Sunteți pe pagina 1din 74

Progress in Neurobiology 85 (2008) 1–74

www.elsevier.com/locate/pneurobio

Neurotransmitter, peptide and cytokine processes in relation to


depressive disorder: Comorbidity between depression and
neurodegenerative disorders
Hymie Anisman a,*, Zul Merali b, Shawn Hayley a
a
Institute of Neuroscience, Carleton University, Ottawa, Ontario K1S 5B6, Canada
b
University of Ottawa, Institute of Mental Health Research, Canada
Received 22 August 2007; received in revised form 4 December 2007; accepted 18 January 2008

Abstract
Given the array of biological changes induced by stressors, it is not surprising that these experiences may provoke a variety of illnesses.
Among others things, stressors promote functional changes of neuropeptide and classical neurotransmitter systems. The peptidergic changes, for
instance, include alterations of corticotropin releasing hormone, arginine vasopressin, and bombesin-like peptides at specific brain sites.
Similarly some of the neurotransmitter systems influenced by stressors include GABAergic and monoamine functioning. Variations of these
processes may limit neurogenesis (and dysregulation of growth factors such as BDNF) and influence cellular viability (through NFkB and MAP
kinase pathways). As well, stressors activate the inflammatory immune system, notably the release of signaling molecules (cytokines), which
may provoke many of the same neuropeptide (and other neurotransmitter) changes. By virtue of their actions on neuronal functioning,
inflammatory processes may influence stress-related illness, such as depression, and may be a common denominator for the comorbidity that
exists between depression and neurological conditions, including Parkinson’s and Alzheimer’s diseases, as well as cardiovascular-related
pathology. The present report provides an overview of biological endophenotypes associated with stressors that are thought to be related to major
depressive disorder and related comorbid conditions. The view is taken that synergy between stressors and inflammatory factors may promote
pathological outcomes through their actions on neuropeptides and several neurotransmitters. As well, stressful events may result in the

Abbreviations: 5-HT, serotonin; 5-HT1A, HT1B, 5-HT2A, and 5-HT2C, serotonin receptor subtypes; 5-HTT, serotonin transporter; 5-HTTLPR, 5-HTT gene
promotor polymorphism; ACTH, adrenocorticotropic hormone; AD, Alzheimer’s disease; AGII, angiotensin II; a-MSH, a-melanocyte stimulating hormone; APAF,
apoptotic protease-activating factor ( ); APP, amyolid precursor protein; AVP, arginine vasopressin; BB, bombesin; BB1, BB2, bombesin receptor subtypes; BBB,
blood brain barrier; BDNF, brain-derived neurotropic factor; BDZ, benzodiazepine; BNST, bed nucleus of the stria terminalis; BSA, basolateral amygdala; CeA,
central amygdala; CGRP, calcitonin gene-related peptide; CNS, central nervous system; COX-2, cyclooxygenase-2; CREB, cAMP response element-binding
protein; CRH, corticotropin releasing hormone; CRH1, CRH2, corticotropin releasing hormone receptor subtypes; CRP, C-reactive protein; CSF, cerebrospinal fluid;
DA, dopamine; DEAF-1, nuclear deformed epidermal auto-regulatory factor; DRN, dorsal raphe nucleus; DVC, dorsal vagal complex; ERK, extracellular signal-
regulated kinase; FGF, fibroblast growth factor; FPC, frontopolar cortex; GABA, gamma-aminobutyric acid; G-CSF, granulocyte colony-stimulating factor; GDNF,
glial derived neurotrophic factor; GR, glucocorticoid receptor; GRP, gastrin releasing hormone; GRPR, gastrin releasing hormone receptor; HPA, hypothalamic-
pituitary-adrenal; HPG, hypothalamic–pituitary–gonadal; IDO, indoleamine-2,3-dioxygenase; IFN, interferon; IGF-I, insulin-like growth factor I; IkB, inhibitory
factor kappa B; IL, interleukin; IL-1ra, IL-1 receptor antagonist; INOS, inducible nitric oxidase synthase; IP3, inositol triphosphate; Ir, immunoreactivity; IRS-1,
insulin receptor substrates 1; JAK, Janus kinase; JNK, c-Jun N-terminal kinase; LIF, leukemia inhibitory factor; LPS, lipopolysaccharide; MAPK, mitogen-activated
protein kinase pathways; MAO-A, monoamine oxidase A; MAOI, monoamine oxidase inhibitor; MAP, mitogen-activated protein; MCAO, middle cerebral carotid
artery occlusion; MDD, major depressive disorder; MnSOD, manganese superoxide dismutase; MR, mineralocorticoid receptor; MS, multiple sclerosis; NE,
norepinephrine; NFkB, nuclear factor kappa B; NK, natural killer cell; NMB, neuromedin B; NMDA, N-methyl-D-aspartate; NO, nitric oxide; NSAID, non-steroidal
anti-inflammatory; NT-3, neurotrophin-3; NTS, nucleus of the solitary tract; OCD, obsessive compulsive disorder; OXT, oxytocin; PI3K, phosphatidylinositol 3-
kinase; PACAP, pituitary adenylate cyclase-activating polypeptide; PD, Parkinson’s disease; PET, positron emission tomography; PFC, prefrontal cortex; PGE2,
prostaglandin; PKC, protein kinase C; PR, progressive ratio; PVN, paraventricular nucleus; sICAM-1, soluble intracellular adhesion molecule; sIL-R, soluble IL
receptors; sIPSC, spontaneous inhibitory postsynaptic currents; SNc, substantia nigra pars compacta; SP, substance P; SSRI, selective serotonin reuptake inhibitor;
STAT, signal transducer and activator of transcription; THDOC, tetrahydrodeoxycorticosterone; TLR4, toll-like receptor 4; TNF, tumor necrosis factor; TNF-R1,
TNF-R2, TNF receptor subtypes; TrkB, tyrosine kinase B; VIP, vasoactive intestinal peptide.
* Corresponding author. Tel.: +1 613 520 2699; fax: +1 613 520 4042.
E-mail address: hanisman@ccs.carleton.ca (H. Anisman).

0301-0082/$ – see front matter # 2008 Elsevier Ltd. All rights reserved.
doi:10.1016/j.pneurobio.2008.01.004
2 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

sensitization of neurochemical and cytokine processes, so that later re-exposure to these stimuli may promote rapid and exaggerated responses
that favor illness recurrence.
# 2008 Elsevier Ltd. All rights reserved.

Keywords: Depression; Neurodegeneration; Comorbidity; Endophenotype; Cytokine; Neuropeptide

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2. Stress and depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.1. Moderators of the stress response in relation to pathology. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.2. Stressor appraisals and coping in relation to pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. Biological correlates of depression. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
4. Stressors induce biogenic amine changes akin to those associated with depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
4.1. Biogenic amines: acute stressor effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4.2. Sensitized biogenic amine variations. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.3. Impact of chronic stressor on biogenic amine activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
5. Serotonin signaling and depression in stress-based models of depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.1. 5-HT manipulations influence depressive-like behavioral impairments in animal models . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.2. Serotonin correlates of depression: human studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
5.3. Serotonin variations in depression and among depressed suicides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
5.4. Genetic links between serotonin functioning and depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
6. Corticotropin releasing hormone (CRH) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
6.1. Stressor-induced CRH functioning: animal studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
6.2. CRH and CRH receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
6.3. CRH in depression/suicide. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
6.4. CRH has multiple actions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
7. Arginine vasopressin (AVP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
7.1. Arginine vasopressin in relation to depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
7.2. Arginine vasopressin and CRH synergy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
8. Bombesin (neuromedin B and gastrin-releasing peptide). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
8.1. Bombesin and CRH interactions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
8.2. Bombesin and 5-HT interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
8.3. Bombesin and anxiety. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
9. Leptin, feeding and stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
9.1. Leptin and cytokines. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
9.2. Leptin and reward processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
9.3. Relation to stress processes and depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
9.4. Leptin involvement in atypical depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
9.5. Ghrelin, food consumption and motivational processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
10. Gamma-aminobutyric acid (GABA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
10.1. GABAA variations associated with suicide/depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
10.2. GABA and CRH inter-relations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
10.3. GABA, CRH and 5-HT inter-relations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
11. Cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
11.1. Cytokines as signaling molecules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
11.2. Paracrine and autocrine actions of cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
11.3. Cytokines influence CNS processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
11.4. Neurochemical effects of cytokines: implications for depressive illness. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
11.5. Neurochemical consequences of IL-1b, IL-6 and TNF-a. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
11.6. Cytokine-induced behavioral changes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
11.7. Proactive effects of cytokines and stressors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
11.8. Cytokines in relation to depression in humans . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
11.9. Immune activation provokes depressive mood state . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
11.10. Mechanisms linking cytokine activation and depressive illness. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
12. Morphological correlates of major depressive illness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
12.1. Stressor and corticoid actions in relation to morphological changes associated with MDD . . . . . . . . . . . . . . . . . . . . . . . 32
13. Growth factors in relation to stressors and depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
13.1. BDNF involvement in depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
13.2. Neurogenesis, growth factors and depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
13.3. Neurodegeneration and depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 3

13.4. Downstream growth factor signaling pathways in depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35


14. Neuropeptide and cytokine influences on neurodegeneration: Contribution of the inflammatory immune system . . . . . . . . . . . . . 37
14.1. Anti-inflammatory properties of CRH . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
14.2. Anti-inflammatory properties of a-MSH, VIP, PACAP and IGF-1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
14.3. Proinflammatory effects of peptides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
15. Neuropeptide and cytokine interactions in the modulation of neurodegeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
15.1. Role of cytokines and CRH in stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
15.2. Role of cytokines and CRH in sequelae of acute head injury. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
15.3. Role of cytokines, VIP, and PACAP in Parkinson’s disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
15.4. Role of IGF-I and cytokines in Alzheimer’s disease (AD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
16. Cytokines mediate the comorbidity between depression and neurodegenerative conditions . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
16.1. Depression and comorbid cardiovascular (heart and stroke) disturbances. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
16.2. Depression and comorbid Parkinson’s disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
16.3. Depression and comorbid Alzheimer’s disease (AD). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
17. Summary and conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47

1. Introduction depression to comorbid illnesses, including those associated


with the aftermath of stroke (and head injury) and those of a
Stressful events engender multiple neurochemical and neurodegenerative nature (Parkinson’s disease and Alzheimer’s
hormonal alterations, presumably as an adaptive response to disease), with the aim of defining some of the shared processes
meet environmental demands. Yet, pathological outcomes are that subserve them. Understanding these common processes
frequent sequelae of stressful experiences, which might not be may offer insights into the mechanisms subserving these
surprising given the wide range of biological changes that are disease states, might provide important clues for effective
engendered. The specific illness that emerges in response to prophylactic and therapeutic treatment, and will reinforce the
stressors might reflect vulnerability to particular biological view (Martin, 2002) that barriers between psychiatry and
alterations and ‘‘weak links’’ in endogenous buffering neurology are counterproductive.
mechanisms. Evidently, in some individuals the neurochemical
alterations are insufficient to deal with the insults (varying with 2. Stress and depression
the nature of the stressor, or the individual’s genetic or
experiential disposition) or may become overly taxed (allostatic Studies in animals have shown that stressors markedly
overload), culminating in adverse outcomes (McEwen, 2006; influence central neurochemical processes that may be pivotal to
Schulkin et al., 1998). As well, in some instances the the provocation of depressive illness. These neurochemical and
‘‘adaptive’’ biological changes themselves may have secondary hormonal changes are thought to serve in an adaptive capacity
adverse consequences. (de Kloet et al., 1999; Kovacs and Sawchenko, 1996; Sapolsky
It seems that illnesses frequently do not occur in isolation of et al., 2000; Sato et al., 1997; Sawchenko et al., 1996), being
one another, and comorbid conditions are common, particularly fundamental in maintaining allostasis (i.e., homeostasis pertain-
in chronic illnesses. In the case of depression, comorbidities ing to the stress response). Among other things, these
have been reported with respect to anxiety (Nutt and Stein, neurochemical changes may prepare the organism to deal (cope)
2006), cardiovascular disease and stroke (Bondy, 2007; with the stressor, blunt its psychological impact, and stimulate
Frasure-Smith and Lesperance, 2005) as well as neurodegen- processes that preclude excessive physiological activation, such
erative disorders such as Parkinson’s and Alzheimer’s diseases, as immune over-reaction (de Kloet et al., 1999; McEwen, 2000;
and multiple sclerosis (Griffin et al., 2006; Lieberman, 2006; Sapolsky et al., 2000). Yet, if the stressor experience is
Mohr, 2007; Owens, 2002; Wheeler and Owens, 2005). The sufficiently severe and/or long lasting (i.e., chronic strain), then
presence of comorbid illnesses may complicate treatment, and the load placed on biological systems may become excessive
there is reason to believe that depression itself (and the (allostatic overload), ultimately resulting in increased vulner-
antecedent or concurrent distress) may limit recovery from co- ability to pathology (McEwen, 2000). Of course, genetic or
occurring pathologies (Rosenthal, 2003). experiential factors might influence the organism’s adaptive
The present report describes the contribution of stressful capacity (or hardiness) in dealing with such insults and it is likely
events to the emergence of various endophenotypes that that these factors, among others, moderate the stress response
comprise depression, including the possible biological pro- (Tannenbaum and Anisman, 2003).
cesses (neurotransmitter, hormone, neuropeptide, growth factor Paralleling the animal studies, there is considerable evidence
and cytokine) that might be responsible for the disorder. In so from human research confirming that stressful experiences
doing, we will outline some of the moderating conditions that promote or exacerbate depressive symptoms (Kessler, 1997;
influence these biological changes, and will relate stress and Paykel, 2003) and may elicit or aggravate several other
4 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

