostasis disruption in, 401403,402f on NMDA receptors,412
nonneuronal factors in,401 Ethanol. See Alcohol
pannexin 1in,12 Ethosuximide. See also Antiepileptic drugs(AEDs)
pathophysiology of,401 on sleep,314
pharmacoresistance in,409 Excitability homeostasis, neuronal,5163
potassium channelrelated,60 cotransportersin,52
prevalenceof,84 epilepsy-related
reactive astrocytosis in, 47,8485 intrinsic plasticity in, 6061,62f
synaptic plasticity in, 6163,62f synaptic plasticity in, 6163,62f
TLR4 signaling in, 157158,157t GABA and glycine receptor inhibition in, 5154,
after traumatic brain injury,426 52f,53f
water and [K+]0 homeostasisin,11 inhibitory plasticity in, neuronal,5760
Epilepsy, homeostatic therapies for, 407412 phosphorylation in,5960
adenosine augmentation in, 407, 408,409 RNA splicing and editing in,5759
age and gender on, 411412 intrinsic plasticity in,5456
anti-inflammatory and immunomodulatory thera- chronic,5657
pies in, 409410 definitionof,54
blood-brain barrier drug transporters in,409 short-term,5455
dietary interventions in, 410411 synaptic plasticityrelated,5556
DNA methylome restoration in, 407f, 408409 mitochondrial function and, 254255,255f
glutamate homeostasis restoration in, 407408 Excitation-spike (E-S) coupling,55
lifestyle choices in,411 Excitatory amino acids(EAA)
methylxanthines on,411 functions of,420
network homeostasis reconstruction in,407 traumatic brain injury on, 420421
Epilepsy, inflammation and immunomodulation in, Excitatory amino acid transporter 2 (EAAT2,
155166 GLT-1),515
adaptive immunity in, 159160 in GABA reuptake,45
comorbidities and, 161165 in glutamate transport,515
autism, 163165,164f in glutamate uptake,46
depression, 162163,163f localizationof,45
memory and cognitive impairments, Excitatory amino acid transporter (EAAT), in gluta-
165166,165f mate transport,515
neurobehavioral disorders, 161162 Excitatory synaptic scaling, 109112
evidence on, clinical,156 in interneurons,112
experimental studies on, 160162 in pyramidal neurons, 109112,111f
antibodies against intracellular antigens in,161 Excitotoxicity, from traumatic brain injury, 420421
antibodies against membrane antigens in, 160161 Exercise, 298308
leukocytes in,161 for addiction, 523524
glial cells in,155 on adenosine signaling,411
inflammatory cascade in, yin and yang of,159 amino acids and,306
inflammatory molecules and epileptogenesis in, for autism,597
158159 behavioral effects of,308
overview of,155 on brain,298
pathophysiology of, 155156 cerebral blood flow and O2 consumption in, 300301
seizure pathogenesis in, 156158,157t on epilepsy,411
therapeutic implications of,166 fatigue in, central, 305308,307f
Epistemological reductionism,232 metabolismin
Equilibrative nucleoside transporters (ENTs) glucose and lactate, 301303
in adenosine release,36 nucleotide and nucleoside, 303304,304f
Index 623

neural control of movement and, 298300,299f in children and adolescents,544


on neurogenesis,411 on cocaine and alcohol addiction behavior,513
neurotransmission in, 304305 on depression,241
dopamine, 304305 with epilepsy,163
glutamate,305 efficacy of,431
serotonin,305 on neurogenesis,213
outlook for,308 on serotonin,513
positive effects of,298 for traumatic brain injury,431
on sleep, 305, 307,308 Fluvoxamine. See also
for traumatic brain injury, 430433 Selective serotonin reuptake inhibitors (SSRIs)
tryptophan and,306 for anxiety disorders,541
Exposure therapy, for anxiety and stress disorders,542 in children and adolescents,544
Extracellular space cleansingf, and CSF circulation, Focal cortical dysplasia (FCD), inflammation in,156
astrocytes in,104 Focal injury,420
Extracted compounds, herbal, 335336 Focal ischemia, nerve growth factor on,376
Folate
Familial hemiplegic migraine-associated spreading functions of,458
depolarization,149 in neuronal homeostasis,193
Fatigue Folate deficiency, in Alzheimers disease,458
central, exercise and, 305308,307f Fragile X mental retardation protein (FMRP), in syn-
peripheral,305 aptic scaling,117
Fat mass and obesity associated (FTO) gene, 579580 Fragile X syndrome
Fatty acid amid hydrolysis (FAAH), in endocannabi- GSK3-signalingin,60
noid addiction,514 synaptic scaling in,117
Fatty acid oxidation(FAO) Free radicals, melatonin on,278
high-fat, low-carbohydrate diet on,252 FRS2/GRB2 complex,380
ketogenic diets on,258 Fructose-1,6-bisphosphate (FBP), on glycolysis,258
FBXO7 (PARK15) Parkinsons disease,485 Functional hyperemia, 104105
Fecal transplantation, for autism,597
Feeding, reward brain circuitry in,521 GABA. See also Glutamate
FGF2/FGFR1,380 in addiction, 515516
on PLC-gamma pathway, 380381 in anxiety and stress disorders,539
Fibroblast growth factor (FGF), 379380 astrocyte release of, 77, 77f,79
Fibroblast growth factor 2 (FGF2),373 discoveryof,51
Fibroblast growth factor (FGF) receptors,380 in epilepsy,403
Fibroblast growth factor (FGF) signaling pathway, estrogen on, 411412
380381 functions of,489
Fingolimod,166 glutamate-glutamine cycle on,4547
Firing rate homeostasis, 115116,118 glutamate in synthesisof,45
5-HT1A receptor, in inflammation-associated depres- on hippocampal neurogenesis, 205206
sion, 163,163f in inhibitory synaptic terminals,44
5-HT1B receptor, in addiction,512 in ketogenic diet,253
5-HT2A receptor, in addiction,512 in Parkinsons disease, 489490
5-HT2C receptor, in addiction,512 insleep
5-HT3, in addiction,512 NREMS, 315f,316f
5-HT3 receptor, in addiction,512 REMS, 315f,317
5-Hydroxyindoleacetic acid, in Parkinsons sourcesof
disease,491 in interneurons,4546
5-Hydroxytryptamine (serotonin) presynaptic,45
exercise on,305 synaptic cleft removalof,44
on hippocampal neurogenesis, 205206 GABA agonists, for anxiety and stress
in sleep,305 disorders,541
5-Nucleotidases,33 GABA A receptors (GABA AR), 51,489
FKBP5 polymorphisms, on cortisol and PTSD,540 in addiction, 515516
Flavonoids, for traumatic brain injury,436 neurosteroids and,520
Fluoxetine. See also anion channelsof,51
Selective serotonin reuptake inhibitors (SSRIs) in anxiety, 539540
on aggression,321 in chloride channel opening, 51,52f
for anxiety,541 phosphorylationof,59
624 Index

GABA B receptors,489 discoveryof,75


in addiction, 515516 in epilepsy, 155, 403404
GABA-enhancing drugs, on REMS sleep,317 Na+/ HCO3- cotransport,100
GABAergic neurotransmission Glial-derived neurotrophic factor (GDNF)
adenosineon,36 in addiction, 521,522
progesterone on,412 in Parkinsons disease,495
Gabapentin. See also Antiepileptic drugs(AEDs) Glial fibrillary acidic protein (GFAP), in epilepsy,85
for addiction,512 Glial scar, 373, 403404
on sleep,314 Glia-lymphatic system, sleep and, 323324
GABA receptors Glibenclamide, on cerebral edema,16,19
in addiction, 515516 Glioblastoma multiforme, 553561
estrogen on, 411412 cytomegalovirus in,557
inhibition via, 5154, 52f,53f epidemiology of,553
physiology of,515 glucose and glutamine on, 556557
GABA vesicular transporter (VIAAT),44 isocitrate dehydrogenase (IDH) gene mutations
Gamma-amino butyric acid. See GABA in,194
Gap junctions management of, current standard ofcare
astrocyte,11 energy metabolism in, acceleration from,
in K+ homeostasis,11 557559,558f
GBA, in Parkinsons disease,484 survival with, 553554, 554f, 558559
GCLM,256 management of, homeostatic
Gender future hopes for,561
on antiepileptic drug elimination,412 ketogenic diet in, calorie-restricted, 559561,559f
on corticotropin-releasing factor receptor,543 mitochondrial dysfunction exploitation in,
in epilepsy, 411412 559560,559f
in post-traumatic stress disorder symptoms,543 metastasis of, systemic,553
Geneenvironment interactions, 233234,234f mitochondrial dysfunction in, 554555,555f
General systems theory,234 reactive oxygen species in,553
Gene silencing,138 tumor-associated macrophages/monocytes in,553
DNA methylation in, 190,195 glutamate use by,558
histone methylation in,194 standard of care on,557
RNA-induced, 191,191f Warburg effect in, 555556,555f
Genetics Gliomas
in Alzheimers disease, 457458 isocitrate dehydrogenase (IDH) gene mutations
in autism, 586587, 590591 in,194
in obesity and diabetes ( See also mitochondrial dysfunction in, 554555,555f
under Obesity and diabetes) Gliotransmission
human, 579580 definitionof,75
mice, 573578,574 in epilepsy, 8690, 403404
in Parkinsons disease, 482485, 495 (See also under glutamate release in,8687
Parkinsons disease) NMDA receptor trafficking in, synaptic, 8790,
in synaptic scaling,180 88f89f
of traumatic brain injury,424 functionsof,75
Gephyrin,58 in sleep pressure accumulation, 8283,83f
calpain as substrate for,61 Gliotransmitters, 7778, 77f,178
in neuropsychiatric disease,60 in epilepsy, 403404
phosphorylation of,5960 Glucocorticoids. See also specifictypes
Ghrelin in anxiety and stress disorders,539
in addiction,521 on sleep and stress responses,320
on alcohol craving,521 sleep deprivation on, 320321
in obesity, 578579 in stress response,535
sleep on,322 Glucose
synthesis and role of, 520521 blood-brain barrier on, 145146,146f
Ginseng, for addiction,526 in brain cancer progression,556
GlialCAM, in megalencephalic exercise and metabolism of, 301303
leukoencephalopathy,19 in glioblastoma multiforme management,
Glialcells 559560,559f
in addiction,512 ventromedial hypothalamus on, 571573
amyloid plaque on,456 Glucose-regulated hypothalamic neurons,572
Index 625

Glucose transporters,145 in brain cancer progression, 556557


Glucostat hypothesis, 572,578 brain cancer therapy on, current, 557,558f
GLUT1,145 cytomegalovirus on,557
GLUT3,145 in epilepsy,403
GLUT4,145 Glutamine synthetase(GS)
Glutamate, 100, 489. See alsoGABA in epilepsy, 47,8586
in addiction, 512, 514515 for seizure prevention,408
astrocyte release of, 77, 77f, 78,408 in temporal lobe epilepsy, 47,403
astrocyte uptake of,408 Glutathione(GSH)
brain cancer treatment on, 557,558f mitochondrial, 255f,256
in epilepsy,8687 sleep deprivation on,323
in epilepsy, homeostasis of,403 Glutathione peroxidase7,262
exercise on,305 Glycemic index (GI),252
functions of,420 low-glycemic index treatment and, 251f,252
in GABA synthesis,45 Glycine receptor (GlyR),51
in ketogenic diet, 253,254 activation of, in chloride channel opening, 51,52f
release-elimination balance of,514 anion channelsof,51
in sleep,305 inhibition via, 5154, 52f,53f
synaptic cleft removalof,44 Glycogen
transport of, excitatory amino acid transporters in energy metabolism, 101102,102f
in,515 [K+]0 elevation in astrocyte metabolism of,103
traumatic brain injury on, 420421 in memory acquisition, 103104
Glutamate cysteine ligase (GCL),256 Glycolytic restriction/diversion, in ketogenic diets,
Glutamate decarboxylase (GAD),253 257258
Glutamate dehydrogenase (GDH),281 Growth arrest and DNA damage 45 (Gadd45)
Glutamate-glutaminecycle genes,189
astrocyte, 100101,101f Growth-promoting extracellular matrix molecules,373
in GABA recycling,4547 GSK3 inhibitors, on neuropsychiatric disease,60
in epilepsy,47 Guanosine
functions of,4445 in CNS signaling, 373374
in inhibitory synaptic transmission,46 on ischemia,382
Glutamate homeostasis,4448 on lipid homeostasis,382
astrocytes in, 100101,101f in neurogenesis and neurorestoration, 381,382
astrogliosis on,4748 neurotrophic effects of,381
branched chain amino acids in, 279280, 280f, signaling transduction cascade of, NGF, BDNF, and
281,282f FGF2 pathways and, 382383,383f
in epilepsy, restoration of, 407408 targeting convergent molecules in, 384385
GABA sources in, presynaptic,45 on stem/progenitor cell proliferation,382
GLT-1 and SN1in,45 Gut microbiota,284
glutamate-glutamine cycle in, astrocyte, 4445, in autism, 589,597
101102,102f on body weight,581
GABA recycling via,4547
transportersin,44 Hallucinogen addiction, 509527. See also Addiction
vesicular packagingin,44 Harvard Epilepsy Botanical Program,336
VIAATin,44 Heart rate variability (HRV), meditation on, 357358
Glutamate receptor 1 (mGluR1),76 Hebbian plasticity, 108, 124126, 136137
Glutamate receptor 5 (mGluR5),76 associative, 124125
Glutamate receptor, in addiction, 514515 runaway dynamics in,125
Glutamatergic neurotransmission spike-timing dependent plasticity rules in,126
adenosine on,3637 synaptic competition in, weak,127
in Parkinsons disease, 489490 Hebbs rule, 108,124
progesterone on,412 Herbal extracted compounds, 335336. See also
Glutamatergic therapies, for epilepsy, 407408 Botanicals
Glutamate transporter-1 (GLT-1, EAAT2),44 Herbal remedies. See Botanicals
in addiction,515 Heroin. See also Opioid addiction
in GABA reuptake,45 on AMPA receptors, 514515
in glutamate uptake,46 on mesocorticolimbic reward circuitry,510
localizationof,45 Heroin addiction, 509527. See also Addiction
Glutamine exercise on,524
626 Index

Heterosynaptic long-term depression,178 posttranslational modification of, 189190, 189f,457


Heterosynaptic long-term potentiation,178 ubiquitination of,190
Heterosynaptic plasticity, 124136 Histone acetyltransferase,189
definition of,130 Histone code, 189,189f
experimental evidence for, 130131 Histone deacetylases (HDACs),457
Hebbian plasticity and, 124126, 136137 (See also -hydroxybutyrate on, 256257,261
Hebbian plasticity) in epilepsy,195
induction scheme for, 137f,138 ketogenic diet on,263f
levels of homeostasis and,124 on learning and memory,193
modeling of, 126128 meditation on genes for,356
competition in, 134136 valproate on,237f
competition in, need for, 127128 Histone deacetylases (HDAC) inhibitors, for
runaway dynamics prevention in, 133134, Alzheimers disease, 459460
135f,136f HMG-CoA synthase 2 (HMG-CoAS2),253
stability through normalization in,127 Homeobox (Hox) genes,192
STDP rule balancing in,126 Homeostasis, brain. See also specifictopics
trigger and timescale in,128 levels of,124
weight changes in, synaptic,126 regulatory mechanisms overview for, 371372
properties of, 132133,132f Homeostatic response recovery,181
role of,124 Homeostatic synaptic scaling. See Synaptic scaling
runaway counteracting mechanisms in, other, Homer1a,180
136137 Homocysteine, in Alzheimers disease,458
synaptic scaling in, 128130 Homosynaptic long-term potentiation, 137f,138
experimental paradigm for, 129,130 heterosynaptic LTD with,130
timescale, competition, and runaway dynamics Homosynaptic plasticity,178
in, 129130 expression mechanisms of,131
tetanization induction of, 132133,132f Hebbian-type learning in, 124,125
trigger for,131 induction of, 126, 137f,138
weight dependence of,131 weight dependence of,131
High mobility group box 1 protein (HMGB1) Hormones, on neuronal excitability, 411412
in epilepsy, cognitive impairments in, 165166,165f 5-HT3, in addiction,512
in seizures, 157158, 157t,406 5-HT receptors
Hippocampal sclerosis in addiction,512
in Alzheimers disease,455 in inflammation-associated depression, 163,163f
in temporal lobe epilepsy, 60, 62f, 85, 87, 209,403 Hub cells,209
Hippocampus. See also specificareas Huntingtons disease
in addiction, 509510 adenosine signaling in,3738
Alzheimers disease on, 214215 basal ganglia dysfunction in,299
antidepressants on neurogenesis in, 208, 213214 BDNF in,376
in anxiety disorders,537 caffeine in,462
cortisol on,359 creatine for, 274275,278
in depression, size of,213 homeostatically controlled mechanismsin,35
in emotional processing, 536,537f mitochondrial dysfunction and NGF signaling
meditation on,360 in,375
neural stem/progenitor cells in, 203f, 204205 neurotrophic factors in,373
regulators of, 205206 Huperzine A(Huperzia serrata), 335336
neurogenesis in, adult, 202204, 203f,208 Hydrocephalus,14
stress-induced atrophy of, 359360 obstructive,14
Histamine Hydrogen peroxide (H2O2),256
in addiction,518 4-Hydroxy-2-nonenal (4-HNE),256
function of,518 3-Hydroxy-3-methylglutaryl CoA (HMG-CoA),
on sleep and cognition, 319320 250f,253
Histamine receptors, in addiction,518 5-Hydroxyindoleacetic acid, in Parkinsons
Histone disease,491
acetylation of, 189, 189f, 457,460 5-Hydroxymethyl-cytosine (5-hmC), in DNA methyla-
in memory formation,193 tion, 188189
in Alzheimers disease, 459460 5-Hydroxytryptamine (serotonin). See Serotonin
methylation of, 189190,189f (5-hydroxytryptamine,5-HT)
in memory consolidation,194 Hyperactivity, neuronal, Ca 2+ in,195
Index 627