psychological and physical disorders. In evaluating the impact influence character traits that affected resilience to severely
of stressors on pathology, however, it ought to be considered traumatic events. No doubt, a set of resiliency factors, including
that there is appreciable variability in how stressors are early experiences and genetic factors, likewise contribute to the
perceived, the emotional reactions to them, as well as the emergence of depression in the face of stressors.
peripheral and central neurochemical changes elicited. Thus,
individuals may differ with respect to the adverse effects 2.2. Stressor appraisals and coping in relation to
elicited by stressful events. Indeed, it is generally thought that pathology
major depressive disorder (MDD) is biochemically hetero-
geneous, wherein symptoms as well as the processes leading to The endorsement of inappropriate or ineffective coping
depression, may vary across individuals; consequently, the strategies to deal with stressors has been implicated as being
treatments effective in alleviating the disorder may vary fundamental in the promotion of psychological illnesses such as
accordingly. This, coupled with the limited success in defining MDD. It is thought that when confronted with a stressor,
the genetic compliments associated with depression, has lead to individuals make appraisals regarding the threat, and this
the suggestion that analysis of MDD might not always be best subjective interpretation influences the coping methods
served when viewed as a syndrome or phenotype. Instead, it endorsed (Lazarus and Folkman, 1984). Appraisals comprise
might be more profitable to evaluate endophenotypes that make a constellation of evaluative dimensions, including the threat or
up the disorder (i.e., focusing on genetic factors and specific risk associated with the event, its severity, controllability,
neurochemical processes related to particular symptoms of predictability, ambiguity, and the meaning imposed. Variations
illness) (e.g., Cryan and Slattery, 2007; Gottesman and Gould, along each of these dimensions are associated with perceived
2003; Hasler et al., 2004). In this regard, the neurochemical distress and may govern the coping methods selected to deal
disturbances associated with depressive disorders have been with the stressor (Anisman and Matheson, 2005).
considered endophenotypes that might be responsible for Coping strategies are often classified into two general types;
different aspects or symptoms of the illness (Hasler et al., problem-focused and emotion-focused (Billings and Moos,
2004). Analyses of endophenotypes likely represent a highly 1985; Lazarus and Folkman, 1984), which subsume multiple
effective and valid approach, so long as it is appreciated that the strategies (e.g., emotion-focused coping may comprise emo-
constellation of symptoms and neurochemical characteristics tional expression, emotional containment, self- or other-blame,
associated with psychiatric illnesses likely reflect complex withdrawal, denial, and passive resignation). In addition, several
interactions between several biological systems. Essentially, it alternative coping methods can be used, including cognitive
may be that a concatenation of neurotransmitter alterations is restructuring (re-evaluating the threat), social support seeking,
fundamental to the emergence of pathology. active distraction, religion, humor, wishful thinking and
rumination (Carver et al., 1989; Endler and Parker, 1990;
2.1. Moderators of the stress response in relation to Matheson and Anisman, 2003; Nolen-Hoeksema, 1998). It is
pathology often taken as axiomatic that in situations in which the individual
has control, problem-focused strategies (e.g., problem solving,
Analyses of stressor effects on pathology based exclusively on cognitive restructuring or positive growth) are adaptive, whereas
animal studies can only go so far in predicting pathological those strategies that encourage undue focus on emotions (e.g.,
outcomes. Although animal studies can be instrumental in rumination, emotional venting, self-blame) are not (Carver et al.,
defining many of the neurochemical mediators and consequences 1989; Lazarus and Folkman, 1984). This said, it was suggested
of stressor outcomes, some of the variables influencing that in emotionally charged situations, emotional-approach
vulnerability to pathology cannot readily be assessed in animal coping might facilitate the individuals ability to come to terms
models. Considerable animal research has focused on the with their feelings and in so doing, distress may be reduced
contribution of genetic factors and early life experiences (as well (Stanton et al., 1994). Likewise, although avoidance often works
as those related to aging) and the nature of the stressor against individuals’ well-being in the long-run, it may provide
experienced (including specific characteristics of the stressor temporary relief from an ongoing stressor, allowing the
such as its predictability, ambiguity, controllability and individual the opportunity to adopt or develop more effective
chronicity) in promoting pathological outcomes. Other pro- strategies (Suls and Fletcher, 1985).
cesses, such as appraisal and coping mechanisms that moderate Any given coping strategy may serve different functions, or
the impact of stressful experiences on the evolution and operate to facilitate or inhibit other strategies. For instance,
exacerbation of pathology are more difficult to assess in animals social support can be used for a variety of purposes, including a
(Anisman and Matheson, 2005). Likewise, several factors have source of information, a shoulder to cry on, a means of
been identified that limit the impact of stressors (i.e., promote distracting oneself, and so forth. However, it is important to
stressor resilience), and although some of these can be evaluated note that as much as social support may provide an effective
in animal models, others cannot. Charney (2004), for instance, buffer against ongoing stressors (Underwood, 2000), the
suggested that neural mechanisms related to reward and perception of unsupport (e.g., not receiving support when it
motivation (hedonia, optimism, and learned helpfulness), was reasonably expected) may have marked repercussions on
responsiveness to fear and fear-related situations, and adaptive depressive state (Ingram et al., 1999). Further to the same point,
social behaviors (altruism, bonding, and teamwork) all acted to although rumination has been associated with depressive illness
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 5

(Nolen-Hoeksema, 1998), this method of dealing with stressors likely that appraisals, coping, and the many factors that moderate
does not occur in isolation of other coping strategies. these responses, may influence the neurochemical alterations
Interestingly, it was observed that rumination ordinarily co- associated with stressors (and the converse is no doubt the case,
occurred, in controls, with a broad constellation of problem- and as well), thereby favoring the development of depressive illness.
emotion-focused strategies, as well as cognitive disengagement.
In contrast, among dysthymic patients, rumination was 3. Biological correlates of depression
associated with a limited array of strategies related to
emotion-focused coping, and inversely related to efforts to It is clear from various studies assessing neurotransmitter
disengage (Kelly et al., 2007). Thus, the effectiveness of a given alterations associated with MDD, that multiple processes may be
strategy may be determined by how it is used in conjunction with perturbed and could potentially subserve depressive illness.
other strategies, and perhaps in how flexible individuals are in These have included variations of brain levels and turnover of 5-
their selection of these strategies. This said, it appears, as well, HT, alterations of the 5-HT transporter (5-HTT) and several 5-HT
that the specific coping strategies endorsed may vary over time receptors (5-HT1A, HT1B, 5-HT2A, and 5-HT2C), DA auto-
and across situations (Delongis and Holtzman, 2005; Tennen receptors, and a1-NE or b-NE receptors (Maes and Meltzer,
et al., 2000), and might also serve different functions as the 1995; Nestler et al., 2002; Nutt, 2002; Wong et al., 2000).
stressor evolves. Of course, individuals bring with them certain Moreover, depression has been associated with alterations of
coping dispositions or styles that influence the strategies adopted peptidergic functioning (e.g., corticotropin releasing hormone;
most readily across different situations (e.g., Carver et al., 1989). CRH), including neuronal release or the number and sensitivity of
Typically, depressive mood is accompanied by the use of peptide receptors (Mitchell, 1998; Nemeroff, 1996; Reul and
emotion-focused coping and rumination, at the expense of Holsboer, 2002), as well as alterations of growth factors (e.g.,
problem-oriented coping and social support seeking (Carver brain-derived neurotrophic factor; BDNF) (Duman and Mon-
et al., 1989; Matheson and Anisman, 2003; Ravindran et al., teggia, 2006; Duman et al., 2000, 2001; Manji et al., 2001) and
1999). However, this still begs the question, as to how certain cytokines that are more typically considered as immune system
coping styles came to predominate. For example, is there a signaling molecules (e.g., interleukin-1, -2 and -6, interferon-a,
genetic disposition to endorse one style over another, or are the tumor necrosis factor-a (Hayley et al., 2005; Maes, 1999)).
styles developed during early childhood subsequently main- Moreover, the downstream signaling factors associated with
tained? Likewise, are particular coping styles secondary to several of these signaling molecules, such as cAMP, CREB and
personality factors such as poor self-esteem? Ultimately, it is protein kinase A, have been implicated in depressive illness

Fig. 1. Schematic representation depicting potential routes by which stressors and cytokines could influence depressive state. A stressor could potentially influence
major depression through two major routes that feed into several interconnected loops. Stressors and cytokines both increase hypothalamic (and extra-hypotalamic)
CRH release, although in the case of stressors this outcome may involve activation of bombesin-like peptides (neuromedin-B and gastrin releasing peptide; NMB and
GRP, respectively). In addition to activating HPA functioning, CRH may influence 5-HT processes, and GABAA activity may act as a mediator in this regard. This, in
turn may influence depression directly, or may do so by impairing neuroplastic processes. An alternative, although not necessarily mutually exclusive pathway
involves cytokine/stress activation of either NFkB, MAP kinases, or JAK/STAT signaling. These would influence oxidative or apoptotic mechanisms, leading to
altered growth factor expression (e.g., BDNF), hence again favoring impaired neuroplastic processes, culminating in major depression.
6 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

(Dwivedi et al., 2002). To this point, however, it is uncertain neurotransmission (using animal models) might provide
whether specific features of depression correspond to particular fundamental information concerning the specific changes
neurochemical changes (e.g., are the DA changes related to associated with this illness. As well, this line of research
anhedonic features of depression, or are 5-HT reductions in may help in the identification of the specific sites at which these
frontal regions related to impulsivity among depressed alterations occur, the factors that favor the provocation of these
suicides?). stressor-induced effects, and conversely, the conditions (and
The multiple neurochemical alterations that have been treatments) that might limit their occurrence or impart
presumed to be associated with depression likely do not act resiliency.
independently of one another. Some may reflect serial variations To a considerable extent, the sections that follow rely
(induction of a cascade of neuronal changes), whereas others may heavily on data derived from animal studies that pertain to
reflect parallel processes stemming from common instigators stressor-provoked neurochemical changes. Of course, such
(e.g., stressful experiences). As will be described in the ensuing studies are fundamental in delineating the effects of stressors,
sections (depicted in Fig. 1), it is our view that stressful events and at least some of the factors that moderate the impact of
favor the development of several neurochemical disturbances that adverse events on neurochemical processes and on behaviors
ultimately come to promote the emergence of depressive that might reflect a depressive-like state. Yet, as already
symptoms (or exacerbate the symptoms of an already existent indicated, attempts to generalize animal-based findings to
depression). In addition to monoaminergic neurotransmitter human pathologies are exceptionally difficult given the
system involvement, we propose that stressors promote variations fundamental role of cognitive (e.g., stressor appraisal)
of several neuropeptidergic (serving as neurotransmitters and/or processes in accounting for the reaction to stressors. This is
neuromodulators) systems (e.g. bidirectional interactions made all the more difficult given that, in humans, certain types
between neuromedin B (NMB), gastrin releasing hormone of stressors (e.g., those dealing with social threats) may engage
(GRP) and CRH systems). In addition, many of these peptidergic different processes than those involving other forms of distress,
systems modulate classical neurotransmitter systems. Illustra- such as shame, loss and rumination (Anisman and Matheson,
tively, CRH and NMB variations influence 5-HT functioning, 2005; Dickerson and Kemeny, 2004). Indeed, these very potent
possibly being moderated by GABAA neuronal activity stressors that could have far reaching neurochemical and
(Brambilla et al., 2003; Shiah and Yatham, 1998; Tunnicliff psychological ramifications, cannot be mimicked in animal
and Malatynska, 2003; Merali et al., 2004a,b). Further, stressors models. Furthermore, in humans, stressors that involve
may come to affect processes associated with neurogenesis and anticipation are generally associated with anxiety, whereas
cell survival (possibly involving growth factors such as BDNF), those involving loss are frequently more closely aligned with
which likewise might contribute to major depressive disorder. In depression (Monroe and Depue, 1991). It appears, as well, that
this regard, a major focus of the model provided in Fig. 1 is that gender differences may exist with respect to the impact of
certain stressors, like immunogenic challenges, may provoke certain types of stressors. For instance, there is reason to
activation of the inflammatory immune system, and the ensuing suppose that depressive symptoms are more likely to be
release of immune signaling molecules (cytokines). These induced by social stressors in females than in males, whereas in
cytokines, in effect, function like stressors in affecting males, factors such as job loss or demotion will be more likely
neurochemical processes, and may act synergistically with to induce depression. Thus, in evaluating the impact of
traditional stressors in provoking these outcomes. As well, stressors, and the mediating role of neurochemical changes in
sensitization of neurochemical responses occur to a variety of promoting depression, caveats concerning social determinants
insults, including neurogenic and psychogenic stressors, so that of the stress response limit the utility of animal studies.
the response to later challenges are increased. In addition to the Although stressors have ubiquitous consequences, affecting
exaggerated responses elicited by stressors similar to those several neurotransmitter systems across a number of brain
initially encountered (or cues that act as reminders of these regions, not all stressors have the same effects, and different
stressors), cross-sensitization between different types of chal- neural circuits may be engaged, depending on the nature of the
lenges may also occur. It is believed that the sensitization of stressor encountered. ‘‘Processive’’ stressors (i.e., those that
neurochemical responses might contribute to the development of involve higher-order processing, e.g., fear conditioning,
pathology, such as depression. Moreover, because these same predator cues) are thought to induce hypothalamic–pituitary–
mechanisms also contribute to neurodegenerative processes, it is adrenal (HPA) activation through limbic processes, whereas
suggested that synergies between stressors and cytokines, and the ‘‘systemic’’ challenges (e.g., respiratory, cardiovascular,
sensitization of the neurochemical processes associated with immune and cytokine challenge) might do so through non-
stressors, may contribute to comorbidity frequently seen between limbic mechanisms (Herman and Cullinan, 1997). Interest-
psychological and neurodegenerative disorders. ingly, despite the broad impact of stressors, some degree of
selectivity seems to occur. Of the processive types of stressors,
4. Stressors induce biogenic amine changes akin to those with physically aversive properties (termed neurogenic
those associated with depression stressors) and those of psychological origin (psychogenic
stressors; learned threats and those that involve innate threats
Given the prominent role of stressors in the provocation and such as predator odor) might also engage different neural
recurrence of MDD, analyses of stressor effects on brain circuits. For example, although different stressors provoked the
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 7

in vivo release of amygdala CRH, antagonists of this peptide did forced swim stressor, 5-HT1A receptor density was reduced in
not attenuate the anxiety elicited by innate threats, but reduced the dorsal raphe nucleus (DRN) and in the hippocampus,
that associated with neurogenic and learned stressors (Merali whereas 5-HT1A receptor density was elevated in the thalamus,
et al., 2004a,b, 2006a). To be sure, it is difficult to compare the hypothalamus, and amygdala (Briones-Aranda et al., 2005).
influence of different types of stressors given that they may be Moreover, the development of behavioral impairments follow-
appraised differently or they may not be equally aversive. ing stressor exposure may be related to hypersensitivity of 5-
Nevertheless, it appears that psychogenic and neurogenic HT2 receptors and desensitization of 5-HT1B receptors
insults differentially influence in vivo amygdala or prefrontal (Bolanos-Jimenez et al., 1995; Nankai et al., 1995). By
cortical NE and CRH activity (Anisman and Matheson, 2005; example, it was reported that a stressor regimen that
Hayley et al., 2001a; Lu et al., 1999; Merali et al., 2001, 2004a; engendered behavioral impairments in a learned helplessness
Morrow et al., 2000) and might thus have different behavioral paradigm, increased mRNA expression of 5-HT1B receptors
consequences. within the DRN, without affecting postsynaptic 5-HT1B
expression within the hippocampus or PFC (Neumaier et al.,
4.1. Biogenic amines: acute stressor effects 1997).
Like neurogenic challenges, psychogenic insults (e.g.,
Like the presumed neurochemical correlates of depression, predator odor, social defeat) influence NE release from
stressors may elicit NE, DA and 5-HT release in several brain mesolimbic neurons (Anisman et al., 1993; Hayley et al.,
regions, varying as a function of experiential and organismic 2001a; Merali et al., 2001, 2004a,b) and 5-HT release at the
factors (e.g., age, strain) (Ritter and Pelzer, 1978; Shanks et al., hippocampus (Keeney et al., 2006). Likewise, social defeat was
1994, 1991; Stanford, 1995). The NE and 5-HT variations are found to increase hypothalamic 5-HT turnover, and the anxiety
notable in specific hypothalamic nuclei (e.g., paraventricular that accompanied the 5-HT variations were attenuated by
nucleus; PVN) and various mesocorticolimbic sites, including chronic antidepressant treatment (Beitia et al., 2005). Despite
the hippocampus, amygdala and prefrontal cortex (PFC). The the obvious relevance of psychosocial stressors in determining
DA changes are also exceedingly sensitive to stressors, being pathological outcomes, to this point analyses of the impact of
notable within the arcuate nucleus, nucleus accumbens and psychogenic stressors on monoamine functioning have not been
other mesocorticolimbic sites (Deutch et al., 1991; Kalivas and examined as extensively as those elicited by neurogenic insults
Duffy, 1995; Puglisi-Allegra et al., 1991; Zacharko and (but see Tamashiro et al., 2005). Nevertheless, it seems that
Anisman, 1991), and there have been reports that different such challenges may instigate anxiety-like effects similar to
neurogenic insults may influence nigrostriatal DA activity those elicited by neurogenic stressors. However, as alluded
(Abercrombie et al., 1989; Nisenbaum et al., 1991). In vivo to earlier, the response to some psychogenic stressors appears to
studies indicated that mesocorticolimbic amine alterations be ‘‘prewired’’ (e.g., predator-related stressors) and mono-
could be induced by relatively mild stimuli (tailpinch, novelty) amine and neuroendocrine alterations may be distinguishable
and by psychosocial stressors (Cenci et al., 1992; Doherty and from those elicited by learned stressors (Anisman et al., 2001;
Gratton, 1992, 1996; Feenstra et al., 1995; Kalen et al., 1989; Hayley et al., 2001a; Merali et al., 2004a; Morrow et al., 2000).
Tidey and Miczek, 1996) and a hemispheric mesocorticolimbic Thus, there is reason to believe that these different types of
DA bias appears to exist in response to stressors (Sullivan and insults will also elicit dissimilar behavioral disturbances.
Gratton, 1998). The latter findings may be important given the In general, it is thought that given a sufficiently severe
finding that a key feature associated with MDD may be related challenge, particularly when termination of the stressor is not
to dysregulation of hemispheric activation within the PFC controllable by the animal, amine release may exceed rate of
(Johnstone et al., 2007). Specifically, in nondepressed synthesis, and as a result depletion of NE and 5-HT stores will
individuals left-lateralized activation in the PFC occurred ensue (Anisman and Zacharko, 1991; Heinsbroek et al., 1989;
when participants were down-regulating negative affect. In Weiss et al., 1981). The diminished monoamine reserves would
contrast, bilateral PFC activation was evident among depressed render animals less able to contend with further insults, and as a
individuals when making such efforts. Furthermore, during an result behavioral impairments would be more likely to occur.
effortful affective reappraisal task, an inverse relationship Consistent with this view, it has frequently been reported that
between activation in left ventrolateral PFC and the amygdala, uncontrollable (inescapable) stressors engender reductions of
likely mediated by the ventromedial PFC was apparent among hypothalamic NE and hippocampal 5-HT relative to the effects
nondepressed individuals, whereas this relationship was provoked by controllable (escapable) stressors (Anisman and
dysregulated among depressed individuals. In effect, these Zacharko, 1991), and are also more potent in provoking 5-HT
data suggest that inappropriate engagement of right prefrontal release at the basolateral amygdala (Amat et al., 1998a), medial
cortex, coupled with limited engagement of left lateral- PFC (Bland et al., 2003a), ventral hippocampus and dorsal
ventromedial prefrontal circuitry may serve an important role periaqueductal gray (Amat et al., 1998b) as well as the nucleus
in regulating affective (and perhaps cognitive) responses to accumbens shell (Bland et al., 2003b). Moreover, just as
negative stimuli (Johnstone et al., 2007). escapable and inescapable stressors differentially influence 5-
In addition to variations of monoamine reserves and HT functioning, it was shown that if rats were first trained in an
turnover, stressors influence the expression of various receptor escapable test (a treatment that immunizes animals against the
subtypes. By example, it was reported that 24 h following a behavioral disturbances elicited by an uncontrollable stressor),
8 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

the increased 5-HT release at the DRN ordinarily elicited by an activity, may be unique for each transmitter system. In the case of
inescapable stressor was prevented. As this effect was stressor-induced sensitization of dorsal raphe 5-HT neuronal
dependent on activation of the ventral aspect of the medial activity, which has been implicated in depression and anxiety, it
PFC, it was suggested that this site might be essential for was suggested that a ligand related to CRH2 receptors might be
storage or plasticity as it relates to information concerning essential in promoting this outcome (Maier and Watkins, 2005).
stressor controllability (Amat et al., 2005, 2006). Moreover, it has been reported that the cross-sensitization
between stressors and amphetamine likely involves NMDA-
4.2. Sensitized biogenic amine variations glutamatergic receptors, as the sensitization can be blocked by
the NMDA antagonist, MK-801, infused directly into the brain
Typically, stressor-provoked amine variations are short-lived, (Pacchioni et al., 2002). Especially intriguing data have been
lasting only minutes or hours, depending upon characteristics of reported by Stewart and her associates (e.g., Flores and Stewart,
the animal (e.g., age, strain) and/or the stressor. However, stressor 2000; Flores et al., 2002) suggesting that bFGF, a neurotrophic
experiences may proactively influence the response to subse- factor produced by astrocytes, may be fundamental for the
quent challenges, so that monoamine responses occur more neuronal plasticity associated with the protracted effects elicited
readily and are more profound than in naive animals (i.e., by stressors and by catecholamine stimulants. At this juncture it
sensitization). By example, upon re-exposure to a mild stressor, is premature to dismiss any of these potential mechanisms in
the utilization of NE and mesocortical DA is enhanced, and this mediating sensitization effects, and it is indeed possible that
occurs even if the later exposure involves a different type of multiple processes may subserve the diverse sensitized responses
stressor (Abercrombie and Jacobs, 1988; Abercrombie et al., that have been associated with stressors.
1989; Anisman and Sklar, 1979; Cassens et al., 1980; Fadda et al.,
1978; Jordan et al., 1994; Kalivas and Duffy, 1995; Pezzone et al., 4.3. Impact of chronic stressor on biogenic amine activity
1992, 1993; Rabin et al., 1995; Roth et al., 1988; Smith et al.,
1992, 1997; Yoshioka et al., 1995). Likewise, 5-HT changes The variations of monoamine activity prompted by repeated
within the ventral medial PFC associated with a stressor can be (chronic) insults are distinct from those engendered by acute
readily re-engendered upon subsequent re-exposure to a stressor insults. Specifically, the reductions of amine levels provoked by
(or upon subsequent morphine treatment), but this effect was acute stressors may not be apparent following protracted or
only apparent if the initial stressor was one that was repeated insults (Deutch and Roth, 1990; Deutch et al., 1991;
uncontrollable (Maier et al., 2006). Irwin et al., 1986; Roth et al., 1988), probably owing to
Among previously stressed animals augmented amine compensatory enhancement of NE and 5-HT synthesis (Haleem
responses are also observed in response to pharmacological and Parveen, 1994; Irwin et al., 1986; Nankova et al., 1993) and
stimulants, such as amphetamine or cocaine (Anisman et al., moderation of DA utilization (Gresch et al., 1994). However,
2003; Antelman and Chiodo, 1984; Deroche et al., 1995; this outcome may be dependent upon the nature of the stressor,
Gresch et al., 1994; Herman et al., 1984; Kalivas and Stewart, as the effects of chronic social stressors appear to be relatively
1991; Nisenbaum and Abercrombie, 1992; Nisenbaum et al., persistent (Buwalda et al., 2005).
1991; Robinson, 1988; Robinson et al., 1987). Thus, stressor The ‘‘adaptation’’ frequently observed with chronic stres-
experiences may not only influence behavioral responses to sors does not necessarily reflect a diminution of the high levels
later stressor challenges, but may also be important in of amine utilization. Indeed, in chronically stressed animals,
moderating the response to certain drugs (Kalivas and Stewart, amine levels do not simply return to pre-stress levels, but may
1991). It has been suggested that basic fibroblast growth factor actually come to exceed basal values, potentially because of the
(bFGF), a neurotrophic factor produced by astrocytes, may be compensatory increase of amine synthesis (Anisman and
essential for the neuronal plasticity leading to the protracted Zacharko, 1991; Blanc et al., 1980). Interestingly, the elevated
responses elicited by stressor and/or catecholamine stimulants monoamine synthesis may persist well after stressor termina-
(Flores and Stewart, 2000; Flores et al., 2002). tion, resulting in elevated monoamine levels, hence rendering
In addition to the altered amine levels and turnover, re- animals better prepared to deal with subsequent stressors
exposure to stressors has been reported to enhance hippocampal (Shanks et al., 1994).
5HT1A receptor density, while decreasing the affinity of those Possibly as a consequence of the excessive monoamine
receptors. In contrast, whereas 5-HT2A receptor density was release, chronic stressors also promote down-regulation of b-
unaffected, its affinity was increased (Harvey et al., 2003). It NE receptor activity and the NE sensitive cAMP response
seems that the sensitized neuronal changes that have been (Stone, 1987) and reduce 5-HT1B receptor expression
reported may be tied to anxiety processes, as the amine changes (Bolanos-Jimenez et al., 1995). It was likewise reported that
elicited by stressor re-exposure or stressor-related cues can be acute and repeated stressors differentially influenced 5-HT2A
attenuated by anxiolytics (Coco et al., 1992; Fadda et al., 1978; receptor mRNA and protein expression, and these effects were
Kaneyuki et al., 1991; Laveille et al., 1978; Wedzony et al., 1996) brain region-specific. In particular, an acute stressor resulted in
and by 5-HT1A receptor manipulations (Wedzony et al., 1996). a decline of 5HT2A receptor mRNA and protein expression in
The processes underlying sensitized neurochemical changes hippocampus. However, this treatment resulted in elevated
have yet to be fully understood, however, it is likely that the frontal cortex 5-HT2A mRNA expression, coupled with a
processes associated with sensitized 5-HT, DA and NE neuronal decrease of receptor expression in the hippocampus and
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 9

hypothalamus of those rats that displayed behavioral impair- with stressors can be attenuated by treatments that influence 5-
ments in a learned helplessness paradigm (Dwivedi et al., HT availability. For example, pharmacological treatments that
2005). It was likewise reported that chronic unpredictable influence hippocampal 5-HT2C receptors may diminish anxiety
stressor exposure was associated with an increase of 5-HT2A in elicited by stressors (Merali et al., 2006a). Likewise, it was
the cortex, coupled with an increase of D1 receptor density repeatedly shown that when administered chronically, anti-
(Ossowska et al., 2001). depressants that affect 5-HT reuptake effectively attenuated the
Like acute insults, chronic stressors proactively influence behavioral interference associated with prior uncontrollable
the response to later challenges; however, whereas acute foot-shock in a learned helplessness paradigm (Anisman and
stressors provoke sensitization of mechanisms associated with Zacharko, 1991), and that the effectiveness of these treatments
NE release, chronic stressors may also induce sensitization of were related to frontal cortex 5-HT variations (Petty et al.,
processes associated with synthesis (Anisman et al., 2003). It is 1992, 1994). Likewise, the learned helplessness effect could be
interesting, as well, that when rats were exposed to a chronic attenuated by bilateral hippocampal infusion of a 5-HT
stressor regimen, subsequent exposure to a different stressor reuptake inhibitor, and this effect could be prevented by the
provoked a still greater increase of hippocampal NE release 5HT1A antagonist 8-OH-DPAT, thus implicating this receptor
(cross-sensitization), which was accompanied by elevated subtype in the stressor-induced interference (Joca et al., 2003).
transmitter synthesis (Finlay et al., 1995; Nisenbaum et al., As performance in shock escape tests may be confounded by
1991). However, it seems that the specific pattern of stressor motoric factors (i.e., disturbances in the ability to initiate and
exposure (and perhaps the nature of the stressor used) is maintain active responses; Anisman et al., 1978), it is
important in determining whether sensitization would be interesting that the effects of antidepressants that influence
established (Jedema et al., 1999). 5-HT or both 5-HT and NE (escitalopram, desipramine) were
Although anxiolytics attenuated the NE and DA release also apparent in cognitive tests, including disturbed cognitive
elicited by acute stressors, in rats chronically stressed and then set-shifting and behavioral flexibility, both of which can be
exposed to a different stressor, the augmented amine release was features of depression (Bondi et al., 2007; Lapiz et al., 2007).
not attenuated by diazepam (Finlay et al., 1995). It seems that Paralleling the effects of pharmacological manipulations, in
following a chronic stressor experience, subsequent exposure to a mice with deletion of 5-HT1A receptors, anxiety responses were
heterotypic stressor (i.e., ones different from the original elevated, as were autonomic indices of anxiety (Toth, 2003).
experience) provokes pronounced neurochemical alterations Yet, increased anxiety was not uniformly evident in either 5-
that are relatively resistant to the effects of anxiolytics. In a like HT1A or 5-HT1B knockouts (Groenink et al., 2003), although
fashion, chronic, unpredictable stressors might promote sus- this may have been due to background strain (genetic context)
tained neurochemical alterations that could render animals more used in these studies. Further to this same point, a gender bias
vulnerable to pathological outcomes. Albeit speculative, these was apparent in a forced swim test in that a reduction of
findings may be relevant to psychological illnesses that appear to immobility was evident among females, but not males (or in
be relatively treatment resistant. wild-type counterparts), with 5-HT1B receptor deletion (Jones
Summarizing, the increased monoamine availability and Lucki, 2005). Based on analyses of 5-HT release in males
(reserves) associated with chronic stressors may enable the and females, coupled with the behavioral effects of the selective
organism to deal with ongoing or impending challenges. serotonin reuptake inhibitor (SSRI), fluoxetine, these investi-
However, the elevated monoamine turnover can only go so far gators suggested that a sex-linked disinhibition of 5-HT release
in limiting adverse outcomes. As indicated earlier, if neuronal was responsible for elevated baseline hippocampal 5-HT levels
activation is sufficiently sustained, then the wear and tear on as well as vulnerability to stressor-elicited 5-HT depletion and
monoamine systems (and those secondary to the amine the ensuing behavioral impairments.
activation) may become excessive, rendering animals vulnerable In addition to the receptor changes, there have been several
to several pathologies, including depression (Fontenot et al., reports implicating the 5-HT transporter (5-HTT) in depres-
1995; McEwen, 2000; McEwen and Magarinos, 2001). In this sion. Specifically, siRNA-mediated knockdown of 5-HTT
regard, vulnerability to pathology is not limited to psychological within the raphe nucleus was associated with attenuated
disturbances, as it was reported that the immunoenhancement immobility in a forced swim test, just as the antidepressant,
provoked by acute stressors may turn to immunosuppression citalopram, provoked this outcome (Thakker et al., 2005). In
after protracted insults (Dhabhar, 2000), potentially influencing contrast to these findings, however, 5-HTT knockout mice
vulnerability to immunologically related disturbances. displayed increased immobility in a repeated forced swim test
(Wellman et al., 2007), and exhibited elevated anxiety and
5. Serotonin signaling and depression in stress-based depression in several behavioral paradigms (Lira et al., 2003).
models of depression The anxiety-like behaviors were particularly pronounced
among knockouts that had been exposed to early life stressor
5.1. 5-HT manipulations influence depressive-like experiences (Carroll et al., 2007), suggesting that the
behavioral impairments in animal models influence of the 5-HTT knockout interacts with such
experiences (just as similar effects were documented in
Consistent with the view that the 5-HT system plays a human studies; see ensuing section). As well, it appeared that
pivotal role in depression, behavioral disturbances associated the behavioral disturbances among 5-HTT knockouts could
10 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