Hyperbaric oxygen therapy (HBO2T), with ketogenic in Parkinsons disease ( See


diet, 560561 Neuroinflammation, in Parkinsons disease)
Hyperemia, functional, 104105 pathophysiology of, 155156
Hyperglycemia, in brain cancer progression,556 on sleep pressure,84
Hyperpolarization-activated, inward rectifier Cl- cur- from traumatic brain injury,421
rent (IClh),15 yin and yang of,159
Hypocretin,317 Infliximab,166
Hypogonadotropic hypogonadism, after traumatic Influenza type-A, amantadine hydrochloride for,430
brain injury,429 Inhalant addiction, 509527. See also Addiction
Hypothalamic craniopharyngioma,572 Inhibitory plasticity, neuronal,5760
Hypothalamic-pituitary-adrenal (HPA)axis phosphorylation in,5960
in allostatic regulation, 358,535 RNA splicing and editing in,5759
meditation on, 358360 Inhibitory synaptic scaling, 112113
in stress response, 535, 537538 Innate immunity,492
Hypothalamus, in obesity in Alzheimers disease, 455456
beta-oxidation in,579 in inflammation, 156158,157t
nutritional signals of, 578579 microglia in,493
Hypothalamus, ventromedial(VMH) Inosine,597
in energy balance and glucose homeostasis, 571573 Input specificity, rule of, 124125
on food intake and metabolic rate, 571572 In silico receptorome screening,241
in obesity, 573576 Insomnia. See also Sleep
melatonin and,278
IL-1R1 signaling, in epilepsy, 157158,157t Insula
IL-1ra, for epilepsy, from inflammation,166 in anxiety disorders,537
Immune function exposure therapy on,543
in epilepsy, 405406 meditation on,360
meditation on, 355357 Insulin
sleep on, 324,324f in addiction,521
Immunoglobulin G autoantibodies, on traumatic brain in obesity,578
injury,424 synthesis and functions of,521
Immunomodulation, in epilepsy, 155166. Insulin-like growth factor 1 (IGF-1), in brain cancer
See also Epilepsy, inflammation and progression,556
immunomodulationin Integrative approaches. See also specifictypes
Immunomodulatory therapies, for epilepsy, 409410 in biology,234
Impulsivity,511 in neuroscience, 232235,234f
in serotonergic drug addiction, 512513 Interleukin-1 receptor antagonist (IL-1ra), for trau-
Inborn errors of metabolism, energy demands in,272t matic brain injury,432
Infectious brain injuries, blood-brain barrier dysfunc- Interleukin-1/Toll-like receptor (IL-1R/TLR) signaling
tion in,149 pathway, in epilepsy, 405406
Inflammation Interleukin-1 (IL-1)
in addiction, 517518 in addiction, 517518
cytokines in, 517518 on epilepsy,410
in Alzheimers disease, 454456 in inflammation, 409410
in autism,590 depression with, 162163,163f
on blood-brain barrier,156 maternal, in autism in offspring, 163164,163f
in brain,155 sleep and, 324,324f
depressionand Interleukin-6 (IL-6)
with epilepsy, 162163,163f in addiction, 517518
after traumatic brain injury, 427428 maternal, in autism in offspring, 163,163f
in epilepsy, 155166, 405406 (See also Epilepsy, Interneurons
inflammation and immunomodulationin) excitatory synaptic scaling,112
IL-1 in, 409410 inhibitory synaptic scaling, 112113
innate immunity and, 156158,157t Interstitial space composition, ionic,98
ketogenic diets on,264 Intrinsic plasticity, 5456,182
maternal immune activation-induced chronic,5657
in autism, 164, 164f,591 definitionof,54
in autism, prevention of,598 in epilepsy, 6061,62f
meditation on,356 flexibilityof,56
in neurodegeneration,492 functionsof,54
628 Index

Intrinsic plasticity (Cont.) mitochondrial permeability transition in,257


short-term,5455 neurotransmitters and ion channel regulation in,
synaptic plasticityrelated,5556 253254
Inverse synaptic tagging, 178179,179f steps of, 250f, 252253
Inward rectifier Cl- current (IClh), TCA cycle effects and anaplerosis in,259
hyperpolarization-activated,15 Ketogenic diet (KD), 248264
Inward rectifying potassium channels (Kir),78 on adiposity,579
Ion channels. See also specificions on Alzheimers disease, 261262
ion currents and equilibrium potentialin,51 on amyotrophic lateral sclerosis, 263264
ketogenic diets on, 253254 on autism, 264, 594595
opening of, 51,52f on brain cancer,262
Ion currents (I),51 branched chain amino acids and, 282283
Ion homeostasis. See also specificions on creatine,277
astrocyteneuron interactions on,98100 definition of,248
[K+]0, 9899,99f with hyperbaric oxygen therapy, 560561
pH0,99100 on migraine headache,264
in epilepsy,403 mitochondrial dysfunction in neurological diseases
Ionotropic glutamate receptors (iGluRs), in Parkinsons and,261
disease,489 neuroprotective effects of, 263f,264
IP3-sensitive calcium, in astrocytes,76 on neurotrauma, 262263
Isocitrate dehydrogenase (IDH) gene mutations, in on pain and inflammation,264
glioblastoma and gliomas,194 on Parkinsons disease,262
Isoleucine, 278283. See also Branched chain amino radiation therapy with,561
acids (BCAAs) restricted consumption of,559
for sleep,324
Jun N-terminal kinase (JNK),378 variations of, 250252
caloric restriction, 252, 559560,559f
K+ low-glycemic index treatment, 251f,252
astrocyte uptakeof,99 medium-chain triglyceride diet,251
in epilepsy,403 modified Atkins diet, 251252,251f
siphoning of,7,8 Ketogenic diet, calorie restricted (KD-R), 252,
spatial buffering of, 67, 7f,8,147 559560
spatial redistribution of,78 for glioblastoma multiforme, 559f, 560561
uptake of,6,7f Ketogenic diet (KD), on epilepsy, 248261,
[K+]0 410411
astrocyte permeabilityto,98 efficacyof
blood-brain barrier on, 146147 clinical studies of, 249250,250f
elevation of, in glycogen metabolism, astrocyte,103 preclinical studies of,250
homeostasis of, 9899,99f energy-sensing signaling pathway activation in,
astrocyteneuron interactions on, 9899,99f 259261
neuronal activityon,98 AMP-activated kinase in,260
Kainate receptor,489 mammalian target of rapamycin in,260
Kava,526 peroxisome proliferator-activated receptors in,
KCC2 cotransporter 259260
on chloride channels, 5254,53f sirtuins in, 260261
phosphorylationon,59 historical aspects of, 248249
RNA splicingon,57 ketogenesis in, 252259 (See also Ketogenesis)
Keratan sulphate proteoglycan,373 variations of, 250252,251f
Ketogenesis, 252259 Ketone bodies, 253, 559560,559f
acetyl-CoA in, 250f, 252253 from branched chain amino acids, 279,280f
antioxidant activity, reactive oxygen species, and fasting on,302
redox state in, 255257 Kineret,166
bioenergetic and mitochondrial changes in, Kir 2.1 inward rectifying channels, 99,99f
254255,255f Kir4.1 channels,89
bioenergetics reserve and mitochondrial respiration in epilepsy,9,403
in, 258259 on K+ spatial buffering,147
fatty acid oxidation in,258 K siphoning,7,8
+

glycolytic restriction/diversion in, 257258 in Mller cells, 99,99f


ketone bodies in,253 Kudzu, for alcohol addiction, 526527
Index 629

Lactate heterosynaptic, 130131,178


astrocyte to axon transfer of, 102103 hippocampal, in associative learning,124
astrocyte-to-neuron shuttling of,100 homosynaptic, 137f, 138,178
metabolism and, exercise and, 301303 heterosynaptic LTD with,130
Lactate dehydrogenase,103 input specificity of, 176,176f
Lactatepyruvate equilibrium,103 in memory, 177,181
Lactate shuttle hypothesis, astrocyte neuron,103 NMDA-type glutamate receptordependent,177
L-DOPA, for Parkinsons disease,489 types of,177
Learning Loving-kindness meditation, 354355, 360. See also
associative, hippocampal long-term potentiation Meditation
in,124 Low-glycemic index treatment (LGIT), 251f,252
COX- 2 on,165 LRRK2 (PARK8) Parkinsons disease,483
dentate granule neurogenesis in,206 Lymphocytes
development and, 108109 in Parkinsons disease, inflammatory reaction
epigenetics in, 193194 in,494
Leptin T
in addiction,521 in Rasmussens encephalitis,160
discovery of,574 in seizure models,161
functions of,578 Lysergic acid diethylamide (LSD) addiction, 509527.
sleep on,322 See also Addiction
synthesis and role of,520 Lysine acetyltransferases (KAT), 257258
Leptin receptor, in obesity,578
Leptin system, proopiomelanocortin in,576 Major depressive disorder(MDD)
Leucine, 278283. See also Branched chain amino acids anxiety disorders with,545
(BCAAs) with epilepsy, inflammation in, 162163,163f
Leukocytes on hippocampus,213
blood-brain barrier migration across,145 neural stem/progenitor cells and neurogenesis for,
in seizure models,161 213214
Leukoencephalopathies, CIC-2in,15 sleep and, 321322
Levetiracetam. See also Antiepileptic drugs(AEDs) after traumatic brain injury, 427428
binding site of,238 inflammation in, 427428
on epileptiform activity in mice,456 neurotrophic support in,427
on ion channels,236 serotonergic dysfunction in,428
on sleep,314 Malonyl-CoA,578
for traumatic brain injury, 408,432 Mammalian target of rapamycin (mTOR)
Levodopa-induced dyskinesia(LID) blockade of, for epilepsy,410
GABA receptors in,490 ketogenic diets on, 260,263f
glutamatergic receptors in,489 Mania, branched chain amino acids for,280
Lewy bodies, in Parkinsons disease,481 MAPK pathway, 378,382
Limbic-hypothalamic-pituitary-adrenal (LHPA) FGF2 in,380
axis tone of, on behavior,321 guanosine-activated cAMP-dependent mechanisms
in sleep and stress responses, 320321 on,382
Lipostatic hypothesis,571 Marijuana addiction, 509527.
Lithium,5960 See also Addiction
Long-chain fatty acid transport, across mitochondrial endocannabinoid homeostasis in, 513514
membrane, 271,272f mindfulness training on,525
Long-term depression (LTD), 175, 176f, 177178 Maternal immune activation (MIA), in autism, 164,
adenosine on,3637 164f,591
calcium sliding threshold for,136 prevention of,598
in cerebellum, 177178 MC4R receptor
excitatory synaptic scaling on, 110,111f destruction of, in obesity,574
heterosynaptic,178 in obesity, 574, 575,578
homosynaptic,178 -MSH on,574
Long-term potentiation (LTP), 175, 176177,176f MDMA addiction. See also Methamphetamine
adenosine on,3637 serotonin homeostasis in, 512513
associativity of,176 Medial prefrontal cortex (mPFC)
calcium sliding threshold for,136 in anxiety and emotional
cooperative,176 behaviors, 536,537f
excitatory synaptic scaling on, 110,111f in anxiety disorders,537
630 Index