be antagonized by repeated antidepressant (fluoxetine) double knockout mice deficient of 5-HTT and BDNF, the
treatment (Holmes et al., 2002) as well as by the 5-HT1A elevation of ACTH in response to a stressor was especially
antagonist WAY 100635, once again implicating 5-HT1A marked, and the presence of dendrites on hypothalamic and
receptor involvement in such effects (Holmes et al., 2003a). hippocampal neurons was diminished (Ren-Patterson et al.,
Interestingly, among 5-HTT knockouts the increased reac- 2006). In such double knockouts, the effects of 5-HT on
tivity to mild stressors and the elevated anxiety-related anxiety-related behaviors were more profound in males than in
behaviors were accompanied by diminished 5-HT1A receptor females. As females also did not exhibit the marked reduction
expression. Thus, the effects of the 5-HTT knockout might be in the expression of TrkB receptors or that of DA, it was
secondary to the 5-HT1A receptor variations. Indeed, suggested that estrogen might afford protection against the
adenoviral assisted elevation of 5-HT1A expression in these TrkB receptor changes (Ren-Patterson et al., 2006). Of course,
5-HTT knockouts attenuated the exaggerated adrenocortico- in relation to depression, such findings beg the question of why
tropic hormone (ACTH) response elicited by a mild stressor. human females are more likely to become depressed relative to
However, other indices of anxiety (exploratory behaviors in males. It appears likely that other biological factors underlie the
an open field and a plus maze) were unaltered. Furthermore, illness, or that females are more likely to encounter stressors,
although 5-HTT knockout mice displayed hypoactivity, appraise stressors differently, utilize different coping styles, or
anhedonia was not apparent in a sucrose preference test are influenced by any of a number of other psychosocial factors.
(Kalueff et al., 2006). It may be that the influence of 5-HT
neuronal functioning (like the effects of CRH manipulations) 5.2. Serotonin correlates of depression: human studies
is situation-specific. Alternatively, it may be that 5-HTT
influences one component of the depressive profile, whereas Although considerable data have implicated both NE and
5-HT1A variations affect a second feature (e.g., anxiety). DA alterations in mediating MDD, particular attention has
From this perspective, genetic deletion may have cascading focused on the involvement of 5-HT processes (Millan, 2006).
effects so that outcomes distal from the original genetic To a considerable extent this stems from the well-documented
manipulation might be responsible for the behavioral influence of 5-HT acting agents in managing depressive illness
changes. Of course, as in the case of many studies involving (Millan, 2006; Pineyro and Blier, 1999). The newer generations
the knockout strategy, it is important to consider that of compounds have considerably reduced side effects
neurodevelopmental changes (either of a compensatory or associated with older antidepressant drugs, increasing their
maladaptive manner) may occur and hence the emergence of tolerability and compliance to their use. However, other things
later neurochemical alterations may stem from these being equal, the efficacy of the treatments has not improved
secondary factors, as opposed to the direct effects of the substantially (Millan, 2006). A characteristic feature of
gene manipulation. Furthermore, it seems that the back- antidepressant pharmacotherapy is that a substantial lag occurs
ground strain upon which a knockout is superimposed may before treatment efficacy is seen. Moreover, still only 60–65%
profoundly influence the behavioral outcomes observed of patients respond favorably to a given drug, remission is often
(Holmes et al., 2003b). Given that polygenic effects (and incomplete, and illness recurrence exceeds 50% (e.g., Fava
background emotionality) might underlie a given phenotype, et al., 2007; Millan, 2006). Clearly, the overall picture
any effects related to a given genetype, as in the case of the 5- regarding the effectiveness of current antidepressant medica-
HTT knockout, can be expected to vary with background tion has not been impressive. Thus, continued attention has
conditions. been devoted to the development of more efficacious treatment
Commensurate with the proposition that depression is not strategies, and identification of biomarkers that may be
subserved by a single neurotransmitter dysfunction, it has been informative in the diagnosis and development of tailored
reported that among 5-HTT knockouts chronic stressor treatments that might be most effective.
exposure provoked the desensitization of 5-HT1A receptors, Several approaches have been taken to identify the neuronal
whereas among glucocortioid receptor knockouts the chronic processes associated with depressive disorders in humans.
stressor did not impair 5-HT1A functioning (Lanfumey et al., These have involved pharmacological studies that influence
2000). Moreover, it was observed that in contrast to wild-type various neurotransmitter systems, imaging studies to assess
mice that exhibited reduced glucocorticoid receptor (GR) blood flow or receptor binding capacity, and analysis of
mRNA expression and binding, among GR knockouts a chronic components of neurotransmitter families in postmortem
mild stressor increased hippocampal, cortical and dorsal raphe (suicide vs. control) brain samples. As well, studies have been
GR mRNA and GR binding sites. Moreover, the desensitization conducted to identify gene polymorphisms in major depression.
of 5-HT1A DRN functioning evident in wild-type mice was not
apparent in the knockouts (Froger et al., 2004). Thus, it seems 5.3. Serotonin variations in depression and among
that genes for 5-HTT and glucocorticoid receptors may act depressed suicides
together to influence processes that might be related to
depression. Numerous advances have been made in the identification of
Gene–gene interactions have also been examined in studies processes that might subserve MDD, but identification of the
where the 5-HTT knockout was combined with either MAO-A, specific serotonergic factors associated with depression has, in
5-HT1B or BDNF knockouts (Hammack et al., 2003). Among several respects, yielded equivocal results. Postmortem
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 11

analyses and imaging studies revealed morphological and was related to hypersensitivity of 5-HT2A receptors (Rosel
functional disturbances within specific aspects of the PFC (e.g., et al., 2004).
dorsomedial and orbital cortex) as part of a wider system of As in the case of the animal models described earlier, there has
neuronal processes that may contribute to the emergence of been considerable focus on the potential involvement of 5-HT1A
MDD (Anguelova et al., 2003a,b; Drevets, 2000a,b, 2001; (auto)receptors in the DRN of depressed patients and depressed
Rajkowska, 2000a,b). However, as will be described shortly, suicides as these raphe changes potentially influence forebrain 5-
studies assessing brain neurochemical processes have not HT availability. It was reported that 5-HT1A receptors were
uniformly supported a relationship between suicide/depression reduced in the DRN (Stockmeier et al., 1998); however, such an
and 5-HT functioning (Anguelova et al., 2003a,b; Lowther outcome was not evident in a study reported by Arango et al.
et al., 1997; Meyer et al., 1999; Purselle and Nemeroff, 2003; (2001), although a reduced volume of the DRN was found. In
Rosel et al., 1997; Stockmeier, 2003). subsequent studies, however, it was observed that differences
Consistent with the involvement of 5-HT in depression, were apparent in the rostral and caudal portions of the dorsal
several investigators reported that 5-HT2A receptors were raphe, being elevated rostrally and diminished caudally (Boldrini
increased in the PFC of suicides relative to controls (Arora and et al., 2007). In our own studies we observed that 5-HT1A
Meltzer, 1989; Faludi et al., 2000; Hrdina et al., 1993; Lemonde receptor mRNA expression was unaltered in the raphe, although
et al., 2003; Mann et al., 1986; Stanley and Mann, 1983; variations of this receptor subtype were detected in other brain
Turecki et al., 1999). Likewise, mRNA expression of 5-HT2A regions (Anisman et al., in press, 2007a,b).
receptors, as well as the protein itself, was elevated in the PFC Although elevated 5-HT1A receptor binding (Arango et al.,
of teenaged suicides (Pandey et al., 2002). In contrast, other 1995) and mRNA expression (Escriba et al., 2004) was reported
studies failed to detect alterations in the expression of 5-HT2A in some aspects of the PFC and in the CA1 of the hippocampus,
receptors between suicides and controls in different portions of most studies indicated reduced 5-HT1A binding (Drevets et al.,
the PFC (Arranz et al., 1994; Cheetham et al., 1988; Crow et al., 1999, 2000, 2007; Lopez et al., 1998; Lowther et al., 1997;
1984; Lowther et al., 1994; Owen et al., 1983, 1986; Rosel Parsey et al., 2006a,b; Sargent et al., 2000; Stockmeier et al.,
et al., 2000; Stockmeier et al., 1997). Further, among depressed 1997), particularly in primary recurrent familial depression
patients that had attempted suicide, PET analysis indicated that (Drevets et al., 1999, 2007). However, analyses of tissues
5-HT2A receptor functioning was unaffected (Meyer et al., obtained from suicides and controls indicated that when factors
1999). Finally, DNA microarray analyses did not reveal such as age of subject, postmortem interval (from the time of
molecular genetic differences within the dorsolateral and suicide), and the time of tissue storage were controlled, the 5-
ventral PFC of suicides and controls (Sibille et al., 2004). HT1A differences between depressed suicides and controls were
The data derived from imaging studies have, unfortunately, not evident (Stockmeier, 2003; Stockmeier et al., 1997). To
been equally confusing. Several studies indicated reduced complicate matters further, Drevets et al. (1999) reported a
ligand binding in aspects of the PFC of depressed individuals decrease of 5-HT1A binding in bipolar patients, but a similar
(Attar-Levy et al., 1999; Biver et al., 1997; Yatham et al., 2000), effect was not apparent in unipolar major depressive patients,
whereas others indicated no differences in this respect (Meltzer unless a familial history of bipolar disorder existed. Moreover,
et al., 1999; Meyer et al., 1999, 2001a,b). In this regard, PET it was reported that the presence of elevated 5-HT1A binding
analysis also indicated that with symptom remission associated potential, coupled with the presence of a 5-HT1A C-1019G
with paroxetine treatment, the density of 5-HT2A receptors polymorphism, predicted poorer treatment response to anti-
increased in the frontal cortex, an effect not evident among depressants (Parsey et al., 2006a,b). It is interesting, however,
treatment non-responders (Zanardi et al., 2001). It is significant, that receptor binding differed between those patients who had
as well, that Meyer et al. (2003) were able to discern increased previously received antidepressant treatment and those who had
5-HT2A binding in the dorsolateral PFC among those patients not (Parsey et al., 2006b). Overall, it seems that MDD was
who scored highly on the dysfunctional attitudes scale. It was associated with diminished 5-HT1A binding in PFC and
suggested that this might have reflected elevated feelings of hippocampus, but these effects were influenced by genetic
pessimism and hopelessness, which may be a precursor to factors, history of depression and drug treatment (see Drevets
suicidal ideation or intent. Of importance, such findings suggest et al., 2007). It is noteworthy that many of these studies did not
that 5-HT receptor variations might not be a reflection of account for the individuals stressor history, which has been
depression/suicide per se, but rather may be tied to shown to interact with certain genetic polymorphisms (e.g.,
endophenotypes comprising the depressive syndrome. Caspi et al., 2003).
Suicide was also associated with fewer 5-HT2A binding sites Recent studies have implicated 5-HT1B receptor functioning
within the hippocampus, as well as lower binding affinity in MDD. In an animal model of depression, chronic stressors
(Cheetham et al., 1988; Mintun et al., 2004; Rosel et al., 1998, reduced 5-HT1B binding (Lopez et al., 1997; Svenningsson and
2000; Sheline et al., 2004), but once again, there have been Greengard, 2006), and the expression of 5-HT1B protein was
reports indicating that 5-HT2A was not altered in this region diminished in the brains of depressed individuals (Svennings-
(Gross-Isseroff et al., 1998; Lowther et al., 1994; Stockmeier son et al., 2006), as was mRNA coding for p11 (also known as
et al., 1997). Interestingly, however, inositol triphosphate (IP3), calpactin I light chain and S100A10), a protein involved in the
a second messenger of 5-HT, was elevated in the hippocampus functional expression of 5-HT1B receptors. Interestingly, in p11
of depressed suicides, suggesting that depression (or suicide) knockout mice, the behavioral profile observed was reminiscent
12 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

of that seen in human depression, supporting the link between Once again, unanimity has not been reached concerning 5-HTT
p11 and 5-HT1B. Our research in depressed suicides likewise changes in suicide brain. Several studies indicated that 5-HTT
indicated that the 5-HT1B and p11 mRNA expression was ligand binding was not altered among suicides (Arango et al.,
diminished among depressed suicides and this outcome was 2001; Bligh-Glover et al., 2000; Stockmeier et al., 1998),
fairly widespread, being evident in the frontopolar cortex, although a marked reduction was evident with respect to the
orbital frontal cortex, hippocampus and amygdala (Anisman number of neurons that expressed 5-HTT mRNA (Arango et al.,
et al., in press, 2007a,b). 2001). Although earlier studies with relatively poor ligands for
Beyond frank changes of 5-HT receptor mRNA expression, 5-HTT had revealed mixed results, several studies using better
elevations of RNA editing was reported at specific sites of the ligands (e.g., 3H-paroxetine and 3H-citalopram) revealed that 5-
5HT2C receptor in depressed suicides. The editing involved the HTT binding was, in fact, reduced in the PFC of depressed
conversion of adenosine to inositol, resulting in alterations of suicides (Arango et al., 1995; Leake et al., 1991; Mann et al.,
the amino acid sequence. It was proposed that this influenced 2000). Likewise, during a major depressive episode, 5-HTT
the efficiency of G-protein coupling, and hence second binding was diminished in the amygdala and midbrain (Parsey
messenger signaling disturbances among the depressed et al., 2006c) and in the brainstem (Malison et al., 1998).
suicides. In line with this, it has been shown that the SSRI, Paralleling their findings of 5-HT1A receptor presence, Meyer
fluoxetine, reduced the frequency of editing, suggesting that et al. (2004) reported that 5-HTT binding potential during the
RNA editing is regulated by 5-HT activity (Gurevich et al., course of a major depressive episode was elevated in various
2002; Schmauss, 2003). At the same time, however, it was brain regions (PFC, anterior cingulate, thalamus, caudate, and
reported that the reduced signaling associated with increased 5- putamen) among patients that displayed extremely negative
HT2C receptor protein levels in suicides was not paralleled by dysfunctional attitudes (it will be recalled that these attitudes
differences of mRNA expression (Pandey et al., 2006). may be aligned with later suicidality). At this juncture, it is
It is particularly interesting that in highly stressor reactive uncertain how the latter findings might map onto the data
BALB/cByJ mice (who also show an inclination toward anxiety reported by Malison et al. (1998) and Parsey et al. (2006c), but
and depressive-like behaviors; Anisman et al., 2007a) stressor they do suggest that individual difference factors might be an
exposure increased 5-HT2C editing, hence increasing the important element in defining whether 5-HTT alterations
mRNA pool that encodes diminished receptor functioning accompany depressive state.
(Alfonso et al., 2005). It was further reported that following
infant maternal separation, increased adult signs of depressive- 5.4. Genetic links between serotonin functioning and
like behaviors were apparent, coupled with increased editing of depression
5-HT2C pre-mRNA expression. In contrast, fluoxetine admi-
nistered during adolescence (but not in adulthood) antagonized Attempts to define the genotypic links to depression have
both the behavioral disturbances and the 5-HT2C editing. become a major focus of research in depression. As several
Unlike the effects of the SSRI, raising these mice in an enriched reviews of this literature have been provided in recent years (e.g.,
environment (following weaning) did not affect 5-HT2C Arango et al., 2003; Serretti et al., 2005), only a capsulated
editing, but the behavioral disturbances were attenuated review of this work is covered here. Briefly, depression was
(Bhansali et al., 2007). Inasmuch as the enriched environment associated with 5HT2A receptor and tryptophan hydroxylase
influenced the G alpha q subunit of the G-protein that couples to gene polymorphisms (Faludi et al., 2000), increased frequency of
5-HT2A/2C receptors, whereas fluoxetine did not have such the long (L) allele of the 5-HT transporter gene (Du et al., 1999),
effects, these investigators suggested that second messenger and a polymorphism was similarly reported with respect to the 5-
systems adjusted to the effects associated with persistent 5- HTT promotor region (Serretti et al., 2005). It is interesting that
HT2C pre-mRNA editing, thereby accounting for the differ- although postmortem analyses indicated that 5-HTT binding was
ential effects of the drug and experiential treatments. When lower in the orbital frontal cortex of suicides (and throughout the
these data are considered in the context of the 5-HT2C editing PFC among those individuals that had a history of major
associated with suicide, two important issues become apparent. depressive disorder), the 5-HTT gene promotor polymorphism
First, early-life events may have their depressogenic effects (5-HTTLPR) was related to depression, but not suicide, and was
through actions on the 5-HT2C receptor, particularly given a not related to 5-HTT binding (Arango et al., 2003; Mann et al.,
genetic disposition toward depressive-like states (recall that 2000). In addition to the potential contribution of the 5-HTT
these studies were conducted in the highly stress-reactive promoter, other candidate genes have been reported to be related
BALB/c mouse). Moreover, despite the profound effects that to depression. These have included those for tryptophan
early life experiences are known to have, these effects are hydroxylase and G-protein beta 3 (see Serretti et al., 2005).
reversible by antidepressants (and to some extent by environ- As well, it was demonstrated that a repressor of the 5-HT1A
mental enrichment) applied during later development. Essen- receptor gene promoter in raphe cells (nuclear deformed
tially, despite a genetic predisposition and early life stressful epidermal auto-regulatory factor (DEAF 1))/suppression)
experiences, the disposition toward depression is not necessa- was rendered inactive by a promoter polymorphism. In effect,
rily fixed. this loss of repression of the 5-HT1A autoreceptor expression
In addition to the receptor changes, considerable interest has potentially reduces 5-HT neurotransmission, hence predisposing
focused on variations of 5-HTT in mediating depressive illness. to depression and suicide (Lemonde et al., 2003).
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 13

It is widely believed that genetic factors alone may not be depressive disorder. Finally, the data indicative of interactions
sufficient to promote depression. Instead genetic contributions between genetic factors (e.g., those related to 5-HTT variations,
might dispose individuals to depression, and expression of the for instance) and stressful experiences, and the data indicating
illness would most likely occur in the face of certain the 5-HT receptor changes associated with suicide may be most
environmental or experiential factors. Consistent with this view, apparent given particular behavioral features attest to the
Caspi et al. (2003) reported that depressive symptoms, and the importance of evaluating the neurochemical substrates of MDD
incidence of depression and suicidality, were more frequent in a broad context.
among individuals with one or two copies of the short allele of the
5-HTT promoter relative to individuals that were homozygous 6. Corticotropin releasing hormone (CRH)
for the long allele. However, the expression of these depressive
symptoms was contingent on individuals having experienced Corticotropin releasing hormone, a primary regulator of the
major stressful life events. It was subsequently demonstrated HPA system, is released at hypothalamic and amygdaloid sites
(Kendler et al., 2005) that individuals with two short alleles for 5- in response to stressors (Herman and Cullinan, 1997; Owens
HTT exhibited greater depressive vulnerability than those with et al., 1990). The transcription of CRH genes within the
one or two large alleles. These individuals appeared more parvocellular portion of the hypothalamus may be regulated
sensitive to the impact of relatively mild life stressors, which through NE processes, whereas arginine vasopressin (AVP)
might thus have promoted their increased depressive symptoms. transcription, a second hypothalamic peptide, was seemingly
Furthermore, among individuals with the two short alleles, less affected by the same NE manipulations (Cole and
tryptophan depletion was likely to promote depressive symp- Sawchenko, 2002; Itoi et al., 1999). As well, it seems that
toms, irrespective of family history of depression (Neumeister other peptides, such as bombesin, may stimulate CRH release
et al., 2002). In accordance with these findings, multimodal (Kent et al., 2001a,b) and GABAA has an inhibitory effect in
magnetic resonance imaging revealed that carriers of the short this regard (Cole and Sawchenko, 2002).
allele had tonically activated amygdala and hippocampal
functioning, but diminished reactivity to negative stimuli (Canli 6.1. Stressor-induced CRH functioning: animal studies
et al., 2006). These investigators suggested that this would
translate as a chronic state of distress, characterized by Ordinarily, acute stressors promote the release of PVN
hypervigilance, threat, or rumination, hence disposing indivi- peptides (CRH and AVP), thus promoting ACTH and adrenal
duals to stress-related disorders. Findings such as these suggest glucocorticoid secretion (Lightman, 1994; Lightman et al.,
that despite the mixed results accrued from postmortem and 2000; Plotsky, 1991; Rivier and Vale, 1985; Windle et al.,
imaging studies, 5-HTT likely may act as an important factor in 1998). These effects were associated with increased anxiety and
depression, but more detailed analyses of gene  environment the initial HPA response was generally not influenced by
interactions are necessary to fully appreciate the contribution of stressor controllability. Essentially, when an animal is first
both these processes in the evolution and maintenance of the confronted by a novel stressor, it is uncertain whether this
depressive phenotype. stressor will be brief or prolonged, controllable or uncontrol-
Overall, it seems that although imaging and postmortem lable, and predictable or unpredictable. Thus, it would be
analyses have provided considerable data concerning the biologically advantageous to mount a strong defensive response
neuronal circuitry that may be associated with depression, the to maintain well-being. As information is acquired about the
inconsistencies that exist preclude definitive conclusions. In an stressor (e.g., its controllability), then it might be appropriate
extensive and insightful review, Stockmeier (2003) was able to for neuroendocrine responses to taper off. Thus, it is not
accommodate some of these inconsistencies, based on among surprising that fast acting neuroendocrine factors would not be
other things, presence of antidepressants (or time of washout markedly influenced by stressor controllability, although one
periods) before PET analyses, differences in outcomes as a might imagine that with sustained stressor experiences,
function of life-time depression, and particular aspects of the differences between the HPA effects of controllable and
brain examined (e.g., different portions of the prefrontal uncontrollable stressors would emerge.
cortex). Moreover, if one considers that multiple neurotrans- In addition to the variations of hypothalamic CRH that
mitters may be associated with depression (and these may differ accompany acute stressors, such treatments increase CRH
to a certain extent in depressed suicides), it is possible that some mRNA expression and increase in vivo CRH release at the
of the between-study inconsistencies reflect the complex central amygdala (CeA) (Davis, 1992; Lee and Davis, 1997;
interactions that may occur between transmitter systems. The Makino et al., 1999; Merali et al., 1998; Pich et al., 1995),
very fact that patients present with varied symptom profiles, possibly through actions of 5-HT and NE processes (Pacak
and are amenable to different drug treatments, would seem to et al., 1995a,b; Stutzmann et al., 1998). It is interesting that
provide prima facie evidence consistent with this proposition. unlike the suppression of hypothalamic CRH release elicited by
For instance, reports indicating that 5-HT receptor subtypes, cortisol, in limbic regions, and specifically at the CeA and bed
such as 5-HT2A and 5-HT2C, may be relevant to anxiety (Merali nucleus of the stria terminalis, cortisol acts to increase CRH
et al., 2006a; Millan, 2005; Weisstaub et al., 2006) raise the expression, as well as GABAA neuronal functioning, that may
possibility that the observed 5-HT receptor changes are actually act to provoke anxiety (Duvarci and Pare, 2007; Schulkin,
a reflection of anxiety that is often a comorbid feature of major 2006; Schulkin et al., 1998; Shepard et al., 2005).
14 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Fear-eliciting stimuli markedly increase CRH expression necessary to consider state and trait anxiety, as well as subtypes
within the CeA and the basolateral amygdala (BSA) (LeDoux, of anxiety disorders. Furthermore, anxiety may be tied to
2000), which may be fundamental for the acquisition (rather significant stimuli or may be stimulus-independent. It is
than the expression) of a fear response (Davis, 1992; Gray, certainly possible that mechanisms other than, or in addition to,
1990; LaBar et al., 1998; LeDoux, 2000). In contrast, diffuse CRH functioning may contribute to these different phenotypes.
stimuli that promote general anxiety elicit greater CRH Less information is available concerning the involvement of
variations at the bed nucleus of the stria terminalis (BNST) CRH2 receptor functioning in anxiety, although interest in this
(Davis, 1992). As well, it appeared that variations of in vivo receptor subtype has been increasing (Takahashi, 2002). Initial
CRH release at the CeA, and the effectiveness of CRH studies had indicated that the contribution of CRH2 was limited,
antagonists in attenuating anxiety, were dependent on the as mice engineered for a deficiency of urocortin expression (a
nature of the stressor to which animals were exposed (Merali peptide that acts as the endogenous ligand for CRH2 receptors)
et al., 1998, 2004a,b). As alluded to earlier, although a variety did not display altered stressor-provoked anxiety (Wang and
of stressors provoked CRH release from amygdala neurons, Shuaib, 2002). In contrast, however, if the CRH2 receptor was
anxiety elicited by naturalistic stressors (e.g., predators), which expressly knocked out, elevated anxiety-like behaviors were
were proposed to involve pre-wired neural circuits, was not apparent (Bale et al., 2000; Kishimoto et al., 2000), but this
altered by manipulations (CRH antagonists) that ordinarily effect was most notable among males (Bale and Vale, 2003;
attenuate the response to neurogenic stressors or learned fear Bale et al., 2002).
cues. Further, although psychogenic and systemic stressors Given the independent effects of the CRH1 and CRH2
have many similar effects, their actions on amygdala manipulations, Preil et al. (2001) assessed corticosterone and
functioning are not identical. Whereas systemic stressors ACTH under basal conditions and in response to social defeat
markedly impacted CeA activity, psychogenic stressors had among double and single knockouts of the CRH1 and CRH2
more potent effects on the medial amygdala, which may genes. Overall, these studies indicated that in both the CRH1
influence HPA activity (Dayas et al., 1999, 2001a,b). knockouts and the double knockouts the corticosterone and
ACTH responses were abrogated, and having the presence of
6.2. CRH and CRH receptors the CRH2 did not compensate for the absence of the CRH1
gene, attesting to the primacy of the type 1 receptor. Yet, it was
Consistent with the assumption that specific CRH receptors observed that among the female CRH1 knockouts, a modest
contribute to anxiety and depression, it was reported that in corticosterone rise was apparent following stressor exposure,
chronically stressed animals, a condition that may be more which was not apparent in males. Further to this point, among
similar to the sustained distress of severe depression, a decrease double mutants (combined CRH1 and CRH2 knockouts),
of CRH1 receptor expression was provoked within the PFC females displayed reduced anxiety, whereas males exhibited
(Iredale et al., 1996). Predictably, administration of the CRH higher levels of anxiety. Moreover, consistent with reports
antagonist CP-154,526 attenuated the behavioral disturbance suggesting a moderating role for experiential factors in
associated with uncontrollable shock (in a learned helplessness determining anxiety responses, the effects of the double
paradigm) (Mansbach et al., 1997). Moreover, studies of knockout varied as a function of maternal factors (Bale et al.,
genetically engineered mice with elevated or reduced CRH 2002). That is, elevated anxiety was apparent among CRH2
receptors revealed, for the most part, predictable alterations of knockouts raised by a similar dam. However, if a pup was born
anxiety and depressive-like behavior (Keck and Muller, 2005), to a heterozygous or mutant dam, then greater anxiety was
and have implicated glucocorticoid receptor (GR) signaling and evident regardless of the pup’s genotype (Bale et al., 2002).
stressor responses in the emergence of depressive symptoms Thus, it seems that both CRH1 and CRH2 receptors may
(Keck and Muller, 2005). In general, mice that overproduced influence anxiety; however, early experiences, and particularly
CRH had higher levels of anxiety, which appeared to be maternal factors (related to either pre- or post-natal events)
dependent on the specific behavioral test used (van Gaalen appear to moderate the actions of the gene on anxiety behaviors.
et al., 2002a,b). Predictably, in CRH1 receptor knockout mice,
anxiety levels were diminished (Contarino et al., 1999; Smith 6.3. CRH in depression/suicide
et al., 1998; Timpl et al., 1998) depending on the basal levels of
emotionality ordinarily apparent (Keck et al., 2001). Further- It has long been known that HPA functioning is altered
more, among conditional knockout mice (Crhr1(loxP/loxP)- among major depressive patients, especially those with
Camk2a-cre), in which CRH1 receptors were inactivated melancholic features. Among other things, MDD has been
postnatally in the anterior forebrain and in limbic brain regions associated with elevated cortisol, early escape from dexa-
(while leaving pituitary receptors unaffected), anxiety-like methasone-induced cortisol suppression, and a blunted ACTH
behaviors were reduced. Conversely, among mice with elevated response to CRH or dexamethasone/CRH challenge (Heuser
CRH1 receptors, hypersensitivity to stressors was apparent et al., 1994; Nemeroff, 1996; Nemeroff and Vale, 2005). As
(Muller et al., 2003). Thus, it would seem that the gene well, in suicides that had been diagnosed with recurrent
subserving this receptor subtype in limbic forebrain regions depressive episodes, MR mRNA expression was diminished in
may be pertinent to the development of anxiety states. the CA3 portion of the hippocampus, relative to controls
However, in relating such findings to human pathology, it is (Lopez et al., 1998). Although it had initially been thought that
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 15