Meditation, 353361. See also specifictypes Metabolic ratio, cerebral, 302303


for addiction, 524525 Metabotropic glutamate receptors (mGluRs), in
on allostasis and allostatic load,353 Parkinsons disease,489
on autonomic regulation of heart, 357358 Metaphysics,234
on blood pressure,355 Metaplasticity, 54, 56, 109, 133, 136,182
on brain homeostasis,353 Methadone, for opioid addiction,523
on brain, research on,353 Methamphetamine
on cortisol, 358359 on AMPA receptors, 514515
on C-reactive protein,356 on histamine release,518
definition of,354 Methamphetamine addiction. See also Addiction
on hippocampus,360 ginseng on,526
on HPA axis regulation, 358359 mindfulness training on,525
on immune function, 355357 serotonin homeostasis in, 512513
on neuroplasticity, 360361 Methioninecycle
on stress response,353 DNA methylation in Alzheimers disease and,458
on telomeres,357 in neuronal homeostasis,193
for traumatic brain injury,433 Methodological reductionism,232
types of practices of,354 1-Methyl-4-phenylpyridinium,263
Medium-chain triglyceride (MCT) diet,251 Methyl-CpG-binding domain (MBD) family,187
for autism,596 Methyl-CpG binding protein 2 (MeCP2), in synaptic
Megalencephalic leukoencephalopathy, astrocyte scaling,117
volume controlin,19 3,4-Methylenedioxymethamphetamine (MDMA). See
Melanocyte concentrating hormone (MCH),577 Methamphetamine
Melatonin, 277278 O6-Methylguanine-DNA methyltransferase (MGMT),
aging and,277 epigenetic alterations in,194
autism and,278 Methylphenidate(MPH)
on free radicals and antioxidants,278 for traumatic brain injury,430
functions of, 277278 use of, by healthy people,238
insomnia and,278 Methylxanthines. See also Caffeine
neurodegenerative disorders and,278 on adenosine receptors, 318,411
sleep deprivation on,323 on epilepsy,411
on sleep-wake transition,318 Microbiome treatment, for autism,597
Membrane-bound monocarboxylate transporters Microbiota, gut,284
(MCT), lactate transport by, 103,302 in autism, 589,597
Memory on body weight,581
adenosine for improvement of, 465467 Microbiotagutbrain axis,284
adenosine kinase on, 466467 Microglia
COX- 2 on,165 in addiction,517
dentate granule neurogenesis in,206 in Alzheimers disease,455
epigenetics in, 193194 cell signaling by,517
epilepsy on, inflammation in, 165166,165f in innate immunity,493
long-term potentiation in, 177,181 regulatory functions of,517
piracetam on,238 in Tau pathology, 455456
synaptic scalingin MicroRNA (MiRNA), 190191,191f
in short- vs. long-term storage,116 Migraine headache
in working memory,116 ketogenic diets on,264
Memory consolidation after traumatic brain injury, 428429
astrocytes in,8283 Mindfulness-based cognitive therapy (MBCT), 354,
histone methylation in,194 359. See also Meditation
Memory formation Mindfulness-based stress reduction (MBSR), 354. See
epigenetic chomatin modifications in,195 also Meditation
glycogen in, 103104 on blood pressure,355
histone acetylation in,193 on cortisol,359
Mesocorticolimbic reward circuitry on heart rate variability,358
in addiction, 509510 on hippocampus,360
in feeding,521 on immune function, 355356
Metabolic disorders. See also specifictypes on neuroplasticity,361
in addiction, 520521 Mindfulness meditation, 354. See also Meditation
in autism, 589590 for addiction, 524525
Index 631

applications of, 524525 on adenosine concentrations, 342,512


components of,524 on endocannabinoids,514
for traumatic brain injury,433 GDNF and,522
Minocycline on glutamate, 514,515
on addiction,517 on mesocorticolimbic reward circuitry,510
for inflammatory diseases,166 on neurosteroids,520
on major depressive disorder,162 Morphine addiction. See also Addiction
for traumatic brain injury,432 acupuncture for,348
Mitochondria cholinergic homeostasis in, 516517
cell damage/injury from, sleep and, 322323,323f ginseng on,526
ketogenic diets on, 254255,255f on progesterone and pregnenolone sulfate,520
respiration in, 258259 Motoneurons,298
neuronal excitability and function of, 254255,255f Motor cortex, 298299
normal function of,555f Movement. See also specific disorders
sleep and, 322323,323f neural control of, 298300,299f
traumatic brain injury on,421 Mller cells,4
Mitochondrial bioenergetics, in epilepsy,406 K+ siphoning in, 99,99f
ketogenic diets on, 254255,255f Multidrug resistance proteins
ketogenic diets on reserve of, 258259 ATP release via,17
Mitochondrial dysfunction at blood-brain barrier,145
in autism, 589590 Multidrug therapies,237
in brain tumors, 554555,555f Multidrug transporters,145
in Huntingtons disease,375 in epilepsy,409
in neurological diseases,261 Multifunctional drugs. See also specifictypes
in Parkinsons disease,487 alcohol as,239
Mitochondrial membrane, long-chain fatty acid trans- anesthetics in,239
port across, 271,272f piracetam in, 238239
Mitochondrial permeability transition (mPT), single-drug molecules as,237
ketogenic diets on,257 Multi-omics,234
Mitogen-activated protein kinase (MAPK) pathway, Multiple sclerosis
378,382 amantadine hydrochloride for,430
FGF2 in,380 blood-brain barrier dysfunction in,149
guanosine-activated cAMP-dependent mechanisms FGF2/FGFR1 signaling manipulation for,380
on,382 fingolimod for,166
Modafinil histamine in,518
for narcolepsy,318 Huperzine Aon,336
use of, by healthy people,238 Multitarget pharmacology, 235237,237f
Modified Atkins diet, 251252,251f Muscarinic (M1-M5) acetylcholine receptors,516
Molecular plasticity, 5760. See also Inhibitory plastic- Myelin-associated glycoprotein,373
ity, neuronal Myelination-related inhibitors,373
Monoacylglycerol lipase (MAGL), in endocannabinoid
addiction,514 N6-(4-hydroxybenzyl) adenosine riboside (T1-11),593
Monoamine neurotransmission, after traumatic brain N-acetylcysteine(NAC)
injury,427 for Alzheimers disease,458
Monoamine oxidase inhibitors (MAOIs) for cocaine addiction,523
for anxiety,542 Na+ channels, voltage-gated, in cell volume,16
for Parkinsons disease,489 Na+-dependent Cl-/ HCO3- exchanger,100
on REMS sleep,317 Na+/ HCO3- cotransport,100
for traumatic brain injury,431 in glial cells,100
Monoamine transporter (5-HTT), in addiction, Na+/ H+ exchanger,100
512513 Na+-K+-2Cl cotransporter (NKCC).
Monocarboxylate transporters (MCT), lactate trans- See Sodium-potassium-chloride
port by, 103,302 cotransporter(NKCC)
Monoclonal antibodies, for epilepsy from Na+/K+-ATPase, exercise on, 301,304f
inflammation Na+/ K+-ATPase pump,6
against IL-1,166 Nalmefene, 522523
against IL-6,166 Naltrexone,522
against TNF-,166 Narcotic addiction, 509527
Morphine Narp, in synaptic scaling,180
632 Index

NCCa-ATP channel, in cell volume homeostasis,16 in subventricular and subgranular zones, 202,203f
Neotrofin (lepteprinim, AIT-082), on nerve growth after traumatic brain injury,425
factor production,381 Neurogenesis homeostasis, for depressive disorders,
Neprilysin gene,457 213214
Nerve growth factor (NGF),373 Neurogenin2,205
AIT-082 on, 381382 Neuroinflammation, in Parkinsons disease, 493494
guanosine on,381 inflammatory response in,492
in neuroprotection,422 postmortem studies of,493
in pain promotion and hyperalgesia,376 therapy for, 494495
in restoration,375 Neuromyelitis optica (NMO), AQP4 OAPsin,10
Netrins,373 Neuronal activity-regulated pentraxin (Narp), in syn-
Network pharmacology, 231243. See also Systems aptic scaling,180
(network) pharmacology Neuronal excitability homeostasis, 5163. See also
Neural/glial antigen,373 Excitability homeostasis, neuronal
Neural restrictive silencing factor (NRSF), 257,410 Neuronal hyperactivity, Ca 2+ in,195
Neural stem/progenitor cells (NSCs), 202218 Neuron restrictive silencing 23 element (NRSE),257
activity of, determining,205 Neurons
in Alzheimers disease, 214215 functionsof,98
in brain repair after stroke, 215217 silencing of, 56, 113, 125, 129, 130,135f
definition of,202 Neuropeptide Y(NPY)
in depressive disorders, 213214 in anxiety and stress disorders,540
functional significance of, 206208 on body weight,577
in hippocampus, 203f, 204205 in obesity, 576577
adult human,208 Neuroplasticity, 175182. See also specifictypes
regulators of, 205206 cortical reorganization and remapping in,
stem vs. progenitor cells in,202 180181,181f
in subventricular and subgranular zones,202 definition of,175
in temporal lobe epilepsy, 208213 functions of,175
early, proliferation and connectivity in, homeostatic response recovery in,181
209211,210f homeostatic synaptic scaling in, 109, 128130, 175,
homeostasis and neurogenesis in, 208209 181 (See also Synaptic scaling)
NSC potential and new granule cells in, 211213, meditation on, 360361
212f213f neurological/psychiatric disorders and,181
Neurocan,373 ocular dominance plasticity in, 180181,181f
Neurodegeneration. See also specific disorders synaptic plasticity in, 175180 (See also
inflammation in,492 Synaptic plasticity)
melatonin and,278 in traumatic brain injury,425
in traumatic brain injury, 425426 Neuropoetic cytokines,521
Neurofibrillary tangles, in Alzheimers disease, 214, Neuroregeneration, 372. See also Neurorestoration
452453 Neurorestoration, 371386, 374377
Neurogenesis bench to bedside gap in,386
definition of,192 definition of,371
epigenetics in, 192193 fibroblast growth factor in, 379380
exercise on, 308,411 fibroblast growth factor signaling pathway in,
from nerve growth factor,376 380381
pathological,411 goal of, 371372
in subventricular and subgranular zones,202 guanosine signaling transduction cascade of, NGF,
Neurogenesis, adult, 202218. See also BDNF, and FGF2 pathways and, 382383,383f
Stem cells, adultneural targeting convergent molecules in, 384385
brain areas of, 202204,203f history of,371
FGF2 in, 379380 homeostasis regulatory mechanisms and, 371372
in hippocampus, 202204,203f neuroregeneration in,372
human,208 neurotrophin (NGF/BDNF) signaling pathway
regulators of, 205206 in,377
neural stem/progenitor cells in, 202 (See also nonadenine-based purinesin
Stem cells, adult neural) AIT-082, 381382
functional significance of, 206208 guanosine,381
in nonneurogenic brain regions, 202203 nonpermissive environment in, reducing,
stem vs. progenitor cells in,202 372373,384
Index 633

p75NTR signaling pathway in, 378379 guanosine,381


permissive environment cues in, stimulating,373 neotrofin,381
permissive environment stimulation in, 374377 Noncoding RNAs, 190191,191f
(See also specifictypes) Nootropic drugs, 231, 237239,240f
BDNF in, 376377 Norepinephrine (noradrenaline)
hormones and neurotransmitters in,374 exercise and,305
interleukins and lymphokines in,374 in fear learning,540
nerve growth factor in, 375376 in pain,490
purinergic molecules in,374 in Parkinsons disease, 490, 491492
research on, early,372 on sleep,321
scope of,372 Norepinephrine-enhancing agents, on sleep, 317318
strategies for,374 Norepinephrine-inhibiting agents, on sleep,317
TRK signaling pathway in, 377378 Norepinephrine neurotransmission
Neurosteroids,520 in anxiety and stress disorders,539
Neurotransmitters. See also specifictypes in Parkinsons disease,491
in addiction, 514517 in stress response,537
astrocyteneuron interactions in homeostasis of, Normalization,127
100101,101f Notch signaling,205
on hippocampal neurogenesis, 205206 NREMS sleep, 315f,316
ketogenic diets on, 253254 Nuclear factor-kappa B (NF-B) cascade, p75NTR
metabolism of, local synaptic,45 activation of,379
packagingof,44 Nucleosome, 189,189f
synaptic cleft removalof,44 Nucleus accumbens(NAc)
vesicular contentof,44 in addiction, 509510
Neurotrophins (neurotrophic factors), 373, 374. See dopamine in,511
also specifictypes Nutrition. See also Diet
in addiction, 521522 on brain function,248
exercise benefits for,524 prenatal, on CNS development and function,193
in neuroprotection, 422423 for sleep,324
in Parkinsons disease, 494495
Neurotrophin (NGF/BDNF) signaling pathway,377 O2 consumption, in exercise, 300301
Neurovascular coupling,148 O6-methylguanine-DNA methyltransferase (MGMT),
Neurovascular unit, 3, 143144, 144f,422 epigenetic alterations in,194
NF E2-related factor 2 (Nrf2),256 obese (ob) gene, 573575
NF-kB (nuclear factor kappa-light-chain-enhancer of Obesity and diabetes, 570581
activated B cells), in addiction,517 calories myth in,580
Nicotine addiction, 509527. See also Addiction dietary composition on,579
acupuncture on, 525526 energy balance and glucose homeostasis in, ventro-
nicotine replacement for,523 medial hypothalamus in, 571573
varenicline for,523 epidemiology of, 570571
Nicotine, on endocannabinoids,514 fat mass and obesity associated (FTO) gene, 579580
Nicotine replacement, for nicotine addiction,523 genetic, in humans, 579580
Nicotinic acetylcholine (nAch) receptors genetic, in mice, 573578,574
in addiction, 516517 Agouti-related peptide in, 576577
classification of,516 arcuate hypothalamus in, 575, 576577
NMDAR antibodies, in epilepsy, 160161 diabetes (db) gene in, 573575
NMDA receptor,489 early studies on,573
astrocytic control of trafficking of, in epilepsy, leptin in,574
8790, 88f89f melanocyte concentrating hormone in,577
estrogen on,412 neuropeptide Y in, 576577
NMDA-type glutamate receptor obese (ob) gene in, 573575
(NMDA-R)-dependent LTP,177 proopiomelanocortin in, 575577
N-methyl-D-aspartate (NMDA), on adenosine ventromedial hypothalamus in, 573576
concentrations,342 -melanin-stimulating hormone in,574
Nociception homeostasis, acupuncture in, 341349. See ghrelin in, 578579
also Acupuncture glucostat hypothesis in, 572,578
NogoA,373 health consequences of,571
Nonadenine-based purines, on nerve growth factor hypothalamic beta-oxidation,579
production hypothalamic nutritional signals in, 578579
634 Index

Obesity and diabetes (Cont.) acupuncture for, 343, 343f (See also Acupuncture)
insulin in,578 definition of,348
lipostatic hypothesis of,571 prevalence of,348
microbiota on, gut,581 Pannexin 1 (panx1)
nature nor nurture in, 580581 functionof,12
Obsessive compulsive disorder (OCD). See also in potassium homeostasis,12
Anxiety and stress disorders Parasympathetic nervous system
deep brain stimulation for,543 in anxiety and stress disorders, 535,537f
DSM on,538 meditation on, 353, 355, 357,358
pharmacological treatment of, 541542 in stress response, 535,537f
in youth,544 PARK1/PARK4 Parkinsons disease, 482483
Ocular dominance plasticity, 180181,181f PARK4 Parkinsons disease, 482483
Olanzapine PARK6 Parkinsons disease,484
on sleep,317 PARK7 Parkinsons disease,484
targets for,236 PARK8 Parkinsons disease,483
Oligodendrocyte myelin glycoprotein,373 PARK15 Parkinsons disease,485
Omega-3 fatty acids, 284285 PARK72 Parkinsons disease,484
for traumatic brain injury,435 PARK genes,482
Omics technologies, 234,242 Parkin (PRKN/PARK72) Parkinsons disease,484
for traumatic brain injury,429 Parkinsons disease, 481497
Ontological reductionism,232 adenosine signaling in, 35,3738
Opioid addiction. See also Addiction amantadine hydrochloride for,430
acupuncture for,348 autonomic nervous system dysfunction in,491
buprenorphine for,523 basal ganglia dysfunction in,299
exercise on,524 basal ganglia in,488
ginseng on,526 BDNF in,376
methadone for,523 brain homeostatic systems in, 487492
Opioids. See also specifictypes adenosine-mediated,491
histaminergic drugs on responses to,518 adrenergic,490
on mesocorticolimbic reward circuitry,510 basal ganglia in,488
Orexin,317 cholinergic, 490491
Orexin receptor blockers, on sleep,317 dopaminergic, 488489
Orkand model, 67,7f glutamatergic, 489490
Orphan CpG islands,188 motor and non-motor symptoms in, 487488
Orthogonal arrays of particles (OAPs),10 norepinephrine-mediated,491
Ovarian hormones, in addiction, 518520 serotonergic,491
Oxidative stress cells in, 493494
ketogenic diets on, 255257 clinical symptoms of,482
in Parkinsons disease,487 motor, 487488
from traumatic brain injury,422 non-motor, 487488
OxPhos creatine for, 274275,278
in glioblastoma multiforme mitochondria, depression in, 482, 487,488
554555,555f GABAergic neurotransmission in,490
insufficiency of, in Warburg effect,556 glutamatergic neurotransmission in,489
noradrengic neurotransmission in,490
P2X receptors, acupuncture and,347 serotonergic neurotransmission in,491
P2Y1 receptor,76 dopamine in, 488489
P2Y receptor, acupuncture and,347 endogenous toxins in, 485486
p75NTR energy demands in,272t
-amyloid protein and,379 environmental factors in, 485, 486487
nuclear factor-kappa B cascade activation by,379 etiology of,482
p75NTR signaling pathway, 378379,385 future directions in, 495497
Pain genetic forms of, 482,495
CNS, BDNF in,376 genetic forms of, autosomal dominant, 482484
ketogenic diets on,264 EIF4G1,484
in Parkinsons disease, 482, 489492 (See also GBA,484
Parkinsons disease) LRRK2 (PARK8),483
Pain, chronic SNCA (PARK1/PARK4), 482483
A1 receptor agonists on, 343,343f VPS35, 483484
Index 635