cortisol changes might simply be a marker of distress or CRH. However, it seemed that unlike the linkage between CRH
depression, the view that it plays a provocative role in this and 5-HT, mixed CRH1/CRH2 antagonists did not modify the
regard has received increasing attention. Indeed, several studies acute or chronic stressor effects on NE and DA activity within
indicated that GR antagonists diminished the symptoms of the PFC (Murphy et al., 2003).
depression (Belanoff et al., 2001; Gold et al., 2002; Murphy, Consistent with the view that CRH may be related to
1997), as did treatments that inhibit cortisol production depression, CRH concentrations in cerebrospinal fluid (CSF)
(Murphy, 1997). Moreover, patients treated with glucocorti- were elevated among depressed patients (Arato et al., 1989;
coids frequently experience pronounced mood disturbances Arborelius et al., 1999; Arranz et al., 1992; Heuser et al., 1998;
(Mitchell and O’Keane, 1998). It is significant, as well, that Nemeroff et al., 1984; Wong et al., 2000), which normalized
among probands of patients with major depression, HPA with pharmacotherapy or electroconvulsive therapy (De Bellis
abnormalities were present prior to the presentation of clinical et al., 1993; Heuser et al., 1998; Kling et al., 1994; Nemeroff
symptoms (Holsboer, 2000). et al., 1991). Although there have also been reports indicating a
The perspective that HPA dysfunctions are not simply a failure to detect elevated CRH within CSF (Kling et al., 1991;
consequence of depression/stress or bystanders of the illness, Pitts et al., 1995; Roy et al., 1987), it seems that CRH CSF
but are aetiologically significant, has gained appreciable levels were elevated in those patients who were dexamethasone
support. At the same time, studies showing that corticoid non-suppressors (Roy et al., 1987).
inhibitors diminish symptoms of depression are inconsistent Increasing evidence has suggested that CRH hyper-
with the perspective that hypothalamic CRH elevations are functioning within hypothalamic and extra-hypothalamic sites
responsible for the depressive state (that is, corticoid inhibition, (locus coeruleus, amygdala, hippocampus, nucleus accum-
through attenuation of the negative feedback ought to promote bens, PFC) is associated with depression, at least that of a
elevated CRH and provoke greater symptoms). Nevertheless, it relatively severe (melancholic) nature, or in which an
has been suggested that the balance between GR and abnormal dexamethasone response was evident (Holsboer,
mineralocorticoid (MR) receptors may be a pivotal factor in 2003; Nemeroff, 1996; Reul and Holsboer, 2002). Although
determining stress reactions and depressive outcomes (de CRH is widely distributed within the brain, the data concerning
Kloet, 2003; Hasler et al., 2004). Specifically, it is thought that concentration of this peptide or its receptors among depressed
several rapid acting processes (catecholamines, CRH/AVP, patients is still limited. Nevertheless, it was reported that CRH
neurosteroids and corticosterone) are engaged in response to mRNA expression and/or CRH immunoreactivity was
stressors. The rapid initiation of corticosterone influences MRs, increased in hypothalamic nuclei, locus coeruleus, PFC and
which may affect defense-related behaviors, including vigi- raphe nuclei of depressed suicides (Austin et al., 2003; Bissette
lance, alertness, arousal, and attention. This is followed by et al., 2003; Merali et al., 2004b; Raadsheer et al., 1994, 1995)
slower changes involving GRs, which have the effect of as was CRH mRNA expression in the PVN (Raadsheer et al.,
normalizing steroid levels (promoting homeostasis) and in 1995).
information storage (or sensitization of neuronal processes) Despite the widespread assumption that disturbances of
concerning potential stressors (de Kloet et al., 2005). Further, frontal cortical processes may be involved in depressive
there is reason to believe that MR activity influences the illnesses, data concerning CRH-related changes within this
functioning of CRH1 receptors (de Kloet, 2003) and may be an region have been sparse and inconsistent. Whereas Nemeroff
essential component of 5-HT1A down-regulation associated et al. (1988) found reduced CRH-binding sites within the
with chronic stressor exposure (Kuroda et al., 1994), whereas 5- frontal cortex of depressed suicides, neither Hucks et al. (1997)
HT2A up-regulation induced by a stressor involves GR nor Leake et al. (1990) observed such effects. Our own findings,
functioning (Karten et al., 1999). Given the relationship however, indicated that CRH levels and receptor mRNA
between CRH and monoamine functioning (Ruggiero et al., expression were altered in selective aspects of the frontal cortex
1991), these findings are consistent with the view that (Merali et al., 2004b). The CRH levels were elevated in the
depression involves the disruption of the interface between right frontopolar (FPC) and dorsomedial prefrontal cortex (only
CRH and 5-HT circuits (Claes, 2004; Price and Lucki, 2001). In tissue from the right side was assessed) among suicides, and
effect, through such cascading processes, CRH activity may mRNA expression of CRH1, but not CRH2, was reduced in the
come to promote or exacerbate depressive symptoms. FPC. Inasmuch as CRH1 desensitization develops upon
It has been suggested that CRH may regulate a subpopula- exposure to physiological concentrations of CRH (Aguilera
tion of raphe neurons, hence influencing PFC 5-HT release et al., 2001), and reduced CRH1 expression occurs after several
(Kirby et al., 2000; Valentino et al., 2001). Just as CRH hours of exposing anterior pituitary cells to CRH (Pozzoli et al.,
administered to the DRN influenced forebrain 5-HT release 1996), it is possible that the diminished receptor expression in
(Price and Lucki, 2001), chronic treatment with the CRH1 suicide brains reflected an outcome secondary to a sustained
antagonist, NBI 30775 (also known as R121919), which has increase of CRH activity. Thus, these data support the
antidepressant actions, influenced hippocampal 5-HT function- involvement of CRH1 receptor in depression/suicide, although
ing (Oshima et al., 2003). Administration of a CRH2 antagonist it is certainly possible that that the CRH1 receptor variations
into the DRN also attenuated the effects of inescapable shock were related to the perceived stress that promoted depression
(Hammack et al., 2002, 2003), hence implicating forebrain 5- (or the stress associated with depression), rather than being
HT involvement in mediating the depressogenic actions of directly related to the affective state.
16 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

6.4. CRH has multiple actions (hyperphagia, significant weight gain, hypersomnia), extreme
fatigue and persistent rejection sensitivity that is not restricted to
Clearly, multiple neurochemical changes are engendered by episodes of depression (Asnis et al., 1995; Quitkin et al., 1991;
stressors, which might ultimately promote depressive symptoms. Stewart et al., 1993). In atypical, unlike typical MDD, HPA
Although CRH might be an especially important factor, it is activity may be diminished as reflected by reduced CRH in CSF
likely that CRH activation represents only one component in a (Geracioti et al., 1997) and lower plasma ACTH (Gold and
sequence of changes that determine behavioral outcomes. It Chrousos, 2002). Furthermore, it seems that elevated CRH may
appears that hippocampal 5-HT release induced by a stressor be less prominent in atypical than in typical major depression
involves a CRH receptor-dependent mechanism (Linthorst et al., (Nemeroff, 1996), and desipramine-elicited plasma cortisol
2002). In fact, protracted intracerebroventricular CRH infusion secretion was greater in atypical patients, suggesting a less
blunted hippocampal 5-HT release ordinarily elicited by LPS dysfunctional noradrenergic system (Asnis et al., 1995; McGinn
(Linthorst et al., 1997). Likewise, paralleling the effects of et al., 1996). From the treatment perspective, it was reported that
stressors, it was shown that CRH infusion increased locus atypical patients, and particularly females (Davidson and Pelton,
coeruleus NE neuronal activity, and conversely, CRH antagonists 1986), responded preferentially to monoamine oxidase inhibitor
attenuated such effects (Emoto et al., 1993a,b,c; Finlay et al., (MAOIs) relative to tricyclic agents (Quitkin et al., 1988;
1997; Li et al., 1998; Shimizu et al., 1994; Valentino et al., 2001). McGrath et al., 1992; Thase et al., 1991). Such findings support
Moreover, as indicated earlier, in addition to attenuating stressor- the importance of differentiating the features of depression
provoked ACTH and corticosterone increases (Ohata et al., (endophenotyping) in evaluating the processes associated with
2002), CRH antagonists also reduced in vivo hippocampal 5-HT illness, and also have implications for the efficacy of treatment
release (Linthorst et al., 2002). Further to this point, among mice strategies.
that overproduce CRH, pharmacological challenges provoked Finally, although CRH is usually considered in the context of
the desensitization of post-synaptic, but not pre-synaptic, 5- stressful encounters, it is clear that this peptide is also released
HT1A receptors (van Gaalen et al., 2002a,b), and it was similarly in response to spontaneous food ingestion (Merali et al., 1998,
reported that HPA functioning and forebrain 5-HT release were 2002b; Plamondon and Merali, 1994, 1998; Plamondon et al.,
altered in CRH1 knockouts (Penalva et al., 2002). There has been 1998). Indeed, both appetitive and rewarding stimuli can
limited work concerning the involvement of specific types of activate the HPA axis, although it is uncertain whether such an
CRH receptors in provoking 5-HT variations, but it has been outcome reflects a response to arousal and/or anxiety or
reported that DRN infusion of CRH or the CRH2 agonist whether food ingestion serves as a coping response to stressful
urocortin 2 increased 5-HT release at the basolateral amygdala, experiences (Dallman et al., 2003; Piazza and Le Moal, 1997).
and these effects were attenuated by selective CRH1 or CRH2 Consistent with the dual effects of CRH in both anxiety and
antagonists (Amat et al., 2004). appetitive processes, central CRH administration provokes both
Several further issues ought to be introduced before ending anxiogenic behaviors and promotes suppression of food intake
this section. First, the fact that clinical depression occurs more (Heinrichs et al., 1992; Heinrichs and Richard, 1999; Hotta
frequently in females than in males begs the question as to et al., 1999; Momose et al., 1999). Conversely, pretreatment
whether females are more vulnerable to glucocorticoid with a CRH antagonist (e.g., a-helical CRH) attenuated the
disturbances. This issue is somewhat removed from the gist anorexia induced by a stressor and also augmented
of the present review, and a detailed examination of this the hyperphagia induced by neuropeptide Y and reduced the
essential issue is provided by Swaab et al. (2005). Suffice it at latency to initiate eating elicited by tail pinch (Heinrichs et al.,
this juncture that cross-talk occurs between the HPA and the 1992). From an ethological perspective, the dual effects of CRH
hypothalamic–pituitary–gonadal (HPG) axis; alterations of (eliciting anxiety and inhibiting eating) are understandable, as
corticoid activity influences HPG functioning, and conversely feeding is incompatible with defensive behaviors, and
estrogen activity will affect HPA functioning, including effects suppression of appetitive behaviors would reflect a highly
on the CRH mRNA expression (Bao et al., 2007; Vamvako- adaptive response in the context of situations that represent
poulos and Chrousos, 1993). Moreover, estrogen is coexpressed danger to the organism. In effect, it would be practically
in approximately 40% of CRH neurons among depressed important for systems subserving anxiety or stressor reactivity
patients (Bao et al., 2007). In fact, the suggestion was offered to be linked, directly or indirectly, with those subserving
that sex steroids might operate to program CRH functioning suppression of consummatory behavior. From this perspective,
(just as early life stressors might be effective in this respect; it would be inappropriate to consider CRH to be exclusively an
Meaney, 2001) so that vulnerability to HPA disturbances, and anxiety/stress peptide. Instead, it may be that the function of
hence depression, would occur upon subsequent stressor CRH, at least at certain brain sites, might be one of making
experiences (Bao et al., 2007; Swaab et al., 2005). salient cues associated with motivational processes or increas-
The second point concerns the fact that subtypes of depression ing the gain in response to aversive and/or rewarding stimuli.
exist that may involve different neurochemical processes, just as
they may comprise distinct symptom profiles. For instance, in 7. Arginine vasopressin (AVP)
contrast to the poor mood, anhedonia and neurovegetative
features of typical MDD, atypical depression comprises mood Parvocellular neurons of the PVN secrete AVP and CRH
reactivity, coupled with reversed neurovegetative symptoms from axon terminals located at the external zone of the median
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 17

eminence. Although AVP itself is a relatively weak secreta- suprachiasmatic nucleus, a site of one the pacemakers for
gogue of pituitary ACTH, it potentiates the effects of CRH in circadian rhythms, the number of AVP immunoreactive
promoting pituitary ACTH release (Antoni, 1993). As well, neurons was increased, but AVP mRNA expression was
magnocellular neurons in the PVN and supraoptic nucleus reduced (Zhou et al., 2001). Thus, the circadian disturbances
project to the pituitary, releasing AVP into the blood in response frequently seen in depression may be related to AVP
to osmotic stimuli (Swaab, 2004). Given the effect of AVP on dysfunction.
HPA functioning, the view was entertained that together with
other peptides, AVP might contribute to anxiety and depression, 7.2. Arginine vasopressin and CRH synergy
and in modulation of the stress response (Landgraf, 2006).
The potential importance of AVP in subserving depression
7.1. Arginine vasopressin in relation to depression comes from its synergistic actions with CRH, and the fact that
these peptides are highly co-localized within the hypothalamus.
Elevated plasma AVP concentrations have been noted in Tilders and his associates (Bartanusz et al., 1993; Schmidt
melancholic patients (van Londen et al., 1997), as well as in et al., 1995, 1996; Tilders et al., 1993) indicated that
patients diagnosed with anxious-retarded depression (motiva- paraventricular neurosecretory neurons have the capacity for
tional inhibition) (Goekoop et al., 2006; de Winter et al., 2003). phenotypic plasticity. In particular, the CRH containing
Interestingly, anxiety and motivational inhibition were highly terminals in the external zone of the median eminence
correlated among patients with high plasma AVP levels, and (originating from the PVN) frequently exhibit AVP co-
were elevated in suicidal depressed patients (Inder et al., 1997). expression. With the passage of time following a stressor
It has also been proposed that AVP may be important in experience or in response to chronic stressor exposure,
mediating the psychomotor retardation and in affecting increased co-storage of CRH and AVP occurs within CRH
memory processes in depressed patients, possibly by altering terminals at this site (Bartanusz et al., 1993; Ma et al., 1999;
arousal and attention (van Londen et al., 1998a,b). Together, Tilders et al., 1993), and increased AVP mRNA expression in
these data support the contention that the inter-relationships the parvocellular portion of the PVN accompanied the elevated
between subtypes of depression and HPA dysfunction may co-expression (Bartanusz et al., 1993).
involve an AVP component. The shift in neuropeptide composition of CRH terminals in
It is interesting that following a challenge with desmopressin the median eminence was not stressor-specific and similar
(an analog of vasopressin that stimulates the V3 receptor), the increases were reported after immobilization or social defeat
ACTH and cortisol responses were appreciably greater in MDD stressors (Bartanusz et al., 1993; de Goeij et al., 1991; Keeney
patients than in controls, suggesting that the V3 receptor was et al., 2006) and after adrenalectomy (Bertini and Kiss, 1991;
more reactive in the depressed condition (Dinan et al., 2004). Sawchenko, 1987). This effect became progressively more
Little information is available concerning molecular biological pronounced over time, peaking during the initial 2–4 weeks
aspects regarding AVP functioning in depression. Nevertheless, after treatment (Schmidt et al., 1995). As CRH and AVP
it may be significant that the haplotype defined by A-T-C-A-G synergistically stimulate ACTH secretion from the anterior
for the AVP1b receptor was under-represented in depressed pituitary, it is significant that when these animals were later
relative to control patients, possibly indicating that this exposed to a neurogenic stressor (footshock), plasma ACTH
haplotype ordinarily acts in a protective capacity insofar as and corticosterone release were augmented. It seems that
this pathology is concerned (van West et al., 2004). stressors increased the availability of secretagogues, such that
In addition to peripheral AVP correlates of depression, later challenges augmented glucocorticoid secretion. More-
immunoreactive AVP and oxytocin (OXT) in the PVN of major over, the coexpression of these sectretagogues was long
depressive and bipolar patients were elevated relative to lasting, thus favoring persistent sensitization to these
controls (although the investigators indicated that there was no treatments.
difference between uni- and bipolar individuals, the sample size It will be recalled that sensitization of neuronal processes
of each was likely insufficient to make adequate appraisals of have been implicated as an important feature in promoting
potential differences that might exist) (Purba et al., 1996). It depressive-like states. According to Post (1992), with each
was similarly reported that AVP mRNA expression was stressor experience and with each successive episode of
elevated in the supraoptic nucleus and PVN in a postmortem depression, neuronal sensitization becomes more pronounced,
analysis of depressed individuals relative to nondepressed and hence the stressor severity necessary to elicit the
controls (Meynen et al., 2006). Importantly, the elevation was neurochemical changes (and hence the depressive episode)
only apparent in the six of nine suicides that had been classified becomes progressively smaller. From this perspective, it could
as being of the melancholic subtype. Paralleling these findings, be argued that stressful events, by virtue of their effects on AVP
immunoreactive AVP was elevated in the PVN and locus co-expression within CRH neurons, may increase vulnerability
coeruleus of suicides. Moreover, although low levels of AVP to depressive episodes. Moreover, as even mild stressors may
are generally found in frontal cortical areas, immunoreactive ultimately come to provoke exaggerated ACTH and cortisol
AVP was elevated in the dorsomedial PFC (but not in the release, the possibility exists that given a sufficiently sensitized
frontopolar cortex), but was reduced in the dorsal vagal neurochemical system, even relatively innocuous stressor
complex (Merali et al., 2006b). It is interesting that within the experiences may promote depressive periods.
18 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Parenthetically, it has also been argued that chronic 8.1. Bombesin and CRH interactions
depression (e.g., dysthymia) may be related to elevated levels
of AVP within CRH neurons, and likewise the high rates of The bombesin (BB) family of peptides was initially
recurrence of depression may be related to the sensitization of implicated in the mediation of peripheral satiety signals (Gibbs
neuronal processes involving CRH/AVP co-expression (Grif- et al., 1979; Kulkosky et al., 1982; Ladenheim et al., 1992;
fiths et al., 2000). It was suggested that in the case of dysthymia, Merali et al., 1999; Moody and Merali, 2004), but members of
AVP was persistently increased within CRH neurons (essen- this peptide family have since been shown to moderate the
tially reflecting a change of basal levels that occurs with a long- stress response (Merali et al., 1998, 2002b), possibly through
standing illness), so that even minor day-to-day annoyances their effects on CRH functioning. Importantly, just as BB-
would trigger excessive CRH/AVP release. This, in turn, would related peptides are influenced by feeding behaviors, it has been
favor the presentation of dysphoric symptoms, and might even shown that stressors altered endogenous levels of hypothalamic
be a factor responsible for triggering double depression (i.e., BB-related peptides and the density of BB binding sites at the
where major depression is superimposed on a dysthymic state nucleus tractus solitarius, arcuate nucleus and PVN (Kent et al.,
and is typically more resistant to effective treatment) (Griffiths 1998). Moreover, in vivo analyses revealed that stressors
et al., 2000). reliably elicited the release of BB-like peptides from the central
amygdala (Merali et al., 1998, 2004a). It is of significance that
8. Bombesin (neuromedin B and gastrin-releasing administration of these peptides promote HPA activation
peptide) (Garrido et al., 1998; Kent et al., 2001a,b; Malendowicz et al.,
1995; Merali et al., 2002a, 2004a; Olsen et al., 1992), and the
Bombesin (BB) is an amphibian peptide that is pharma- variations of ACTH elicited by BB could be attenuated by the
cologically active in mammals (Battey and Wada, 1991), and CRH antagonist, a-helical CRH (Kent et al., 2001a,b). Thus, it
its mammalian analogues include the endogenous peptides, appears that BB-related peptides and CRH processes are
neuromedin B (NMB) and gastrin-releasing peptide (GRP). intertwined in the modulation of stress responses.
Whereas NMB binds preferentially to BB1 receptors, GRP
(GRP1–27) and neuromedin C (NMC (GRP18–27)) have a 8.2. Bombesin and 5-HT interactions
greater affinity for BB2 receptors. Both GRP and NMB are
present in each of the major nodes of the HPA axis (Domin It will be recalled that anxiety has been associated with
et al., 1988; Houben et al., 1993; Minamino et al., 1984; Moody altered 5-HT functioning, and particularly 5-HT1A and/or 5-
and Merali, 2004) and have been identified within several HT2C receptor changes (Alves et al., 2004; Gordon and Hen,
limbic regions (Merali et al., 2006b). These peptides and their 2004; Lesch et al., 2003; Millan, 2005). It was argued that
receptors are concentrated in brain regions associated with bombesin-related peptides may, through their actions on 5-HT
stress and appetitive responses, including hypothalamic nuclei and CRH, play a fundamental role in promoting anxiety (Merali
(Battey and Wada, 1991; Ladenheim et al., 1992; Pinnock et al., 2006a). It was observed that in addition to the HPA
et al., 1994; Wada et al., 1990), the nucleus of the solitary tract changes, NMB increased the firing rate of 5-HT cells in the
(NTS), the BNST, several amygdaloid nuclei, and the DRN (Pinnock and Woodruff, 1991), and, as expected, dorsal
hippocampus (Chronwall et al., 1985; Moody et al., 1981; raphe microinjection of NMB provoked the release of 5-HT at
Shumyatsky et al., 2002). In humans, NMB has also been the ventral hippocampus (terminal region). Conversely,
identified in several brain regions, being high in the dorsal microinfusion of the BB1/BB2 antagonist caused a diminution
vagal complex, median eminence and locus coeruleus, of extracellular 5-HT levels at this site (Merali et al., 2006a).
whereas the concentrations of GRP were most prominent in Thus, it seems that BB1 receptors (stimulated by NMB) are
the PVN, median eminence and frontopolar cortex (Merali located on the 5-HT cell bodies of neurons that project to the
et al., 2006b). ventral hippocampus. Localized infusion of the NMB
A strong case has been made that bombesin-like peptides antagonist PD 176252 into the DRN attenuated anxiety in a
contribute to the emergence of psychiatric and neurological social interaction test, and provoked a marked reduction of 5-
disorders (Roesler et al., 2006). These investigators indicated HT release (60% of baseline release) from the ventral
that GRP and its receptor (GRPR) were altered in the CNS of hippocampus (Merali et al., 2006a). As the activity of forebrain
patients with schizophrenia and Parkinson’s disease, and serotonergic neurons appear to be modulated by the BB1
disturbed GRPR-induced cellular calcium signaling was receptor at the dorsal raphe, it is possible that anxiety/
observed in fibroblasts from patients with Alzheimer’s disease. depression may be associated with alterations in the functioning
Pharmacological and genetic studies in rodents have shown that of these receptors.
GRPRs are present in brain areas such as the dorsal
hippocampus and amygdala, which are likely involved in 8.3. Bombesin and anxiety
anxiety, schizophrenia, depression, and dementia. Coupled
with animal studies supporting a role for GRP in modulating It has been shown that BB-like peptides influenced anxiety
behavioral changes and that stressors influence GRP function- responses in several behavioral paradigms. By example, central
ing, these findings support the suggestion that the GRPR should BB administration dose-dependently increased locomotor
be considered a therapeutic target for a subset of CNS diseases. activity in a familiar situation, but the same treatment provoked
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 19