genetic forms of, autosomal recessive, 484485 pH


ATP13A2 (PARK9), 484485 blood-brain barrier on,147
DJ-1 (PARK7),484 definitionof,99
FBXO7 (PARK15),485 pH0
Parkin (PRKN/PARK72),484 activity-induced changes in, and regulation,100
PINK1 (PARK6),484 astrocyte regulation of,99100
PLA2G6,485 homeostasis of,99100
homeostatically controlled mechanismsin,35 Phenytoin
ketogenic diets on,262 P-glycoprotein on brain penetration of,409
Lewy bodies in,481 on sleep,314
neuroinflammationin targets and mechanisms of action of,407
inflammatory response in,492 for traumatic brain injury,432
postmortem studies of,493 Phosphatidylinositol 3-kinase (PI3K) pathway,378
therapy for,494 Phospholipase C gamma (PLC-gamma) pathway,378
neuroinflammatory pathways in, 493494 FGF2/FGFR1 triggering of, 380381
neurotrophic factors in, 373, 494495 Phosphorylation, in inhibitory plasticity,5960
pain in,482 Physical exercise, 298308. See also Exercise
adenosine pathways in,492 Phytocannabinoids,335
GABAergic neurotransmission in,490 PI3K/Akt signaling pathway, tumor radiation on,557
glutamatergic neurotransmission in,489 Pia, glial membrane of,104
noradrenaline in,490 PINK1 (PARK6) Parkinsons disease,484
serotonergic pathways in,491 Piracetam, 231,238
pathogenesis of, 384, 486487 on cell membranes, 238239
mitochondrial dysfunction in,487 lack of specificity of, 238239
oxidative stress in,487 on memory, 231,238
protein aggregation and misfolding in,486 PLA2G6 Parkinsons disease,485
ubiquitination and related stress in, 486487 Plasticity, 308. See also Neuroplasticity; specifictypes
pathophysiology and neurochemical Hebbian, 108, 124126, 136137
features of,481 intrinsic, 5456, 182 (See also Intrinsic plasticity)
prevalence of,481 epilepsy-related, 6061,62f
reactive astrocytosisin,47 molecular (inhibitory), 5760 (See also
risk factors for,481 Inhibitory plasticity, neuronal)
sleep disorders in, 482, 487, 489491 structural,182
adenosine in,492 PLC-gamma. See Phospholipase C gamma
autonomic nervous system in,491 (PLC-gamma) pathway
GABA in,490 Polycomb (PcG) system,192
glutamatergic receptors in,489 Polyglutamine diseases,384
serotonin in,491 Polypharmacology, 235237,237f
therapeutic strategies for,495 Polyphenols, dietary, for traumatic brain injury,436
Paroxetine. See also Polytherapy, rational,237
Selective serotonin reuptake inhibitors (SSRIs) Polyunsaturated fatty acids (PUFA), ketogenic diets
for anxiety disorders,541 on,258
on depression,241 Posttraumatic epilepsy,426
Pattern separation,207 Posttraumatic headache,428
Pedunculopontine nucleus (PPN), in Parkinsons Post-traumatic stress disorder (PTSD)
disease,490 amygdala in,537
Pentobarbital, for traumatic brain injury,432 cortisol in,540
Pericytes, 144,144f deep transcranial magnetic stimulation for,543
Peripheral fatigue,305 development of,538
Peroxiredoxin4,262 gender differences in,543
Peroxisome proliferator-activated receptors sleep in,321
(PPARs),579 THC for,541
ketogenic diets on, 259260,263f Potassium channels
Persistent hypogonadotropic hypogonadism (PHH), in astrocyte-mediated potassium homeostasis,79
after traumatic brain injury,429 in astrocyte volume regulation,14
PGE2, in inflammatory cascade, 157t,159 inward rectifying (Kir),78
P-glycoprotein, in epilepsy,409 phosphorylationof,60
P-glycoprotein-like proteins, at blood-brain Potassium-chloride cotransporter (KCC), on chloride
barrier,145 channels, 5254,53f
636 Index

Potassium homeostasis microdynamics, in autism,593


extracellular,612 on neural stem cells, 380381
astrocytesin Purines. See also
aquaporins in,1011 Adenosine; ATP (adenosine triphosphate)
connexinsin,11 homeostatic regulationof,31
pannexinsin,12 salvage of,3132
potassium channels in,79 Pyramidal neurons, synaptic scalingin
astroglial,11 excitatory, 109112,111f
Kir4.1 in, 89,403 inhibitory, 112113
K+ siphoning in,7,8
K+ uptake and spatial K+ buffering in, 67,7f,8 Quetiapine, targets for,236
Orkand model of, 67,7f
potassium and sodium ion concentrationsin,6 Radiation therapy
TREK and TWIK in,9,14 on brain tumor energy metabolism, 557558,557f
Potassium ion ( K+). See also K+ with ketogenic diet,561
astrocyte uptakeof,99 on PI3K/Akt signaling pathway,557
in epilepsy, elevation of,403 Ramelteon, on sleep-wake transition,318
siphoning of,7,8 Rapamycin, in epilepsy,410
spatial buffering of, 67, 7f,8,147 Ras/extracellular signalregulated kinase (ERK),
spatial redistribution of,78 377378,384
uptake of,6,7f Rasmussens encephalitis (RE), 156, 159160
Prazosin, on sleep,321 Rational drug discovery,235
Prefrontal cortex(PFC) Rational polytherapy,237
in addiction, 509510 Reactive astrocytosis,47
in anxiety disorders,537 in Alzheimers disease, 47,454
in emotional processing, 536,537f in epilepsy, 47,8485
meditation on,360 in Parkinsons disease, 493494
Pregnenolone sulfate in stroke,47
in addiction,520 from traumatic brain injury, 47, 421422
morphine addiction on,520 Reactive astrogliosis. See Reactive astrocytosis
Presenilin-1 (PSEN1) promoter, 457458 Reactive oxygen species(ROS)
PRKN/PARK72 Parkinsons disease,484 ketogenic diets on, 255257
Probiotics,284 n tumor cells,553
Progenitor cells,202 in Parkinsons disease,487
Progesterone from traumatic brain injury,422
on GABAergic and glutamatergic Receptor for advance glycation end products (RAGE),
neurotransmission,412 in epileptogenesis, 157t, 159, 165,166
morphine addiction on,520 Receptor-mediated transcytosis,147
for traumatic brain injury, 423,435 Redox state, ketogenic diets on, 255257
Programmed cell death,494 Reductionism, 231, 232233
Proopiomelanocortin (POMC) epistemological,232
in leptin system,576 methodological,232
in obesity, 575577,579 ontological,232
Proportional-integral-derivative controller,115 Reelin,209
Prostaglandin 2 (PGD2), sleep and, 324,324f Regulatory volume decrease (RVD), 12,13f
Protein aggregation and misfolding, in Parkinsons VRAC in,13f
disease,486 Rehabilomics,429
Pseudoginsenoside-F11 (PF11),526 Relaxation Response, 354. See also Meditation
Psychostimulants. See also specifictypes on blood pressure,355
addiction to, 509527 (See also Addiction) on immune function, 356357
GDNF and,522 REMS sleep, 315f, 316317
on sleep, 317318 Repressor element 1-silencing transcription factor,
for traumatic brain injury,430 205,209
use of, by healthy people,238 Reservatrol,436
Psychotherapy, for anxiety and stress disorders, Retaliatory metabolite, 31, 38,593
542543 Rett syndrome, 117, 181,596
Purinergic signaling,373 RhoA,379
Purinergic system Risperidone, on sleep,317
acupuncture on, 346347 RNA editing, in inhibitory plasticity,5859
Index 637

RNAs, noncoding, 190191,191f in MDMA addiction, 512513


RNA splicing, in inhibitory plasticity,5758 Serotonin-inhibiting agents, on sleep,317
Rubinstein-Taybi syndrome,193 Serotonin neurotransmission system
Runaway dynamics in anxiety and stress disorders,539
heterosynaptic plasticity in prevention of, 133134, in exercise,305
135f,136f Serotonin-norepinephrine reuptake inhibitors (SNRIs)
mechanisms counteracting, other, 136137 for anxiety disorders, 540541
normalization on,127 on REMS sleep,317
positive feedback on synaptic weight changes on,125 Serotonin receptors, in addiction, 512513
spike-timing dependent plasticity rules and, Sertraline
134,135f for anxiety in children and adolescents,544
synaptic scaling in, 129130 for traumatic brain injury,431
SeSAME syndrome,8
S100b,424 Set points,372
S-adenosylhomocysteine hydrolase (SAH), 33, Sex steroids, in addiction, 518520
406407,407f Short-term intrinsic plasticity,5455
in DNA methylation, 458,465 Silencing
S-adenosyl methionine (SAM),458 epigenetic, of tumor suppressor genes,194
in DNA methylation, 188, 189f, 193,465 gene,138
Schizophrenia DNA methylation in, 190,195
adenosine signaling in, 35,3738 histone methylation in,194
antipsychotics for, targets for,236 RNA-induced, 191,191f
energy demands in,272t neuron, 56, 113, 125, 129, 130,135f
GSK3-signalingin,60 transcriptional, 194,457
histamine in,518 Siltuximab,166
homeostatically controlled mechanismsin,35 Similarity ensemble approach,241
Secondary injury, in traumatic brain injury,420 in silico receptorome screening in,241
Seizures. See also Epilepsy Sirtuins (SIR1-7), ketogenic diets on, 260261,263f
in Alzheimers disease,465 SLC6A4 variation, after traumatic brain injury,424
from astrogliosis,465 Sleep, 314325
inflammation in pathogenesis of, 156158,157t adenosine on, 36, 81, 305,307
Selective serotonin reuptake inhibitors (SSRIs). See also on adenosine signaling,411
specificagents astrocytes in,8184
for anxiety disorders, 540541 on brain activity at circuit level,81
in children and adolescents,544 in inflammatory response and sleep behavior,84
on cocaine and alcohol addiction behavior,513 on sleep homeostasis, 77f, 8182,83f
on REMS sleep,317 sleep loss, depression, and,8384
for traumatic brain injury,431 sleep loss, memory consolidation, and,8283
Semaphorins,373 on ATP, brain,322
D-Serine, 77f, 7879,404 circadian pacemaker in,317
Serotonergic neurotransmission on cognition, 319320
in depression after traumatic brain injury,428 on damage and waste removal, 323324
in Parkinsons disease,491 on damage, pathology, and neurological disorders,
Serotonin (5-hydroxytryptamine,5-HT) 322323,323f
in addiction, 512513 dopamine-enhancing agents on, 317318
on adenosine concentrations,342 dopamine-inhibiting agents on,317
biosynthesis of,512 exercise on,308
exercise on,305 on ghrelin, leptin, and body weight,322
functions of,512 glutamate in,305
on neurogenesis, 205206 immune factors on, 324,324f
in Parkinsons disease,491 on immune responses, 324,324f
receptors on action of,512 mitochondria and, 322323,323f
on sleep, 305,321 neuro-immune response factors and, 324,324f
Serotonin-altering drugs, addictive, 512513 norepinephrine-enhancing agents on, 317318
Serotonin-enhancing agents norepinephrine-inhibiting agents on,317
on REMS sleep, 315f,317 nutritional pharmacology for,324
on sleep, 317318 in Parkinsons disease, 482,487
Serotonin homeostasis adenosine in,492
in addiction, 512513 autonomic nervous system in,491
638 Index

Sleep (Cont.) Spatial K+ buffering, 67, 7f,8,147


GABA in,490 Sphingomyelinase,379
glutamatergic receptors in,489 Spikes, bursts of, in heterosynaptic plasticity, 131,
serotonin in,491 132,132f
serotonin-enhancing agents on, 317318 Spike-timing dependent plasticity (STDP),125
serotonin in,305 Spike-timing dependent plasticity (STDP)rules
serotonin-inhibiting agents on,317 competition between synapses in,126
sleep-wake transition in,318 in Hebbian plasticity,126
on stress and behavior, 320321,320f heterogeneity in parameters of,126
sufficient,314 heterosynaptic plasticityon
synaptic scaling in, 116117 competition in, 134135
Sleep architecture, neurobiology and, 315318,315f runaway dynamics prevention and, 134,135f
definition of,315 in stable neuron operation, 134,136f
NREMS, 315f,316 learning,125
NREMS-REMS cycles in, 315316,315f neuron stabilization from,126
REMS, 315f, 316317 regular pairing protocols on,133
wakefulness, 317318 timescale in,128
Sleep disorders Spinocerebellar ataxia type 1 (SCA1),384
with autism,589 Spiny projection neurons (SPNs)
with major depressive disorder, 321322 dopaminergic,488
melatonin and,278 GABAergic, 489490
in Parkinsons disease, 482, 487, 489491 Spontaneous recurrent seizures (SRS), in temporal lobe
in post-traumatic stress disorder,321 epilepsy,209
Sleep hygiene, 324325 Stability, change and,108
Sleep, insufficient Stem cell related transplant,374
adenosine signaling in,3738 Stem cells, adult neural, 202208. See also Neural stem/
astrocytes and,8283 progenitor cells(NSCs)
astrocytes, depression, and,8384 Stimulant addiction, 509527. See also Addiction
on behavior,321 Stimulants. See also specifictypes
on brain,314 GDNF and,522
on glucocorticoids, 320321 on sleep, 317318
on glutathione,323 for traumatic brain injury,430
homeostatically controlled mechanismsin,35 use of, by healthy people,238
on melatonin,323 STITCH (Search Tool for Interactions of
on stress response, 320321 Chemicals),241
Sleep pressure St. Johns wort,526
gliotransmission in accumulation of, 8283,83f Strategy training, for traumatic brain injury, 433434
inflammationon,84 Stress-activated protein kinase signaling cascade,378
Sleep therapy,322 Stress, prenatal, on neurodevelopmental and psychiat-
for cognition,219 ric disorders,193
Sleep-wake transition,318 Stress response
Slits,373 meditation on,353
Slow inward currents (SICs) sleep on, 320,320f
from astrocyte glutamate release,78 sleep on, insufficient, 320321,320f
in epilepsy,86 Striatum,489
Slow outward currents (SOCs), from astrocyte GABA Stroke,118
release,79 on ATP production,101
Small guanosine triphosphatases,379 disability from,214
SNCA (PARK1/PARK4) Parkinsons disease, 482483 epidemiology of,214
Social anxiety disorder (SAD), amygdala in,537 GSK3-signalingon,60
Sodium, intracellular ([Na+]i), in astrocyte homeostasis nerve growth factor on,376
regulation,18 neural stem/progenitor cells for brain repair after,
Sodium-potassium-chloride cotransporter (NKCC),6 215217
on chloride channels, 52,53f reactive astrocytosisin,47
phosphorylationon,59 synaptic scaling in,118
Soluble N-ethylmaleimide-sensitive fusion protein Structural plasticity,182
receptor (SNARE) complex, in astrocytes, Subarachnoid hemorrhage, blood-brain barrier dys-
77,77f function in,149
Solute carriers (SLC),146 Substance abuse. See also specific substances
Index 639

addiction in, 509527 (See also Addiction) weight dependence of changes in,126
anxiety disorders with,545 Synaptic plasticityrelated intrinsic plasticity,5556
classification of,509 Synaptic scaling, 54, 108118, 109, 128130, 175,
Substance P, on adenosine concentrations,342 179180,179f
Substantia nigra,299 in Alzheimers disease,118
acetoacetate and -hydroxybutyrate on,254 canonical, 110,111f
BDNF in,376 as cell autonomous, 111112
dopamine from,511 computational ideas on, 115117
dopamine neurons in, 262, 317,536 in control theory, 113115,114f
neurogenesis in, 202203 definition of, 109, 128129
in Parkinsons disease, 262, 381, 481, 484, 490,492 in development and learning, 108109
on sleep architecture,315f excitatory, 109112
in traumatic brain injury,427 in interneurons,112
Succinyl-CoA,596 in pyramidal neurons, 109112,111f
Sudden unexpected death in epilepsy (SUDEP), caf- experimental studies on, 128129
feine in,411 as feedback control mechanism, 113114
Suprachiasmatic nucleus,317 firing rate homeostasis in, 115116
Suramin,593 fragile X mental retardation protein in,117
Susceptibility genes, DNA methylation in Alzheimers in fragile X syndrome,117
disease and, 457458 genes in,180
SWItch/ Sucrose Nonfermentable (SWI/SNF) inhibitory, 112113
complex,192 local and rapid forms of,110
Symbiosis,284 MeCP2 in,117
Sympathetic nervous system in memory
in anxiety and stress disorders, 535,537f short- vs. long-term storage of,116
ketone body effects in,254 working,116
meditation on, 353, 355, 357,358 Narp in,180
norepinephrine in, 305, 320,321 in neocortex,129
sleep and,320 neurological/psychiatric disorders and,181
in stress response, 320, 535,537f realism of,130
Synapse in Rett syndrome,117
development on,108 scaling up and down in,112
Hebbs rule on,108 in sleep, 116117
neurotransmitter metabolism in, local,45 in stroke,118
Synaptic competition study methods for,109
Hebbian plasticity and, 126,127 synaptic competition in,116
in synaptic scaling,116 timescale, competition, and runaway dynamics in,
Synaptic drive,108 129130
Synaptic plasticity, 124, 175180 timescale in,116
adenosine in,3637 Synaptic tagging, 178,179f
associative,124 inverse, 178179,179f
definition of,175 Synaptic tagging and capture theory,178
epigenetics, learning, and memory in, 193194 System N glutamine transporter(SN1)
epilepsy-related, 6163,62f in glutamine release,46
inverse synaptic tagging in, 178179,179f localizationof,45
long-term depression in, 175, 176f, 177178 (See also Systems biology,232
Long-term depression(LTD)) Systems (network) pharmacology, 231243,333
heterosynaptic,178 ab initio drugs in, 231,237
homosynaptic,178 brain functionality and complexity in, 231232
long-term potentiation in, 175, 176177, 176f (See for brain functionality restoration, 231243
also Long-term potentiation(LTP)) chemoinformatics tools for, 240241
heterosynaptic,178 disease-associated molecular networks in, 240241
homosynaptic,178 on drug action,235
neurological/psychiatric disorders and,181 emergent properties in,233
saturation of,136 epigenetics and geneenvironment interactions in,
synaptic scaling in, 179180, 179f (See also Synaptic 233234,234f
scaling) fundamental tenets of,231
synaptic tagging in, 178,179f integrative neuroscience approaches in,
weight dependence of,136 232235,234f
640 Index