a decline of exploration in a novel (presumably stressful) 9. Leptin, feeding and stress


context (Kent et al., 1998, 2001a,b; Itoh et al., 1994).
Conversely, antagonism of BB1 attenuated signs of anxiety Leptin, a hormone primarily secreted from adipocytes, is
across a range of situations that included those that involved usually associated with appetite or energy regulation (Mal-
conditioned fear (fear potentiated startle) as well as naturalistic endowicz et al., 2007), likely involving the central actions of
(unlearned) stressors, such as pup vocalization upon removal of this peptide (Banks, 2006). However, this hormone has also
the dam, a social interaction test, and approach to a snack in a been implicated in subserving symptoms of major depressive
novel environment (Merali et al., 2006a). Furthermore, bilateral disorder. Although many of the regulatory effects of leptin
infusions of a GRP antagonist into the prelimbic cortex, involve hypothalamic nuclei, it seems that this hormone has
infralimbic cortex or CeA effectively reduced freezing in multiple effects beyond that of feeding and energy regulation.
response to both contextual or explicit cues that had been paired Indeed leptin receptors are widely expressed within brain
with a neurogenic stressor (Mountney et al., 2006). Yet, it regions, including the hippocampus, cortex and cerebellum,
seems that the effects of BB manipulations may be situation which have not typically been linked to feeding or energy
specific, depending on the specific receptors activated or regulation (Harvey, 2007).
blocked. It was reported that a NMB (BB1) receptor antagonist
elicited anxiolytic effects in the elevated plus maze, whereas 9.1. Leptin and cytokines
such an effect was not elicited by a GRP (BB2) antagonist.
However, in a fear potentiated startle paradigm, pretreatment There is impressive evidence suggesting that leptin
with either the BB1 receptor antagonist or a BB2 receptor operates through IL-1b in modulating core body temperature
agonist attenuated the fear potentiated startle response, without as well as feeding, although they may do so through different
affecting basal startle amplitude. Thus, it seems that NMB processes (see Section 11 for a more detailed discussion of
manipulations affected both anxiety and fear responses, cytokines and other inflammatory factors in relation to major
whereas GRP alterations selectively affected fear-related depressive disorder). In this regard, leptin may influence
responses (Bedard et al., 2007). In contrast to BB1 receptors, appetite regulation through its neuronal actions, whereas the
it was reported that within the lateral amygdala, manipulation effects on body temperature are subserved by COX-2 actions
of BB2 receptors influenced the acquisition and performance of involving endothelial cells (Harden et al., 2006; Inoue et al.,
a conditioned fear response. Thus, there is reason to believe that 2006; Luheshi et al., 1999). Specifically, administration of
the BB2 receptor subtype might also contribute to anxiogenic leptin influenced hypothalamic IL-1b in rats, and concur-
processes (Shumyatsky et al., 2002). rently diminished food intake and increased core body
Consistent with the effects of pharmacological manipulations temperature. Conversely, administration of the IL-1 antago-
of BB functioning, elevated 5-HT expression at the DRN was nist (IL-1ra) attenuated the suppressed food intake ordinarily
reported in mice with targeted disruption of the BB1 receptor associated with either peripheral or central leptin adminis-
gene. Moreover, the stressor-provoked 5-HT expression noted in tration (Luheshi et al., 1999). Further support for the IL-1b
wild-type mice was not apparent in mice lacking the BB1 involvement in the effects of leptin came from the
receptor (Yamano et al., 2002). These data suggest that these demonstration that mice lacking the Type 1 IL-1 receptor,
receptors are important in fine-tuning subsets of 5-HT neurons. In did not exhibit the reduction of food intake ordinarily
fact, 5-HT1A receptor gene expression was down-regulated in provoked by leptin (Luheshi et al., 1999). It seems, as well,
BB1-deficient mice. Inasmuch as these mice also exhibited that microglia, a major source for brain cytokines, express
increased indices of anxiety, particularly with respect to elevated certain isoforms of leptin receptors, and that the release of
risk-assessment (reflected by higher frequencies of stretch attend IL-1b likely occurs through a signal transducer and activator
responses in anxiety provoking situations), these data implicate of transcription 3 (STAT3)-dependent mechanism (Pinteaux
bombesin in promoting anxiety (Yamada et al., 2002). et al., 2007).
The use of BB manipulations within a clinical context has
received little attention, despite the fact that (a) pharmacolo- 9.2. Leptin and reward processes
gical treatments profoundly influence anxiety responses in
animal studies and (b) GRP-ir within the locus coeruleus of Leptin also interacts with processes other than cytokines in
suicides was higher than in controls, whereas NMB-ir was moderating food intake. For instance, it has been shown that
reduced at the dorsal vagal complex (DVC) (Merali et al., prolactin treatment attenuated the effects of leptin on food
2006b). It was suggested that BB receptors might turn out to be intake, possibly through STAT-3 changes (Naef and Woodside,
a significant target for the treatment of anxiety disorders, and 2007). Furthermore, intracerebroventricular infusion of leptin
perhaps for depression (Merali et al., 2006b). In this regard, as diminished 5-HTT binding sites in the rat PFC (Charnay et al.,
GABAA subunits have been implicated in anxiety processes, 2000), which coincides with changes thought to be associated
and inter-relations exist between GABA and 5-HT functioning with MDD. Feeding processes have also been associated with
(Sibille et al., 2000), which might be moderated by CRH, it is 5-HT variations, and among obese ob/ob (leptin deficient) mice
conceivable that BB manipulations might affect anxiety that also exhibited a depressive-like behavioral profile, 5-HTT
through a cascade of actions that involve CRH, GABA and mRNA expression was reduced within dorsal raphe neurons
5-HT functioning (see Section 10). (Collin et al., 2000).
20 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

It is of particular significance from the present perspective that depressed patients were accompanied by elevated IL-6 and
leptin administration may alter the rewarding value of otherwise TNF-a levels (Yang et al., 2007). In contrast to these findings,
positive stimuli, as reflected by an increase in the reward however, other investigators reported either no differences of
threshold (i.e., reduced value of the reward) among rats leptin levels between controls and depressed patients (Deuschle
responding for electrical stimulation from the lateral hypotha- et al., 1996; Kauffman et al., 2005) or elevated levels of the
lamus (Fulton et al., 2004). Interestingly, this outcome was only hormone (Antonijevic et al., 1998; Esel et al., 2005; Gecici
evident when the stimulating electrodes were present at sites et al., 2005). Moreover, elevated perceived life stressors
dorsal or dorsolateral to the fornix, but not in closely adjacent (common in MDD patients) were associated with elevated
regions (i.e., brain sites that did not support increased responding leptin levels (Otsuka et al., 2006). Still other reports indicated
for brain stimulation reward that ordinarily occurs from the that elevated serum leptin levels were associated with a lifetime
lateral hypothalamus among food deprived rats). It seems that history of depression and that the levels predicted subsequent
leptin may contribute to reward processes, and it is possible that development of a depressive disorder (Pasco et al., 2007).
some of the appetitive behavioral changes exerted by leptin may
involve changes in motivation to perform (Fulton et al., 2000). 9.4. Leptin involvement in atypical depression

9.3. Relation to stress processes and depression Although these marked inconsistencies might, at first blush,
be disconcerting, they raise the question as to whether analyses
Like several other hormones, it seems that in addition to of a leptin–depression relationship would be better served by
being involved in regulation of feeding (and energy regulation), assessing specific endophenotypes or at least to differentiate
leptin may also be influenced by stressors, and might thus be between subtypes of depression. As already indicated, typical
involved in stress-related pathology. Specifically, stressors and atypical depression are distinguishable from one another on
increased serum and adipose leptin levels in rodents subjected the basis of the vegetative symptoms that accompany illness.
to the distress of water immersion (Konishi et al., 2006). It was Likewise, stressor experiences in some individuals are
similarly observed that in female mice, a chronic (10 days) accompanied by reduced eating, whereas in others these events
restraint stressor regimen provoked an increase of hypotha- are accompanied by increased eating and especially carbohy-
lamic leptin levels. This same treatment, however, reduced drate craving. Dallman and her associates (Dallman et al., 2003;
leptin levels in the thalamus of males and females (Manni et al., Pecoraro et al., 2004) have provided an intriguing conceptua-
2007). The functional significance of these changes cannot be lization of how stressful events might come to produce these
deduced from this study; however, given that eating disorders effects, and as will be seen, although the focus of the model
are more common in females than in males, the unique effects deals with HPA functioning as well as glucocorticoid and CRH
of the treatment on hypothalamic leptin may turn out to be variations at limbic sites, changes of cytokines and leptin might
functionally significant. also be relevant in this regard. According to Dallman,
In light of the belief that stressors influence leptin chronically elevated levels of glucocorticoids (as described
concentrations, and that leptin influences reward processes, earlier) have the effect of stimulating CRH release at the central
it has been suggested that leptin might also be associated with amygdala, thus instigating a stress-like cascade. As well, it was
major depressive illness, possibly subserving the neurovege- suggested that glucocorticoids (or CRH variations) encourage
tative features of depression (e.g., altered food intake and or make salient pleasure-related stimuli and compulsive
weight change) (Lu et al., 2006). Studies in animals not only behaviors, including sucrose and fat consumption, thereby
demonstrated low leptin levels in association with stressor motivating the consumption of ‘‘comfort foods.’’ The
exposure, but also that the depressive-like behavioral dis- consumption of these comfort foods, which promote abdominal
turbances introduced by a chronic stressor could be antagonized obesity, has the effect of inhibiting brainstem catecholamines
by systemic or intrahippocampal (but not hypothalamic) leptin and hence limiting processes that lead to further CRH
administration (Lu et al., 2006). In fact, these investigators variations, thereby diminishing anxiety. Given the relation
suggested that leptin ought to be considered as a possible between leptin and CRH (and cytokine) and glucocorticoid
therapeutic target for the treatment of depression. processes, it can be readily imagined that this peptide might
Unfortunately, the data from human studies concerning contribute to changes of ingestion associated with depressive
leptin variations in relation to depression have been incon- features in a subset of individuals.
sistent. Several investigators reported the expected low levels of In his insightful review, Black (2006) tied stress, various
leptin in depressed patients (Jow et al., 2006; Kraus et al., 2001) hormones and neurotransmitters, cytokines, and free fatty acids
and in those who attempted suicide (Atmaca et al., 2002; to several pathological conditions. This perspective, like that
Westling et al., 2004), as well as potential changes of advanced by Dallman, incorporates CRH changes into a model
glucocorticoid receptor sensitivity leading to leptin alterations that predicts variations of fat consumption and the development
with successful treatment of depression (Himmerich et al., of pathology. Essentially, it was suggested that stressors
2007). As well, brain leptin mRNA levels were diminished in stimulate NE release and hence NFkB activation in macro-
suicides relative to controls (Eikelis et al., 2006). Furthermore, phages and visceral fat, ultimately resulting in the provocation
in line with the view that leptin and cytokine variations may be of an acute phase response. Visceral fat, from this perspective,
linked, it was reported that reduced leptin levels among is a key player in this cascade, as cytokines (particularly IL-6
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 21