Systems (network) pharmacology (Cont.) spontaneous recurrent seizures in,209


multitargets in, 235237,237f Testosterone
network dynamics discovery in,241 in addiction, 518519,520
nootropic drugs in, 231, 237239,240f on traumatic brain injury, 423,429
outlook for, 242243 Tetanization, heterosynaptic plasticity induction via,
practical matters of, for CNS drugs, 240242 132133,132f
PubMed papers on, 235,235f Tetrahydrocannabinol (THC), 513514
purpose of,232 for posttraumatic stress disorder,541
reductionism and, 232233 Theophylline
similarity ensemble approach in,241 on adenosine receptors, 318,411
systems biology in,232 from caffeine metabolism,461
on epilepsy,411
T1-11,593 Theravadan Buddhist meditation, 360. See also
Tai Chi Chuan Qigong, for traumatic brain injury,433 Meditation
Tamoxifen 3-Hydroxy-3-methylglutaryl CoA (HMG-CoA),
on focal ischemia,15,18 250f,253
on VRAC channels,15,18 Thrombin, on brain, 148,150
Tardive dyskinesia, branched chain amino acids for, Tibetan Dzogchen meditation, 360. See also
280281 Meditation
Target-selective pharmacology,231 Tight junctions,144
drug discovery in, 234235 TLR4 signaling, in epilepsy, 157158,157t
Tauopathies. See also specific disorders T lymphocytes
Alzheimers disease as, 455 (See also in Rasmussens encephalitis,160
Alzheimers disease) in seizure models,161
astrocytes in,455 Traditional Chinese medicine(TCM)
chronic traumatic encephalopathy as,426 acupuncture in, 341349 (See also Acupuncture)
epileptic activity and seizure thresholds in,465 botanicals in, 334 (See also Botanicals)
microglia in, 455456 combination therapy in,334
tau acetylation on,459 on health,334
Tau protein, in Alzheimers disease,454 Transcendental Meditation. See also Meditation
Tbr2,205 on blood pressure,355
Tele-methylhistamine, in addiction,518 research on, early,354
Telomeres, meditation on,357 Transcranial magnetic stimulation, for PTSD,543
Temozolomide, for gliobastoma multiforme, 553554 Transcriptional silencing, 194,457
Temporal lobe epilepsy (TLE). See also Epilepsy Transcytosis
astrogliosisin,47 adsorptive-mediated,147
clinical picture of,6263 receptor-mediated,147
COX-2 in,165 Transforming growth factor (TGF-B) signaling,
dentate gyrus granule cellsin,60 blood-brain barrier disruption on,405
etiology of,209 Transient receptor potential (TRP) channel genes, in
gephyrinin,61 cell volume homeostasis,1617
gephyrin splice variantsin,58 Transient receptor potential (TRP) channel, in
gliotransmission in, 8690, 88f89f (See also osmotransduction,13f
Gliotransmission, in epilepsy) Traumatic brain injury (TBI), 420429
glutamate-glutamine cyclein,47 autoimmunity in, 424425
glutamine synthetase in,403 brain plasticity in,425
GlyR RNA splice variantsin,57 branched chain amino acids for,280
GlyR 2- and 3-coding gene transcriptsin,58 cognitive dysfunction in,427
granule cell upregulations in,6061 creatine and, 275276
hippocampal sclerosis in, 60, 62f, 85, 87, 209,403 depression in, 427428
IL-1R/TLR signaling pathway in, 405406 DNA hypomethylation in,409
Kir4.1 in,9,403 epilepsy in,426
neural stem/progenitor cells in, 208213 etiology of,422
homeostasis and neurogenesis in, 208209 genetics and epigenetics of,424
NSC potential and new granule cells in, 211213, headache and migraine in, 428429
212f213f history of,371
proliferation and connectivity in, 209211,210f innate homeostatic heterogeneity in, 423426
pathophysiology of,209 ketogenic diets on, 262263
reactive astrocytosis in,8485 neurodegeneration and atrophy in, 425426
Index 641

neuroendocrine dysfunction in,429 standard of care on,557


neuroprotectants for, potential, 422423 Tumor necrosis factor (TNF) receptor family,378
neurotrophic factors in,373 p75NTR signaling pathway in, 378379
pathophysiology of, 420423 Tumor necrosis factor (TNF-)
blood-brain barrier and reactive astrocytosis in, in addiction, 517518
47, 421422 in Alzheimers disease,455
contusions and axonal stretch/strain in,420 in inflammatory cascade, 157t,159
excitotoxicity in, 420421 sleep and, 324,324f
inflammation in,421 TWIK,9
integrative summary on,423 2-arachidonoylglycerol (2-AG), 513514
ischemia and oxidative stress in,422
stress response from,423 Ubiquitination, in Parkinsons disease, 486487
Traumatic brain injury (TBI) intervention strategies, Ubiquitin-proteasome system (UPS),486
429436 Uncoupling proteins (UCPs),256
biologicsin
omega-3 fatty acids,435 Valine, 278283. See also Branched chain amino acids
polyphenols, dietary,436 (BCAAs)
progesterone/estradiol,435 Valproate. See also Antiepileptic drugs(AEDs)
vitaminD,435 carnitine and, 273,274
common considerations in, 429430 multimechanistic actions of, 236, 237f,242
omics and rehabilomics in, 429430 targets and mechanisms of action of,407
pharmacotherapies in, 430432 Vanillinoid receptor (TRPV1) activation, on adenosine
anticonvulsants,432 release,342
antidepressants,431 Varenicline, for nicotine addiction,523
anti-inflammatory strategies,432 Vasogenic edema,12,14
cholinergics,431 Ventral tegmental area(VTA)
stimulants,430 in addiction, 509510
physiotherapies in, 432435 dopamine in,511
acupuncture, 434435 pregnenolone sulfate on,520
cognitive rehabilitation and strategy training, Ventromedial hypothalamus(VHM)
433434 in energy balance and glucose homeostasis, 571573
exercise, 432433 on food intake and metabolic rate, 571572
mindfulness meditation,433 in obesity, 573576
TREK (TWIK-related K+ channels),9,14 VIAAT,44
Tricarboxylic acid (TCA)cycle Vigabatrin, for addiction,512
GABA from substrates of, presynaptic,45 Vimentin,204
ketogenic diets on,259 in epilepsy, 47, 85, 87, 88f89f
acetyl-CoA and glutamate in, 252253 Vipassana meditation, 360. See also Meditation
mitochondrial redox changes in, 255,255f VitaminB
via glycolytic restriction/diversion,257 deficiency of, hypomethylation from,458
Tricyclic antidepressants (TCAs) on DNA methylation,458
for anxiety,542 VitaminB12
for traumatic brain injury,431 deficiency of, in Alzheimers disease,458
Triheptanoin functions of,458
for autism,596 Vitamin D, for traumatic brain injury,435
for epilepsy, 259,411 Vitamin D hormone (VDH), for traumatic brain
Trithorax (trxG) system,192 injury,435
TrkB signaling Volume-activated anion channels (VAAC), astrocyte
in epilepsy, 59,405 release of ATP via,77f
glycolytic restriction/diversion on,257 Volume-regulated anion channel (VRAC)
Trk receptors,377 in Cl- conductance,15
Trk signaling pathway, 377378 on regulatory volume decrease, 13f,15
TRPCs,17 tamoxifen on,15,18
Tryptophan VPS35 Parkinsons disease, 483484
in central fatigue,306 VX-765, for epilepsy from inflammation,166
exercise and,306
Tumor-associated macrophages/monocytes (TAM), in Wakefulness, 317318
gliobastoma multiforme,553 in Parkinsons disease, 482, 487, 489, 490, 491,492
glutamate use by,558 Warburg effect, 262, 555556,555f
642 Index

Waste removal, sleep on, 323324 BDNF on,577


Water and ion microdynamics, 319. See also excess ( See Obesity and diabetes)
specificions gut microbiota on,581
astrocytes cells in, 36,5f regulation of, 570571
calcium and sodium dynamics in,1718 sleep on,322
cell volume homeostasis in, 1217 (See also Weight dependence, of plastic changes,126
Cell volume homeostasis microdynamics)
potassium homeostasis in, 612 (See also Yellow mouse,573
Potassium homeostasis microdynamics)
Water channels, in astrocyte volume regulation,1314 Zen meditation, 360. See also Meditation
Water homeostasis, in epilepsy,403 Zonisamide, 314, 412. See also Antiepileptic drugs
Weight,body (AEDs)

S-ar putea să vă placă și