and TNF-a) as well as leptin, are released in response to coordinated, and whether these act together to influence
sympathetic signals. Significantly, stressors and inflammation cytokine functioning, is uncertain. Nevertheless, as will be seen
give rise to several hormone and enzyme changes (cortisol, IL- in Section 11, cytokines have been implicated in depressive
6, angiotensin II, and 11b-hydroxysteroid dehydrogenase-1) illness, and as such the leptin–ghrelin connection to depression
that might contribute to appetite stimulation and further fat warrants further consideration.
accumulation. In addition to these peripheral processes, leptin
released from fat cells come to stimulate hypothalamic CRH, 10. Gamma-aminobutyric acid (GABA)
which then influences sympathetic and HPA functioning, and
may inhibit fat consumption. Moreover, by virtue of CRH Several lines of evidence have implicated GABAergic
changes associated with this cascade, emotional changes can be processes in anxiety and depressive illness. These include the
expected. demonstrations that (a) plasma and CSF GABA levels were
In view of the apparent linkage between leptin, food increased in stress and anxiety situations, (b) stressor exposure
consumption and depressive disorder, the proposition was influenced GABAA receptor functioning, (c) factors that
advanced that cytokines and leptin may be differentially promote vulnerability to elevated anxiety and depressive-like
expressed in typical and atypical depression and may be behaviors, such as early life stressors, also influence GABAA
responsible for the symptom profiles expressed in these illness g2 subunit expression, (d) GABAA acting agents influence
subtypes (Andreasson et al., 2007). Commensurate with this anxiety, (e) antidepressants influence interneuron functioning
view, plasma leptin levels among patients with seasonal (Akinci and Johnston, 1993; Brambilla et al., 2003; Caldji
affective disorder (characterized by hyperphagia with cravings et al., 2000, 2004; Krystal et al., 2002; Shiah and Yatham,
for carbohydrates and moderate weight gain) appeared to be 1998; Tunnicliff and Malatynska, 2003), and (f) marked
tied to seasonal changes (Cizza et al., 2005). Yet, it was also GABAA receptor subunit expression is disrupted in some brain
observed that serum leptin levels were higher among patients regions (e.g., frontopolar cortex) of suicide victims (Merali
presenting with atypical depression than in those with typical et al., 2004a,b). The potential involvement of GABA in
features (Gecici et al., 2005) contrary to the view advanced by mediating depression and anxiety was reinforced by findings
Andreasson. At this juncture there are simply too few published that reciprocal innervation might exist between GABAA and 5-
studies to make definitive conclusions regarding the contribu- HT processes within brain regions that have been associated
tion of leptin to typical vs. atypical major depressive disorder. with depression, such as the PFC and hippocampus (Brambilla
Nevertheless, the views expressed by Dallman, Black and et al., 2003), and GABAA and DA activity appeared to
Andreasson provide a framework that encourages further modulate amygdala functioning (Marowsky et al., 2005).
analyses of the linkage between leptin and specific endophe- Likewise, estrogen and progesterone as well as the progester-
notypes of these illnesses. one metabolite, allopregnanolone, act on the GABAA receptor,
and may thus influence gender-based vulnerability to mood
9.5. Ghrelin, food consumption and motivational processes states (Birzniece et al., 2006). Taken together, these findings
raise the possibility that treatments that target neuroactive
Unlike most peptides that have been implicated in feeding steroids and the GABAA receptor may be fruitful in the
through their actions on satiety processes, ghrelin, a relatively treatment of depression.
recent addition to the group of peptides associated with food
ingestion, operates by stimulating food intake (Van der Lely 10.1. GABAA variations associated with suicide/depression
et al., 2004). Moreover, it was reported that the ghrelin receptor
growth hormone secretagogue 1 receptor (GHSR) is present Inconsistent results have been reported concerning GABA
centrally, not only at hypothalamic sites involved in energy variations in the brain of depressed suicides. It had been
metabolism but also within the ventral tegmentum, which it will reported that GABA levels within the PFC were inversely
be recalled is fundamental in reward processes, and is sensitive related to severity of depression (Honig et al., 1988), but
to stressor actions. Indeed, paralleling the increased food subsequent studies indicated that neither GABA levels (Korpi
consumption, ghrelin stimulated increased DA neuronal et al., 1988), GABA-related enzymes (Cheetham et al., 1988;
activity at the ventral tegmentum and influenced DA turnover Sherif et al., 1991), the GABA transporter (Sundman-Eriksson
at terminal sites within the nucleus accumbens (Abizaid et al., and Allard, 2002) nor GABAB binding in the frontal cortex
2006). Thus, it was suggested that beyond its other actions, differed between drug-free depressed suicide victims and
ghrelin might regulate feeding through its effects on controls (Arranz et al., 1992; Cross et al., 1988; Sundman et al.,
mesolimbic reward processes. Interestingly as well, it was 1997). Despite these negative findings, it is significant that
reported that intravenous ghrelin administration had the effect GABAA/BDZ (benzodiazepine) binding sites were elevated in
of inhibiting cytokine production otherwise provoked by depressed suicides (Cheetham et al., 1988), indicating that
endotoxin administration (Li et al., 2004a,b; Taub, 2007). In GABAA receptor subunit expression is likely changed. More-
addition, ghrelin also had anti-inflammatory effects through its over, whole genome analyses (using microarrays) have
action on vagal afferents (Wu et al., 2007). In effect, the linkage indicated the presence of GABA-glutamate related genes in
that exists between leptin and cytokines is also evident with depression and suicide (Sequeira and Turecki, 2006). In line
respect to ghrelin. Whether ghrelin and leptin variations are with this broad analysis, it was reported that the GABAA a1 and
22 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

a6 polymorphisms were associated with mood disorders in planning (Kalivas and Nakamura, 1999; Tanji and Hoshi,
female patients (Yamada et al., 2003a,b). 2001), coupled with its influence on nucleus accumbens
As the function/pharmacology of the GABAA receptor functioning (a region that may be fundamental in the anhedonic
complex is largely controlled by the expression of the a (a1, a2, features of depression; Stevenson et al., 2003), the interactive
a3, a4, a5), d and g2 subunits, we assessed their expression and effects of GABAA and CRH neuronal activity may contribute to
inter-relations in control and suicide brains. In addition to CRH aspects of depressive symptomatology.
receptor variations, mRNA expression of the a1, a3, a4, and d
receptor subunits were reduced in the frontopolar region of 10.3. GABA, CRH and 5-HT inter-relations
suicide victims. It might be of significance that our partial
analysis of the GABAA receptor functional genome revealed An interesting link between 5-HT and GABA receptors has
that within the frontopolar and dorsomedial prefrontal cortex been reported, essentially indicating that 5HT1A receptors
pronounced cross-correlations of GABAA subunit expression regulate GABAA receptor expression (Sibille et al., 2000), and
was evident among nondepressed individuals, suggesting a high indeed it appears that PFC 5-HT neuronal functioning plays a
degree of coordinated gene regulation. In contrast, the GABA prominent role in regulating GABAergic inhibitory transmis-
subunit intercorrelations were markedly diminished in suicide sion. Thus, it is significant that electrophysiological studies
brains (with the exception of the inter-relations between the a2, indicated that 5-HT application induced a marked desensitiza-
a3 and a4 subunits). The frequency of significant relations, and tion of the amplitude and frequency of spontaneous inhibitory
the specific profiles of these relations (in PFC regions) suggests postsynaptic currents (sIPSC) and also provoked a marked
that the findings were not spurious (Merali et al., 2004b), and it reduction of electrically evoked IPSC (eIPSC). Further, chronic
has, indeed, been reported that the expression of several (but not acute) SSRI treatment provoked the down-regulation of
GABAA receptor subunits (a1, a2 a3, b2/b3, g2, d) within the synaptic activity of forebrain 5-HT2 receptors (Zhong and
dentate granule neurons also appeared to be coordinated (Benke Yan, 2004). It is interesting, as well, that CRH treatment
et al., 1997; Brooks-Kayal et al., 1999; Kern and Sieghart, prolonged the regulation of sIPSCs elicited by 5-HT in PFC
1994; Khan et al., 1996; Mertens et al., 1993). As the genes for slices, and such an effect was also apparent in slices obtained
these subunits appear on different chromosomes, the subunit from previously stressed animals. Conversely, the effects of
coordination could not be attributed to factors related to shared CRH and of the stressor treatments were blocked by the CRH
chromosomal localization (e.g., linkage effects; Brooks-Kayal antagonist a-helical CRH and by the CRH1 antagonist
et al., 1999). These findings raise the possibility that the astressin. Likewise, if phospholipase C or protein kinase C
GABAA receptor subunit changes, or the disturbed coordina- (PKC) was inhibited, then the effects of CRH were also
tion between the GABAA receptor subunits, contribute to antagonized (Tan et al., 2004). Taken together, it seems that
depression/suicide or are secondary to the illness/distress stressors and CRH (or CRH activation in response to stressors)
associated with it (Merali et al., 2004b). influence 5-HT regulation of GABA transmission, and PKC
may be fundamental in mediating this action. In view of the
10.2. GABA and CRH inter-relations fundamental role of GABAergic inhibitory transmission in PFC
functioning, it was suggested that chronic SSRI treatments may
There is reason to believe that CRH and GABAA functioning ultimately have their therapeutic effects through actions on
may be inter-related within hypothalamic processes as well as GABA processes (Zhong and Yan, 2004). Yet, it has also been
in limbic neural circuits. Interestingly, in vitro application of the reported that 5-HT1A knockout mice respond poorly to
GABAA antagonist bicuculline methiodide influenced AVP benzodiazepine (BDZ) anxiolysis, an effect that was first
mRNA, without affecting CRH expression (Bali and Kovacs, attributed to a loss of GABA binding sites, and specifically to a1
2003), although in vivo, both CRH and AVP gene expression and a2 GABAA subunit reductions in the CA1 and dentate of
were increased (Cole and Sawchenko, 2002). In line with the the hippocampus. This link between 5-HT receptor status and
supposition that GABAA may have functional influences on GABAA receptor expression are consistent with the findings
CRH under stressor conditions, it was shown that the basal that BDZ treatment when coupled with an antidepressant has
expression of transcripts encoding several subunits of the modest, albeit short-term, positive effects in treating depression
GABAA receptor were present within CRH neurons funda- (Furukawa et al., 2002). However, given the limited actions of
mental in the stress response (Cullinan, 2000; Cullinan and such manipulations, it is unlikely that GABA functioning alone
Wolfe, 2000). accounts for depression, and other neurotransmitter processes
Beyond potential interactions between CRH and GABAA would have to be concomitantly altered for antidepressive
within the hypothalamus, CRH is uniquely expressed in GAD actions to become manifest.
positive interneurons in rat cortex and chronic stressors can In addition to 5-HT influences on GABAergic functioning
affect GABAA receptor expression (Cullinan and Wolfe, 2000; within limbic regions, the activity of 5-HT neurons within the
Yan et al., 1998). Likewise, GABAergic agents may affect CRH DRN appear to be regulated by GABAA and 5-HT1A inhibitory
mRNA expression within limbic sites (Cullinan and Wolfe, receptors (Boothman et al., 2006; Cremers et al., 2007; Judge
2000; Gilmor et al., 2003; Skelton et al., 2000; Stout et al., et al., 2006). Given that raphe 5-HT1A autoreceptors influence
2001). Given the important role for the PFC in subserving forebrain 5-HT release, it is likely that reciprocal influences
various cognitive processes, including hedonia and behavioral between GABA and 5-HT processes may affect mood states.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 23

The inter-relations with GABAA functioning may also involve inflammatory actions. As well, IL-1b and IFN-g are examples
other receptor subtypes, as 5-HT2C antagonists augmented the of pyrogens, in that they provoke febrile responses, whereas IL-
acute effect of SSRIs on hippocampal 5-HT release, and this 2 is a non-pyrogenic cytokine. Interestingly, although TNF-a
effect was modifiable by GABA manipulations (Cremers et al., and IL-6 are often referred to as pyrogens, under certain
2007). conditions they may actually reduce the fever provoked by IL-
Just as CRH may influence both NE and 5-HT neuronal 1b, thus acting in a cryogenic fashion (e.g., Long et al., 1992).
activity, which could potentially impact upon depressive states
(Ruggiero et al., 1991), it is possible that CRH-GABA inter- 11.1. Cytokines as signaling molecules
relations have such an effect. Indeed, based on studies using
dual immunoelectron microscopy to assess different types of Signaling involving IL-1b is dependent upon an interaction
synaptic contacts, it was suggested that CRH has both direct between its type 1 receptor and the IL-1b receptor accessory
and indirect effects on DRN 5-HT neurons, and that GABA protein (Rothwell, 1999; Huang et al., 1997; Greenfeder et al.,
might serve as an essential mediator by which CRH (and 1995), both of which are found on the surface of numerous cell
possibly stressors) come to influence DRN 5-HT functioning types, including lymphocytes, macrophages/monocytes,
(Waselus et al., 2005). Alternatively, CRH variations may endothelium and even neurons and glial cells within the brain
instigate 5-HT receptor changes, which then influence frontal (Mantovani et al., 1996; Vitkovic et al., 2000). A second IL-1
cortical GABAA functioning (Tan et al., 2004). Of course, receptor, termed type II, acts as a ‘‘decoy receptor,’’ in that it
further GABA-related interactions likely exist that could does not transduce a signal upon binding the cytokine (Colotta
influence depressive illnesses. For instance, it has been reported et al., 1994). As well, there is an endogenous antagonistic
that the progesterone metabolite allopregnanolone potentiated ligand, namely the IL-1b receptor antagonist (IL-1ra), which
the GABAA-mediated inhibition of DRN 5-HT neurons (Kaura competes with IL-1 for binding sites, in effect mopping up IL-
et al., 2007). Moreover, GABAA d subunit expression may be 1b (Mantovani et al., 1996; Lucas et al., 2004). Essentially, the
influenced by progesterone, and the highly stressor-sensitive actions of IL-1b are under tight regulation, involving a series of
neurosteroid 3 alpha,5 alpha-tetrahydrodeoxycorticosterone different factors that comprise the IL-1 family. Ultimately,
(THDOC) (Reddy, 2003) might mediate this effect (Maguire activation of the IL-1b type 1 receptor initiates a cascade of
and Mody, 2007). As such, these data raise the possibility that events culminating in the phosphorylation and consequent
stressor effects on depressive symptoms, and the sex degradation of the endogenous inhibitory factor kappa B (IkB),
differences that exist, may involve interactions between resulting in the translocation of nuclear factor kappa B (NFkB)
GABAA subunits and THDOC and/or allopregnanolone. to the nucleus where it promotes the expression of several genes
that play an important role in neuronal homeostasis and
11. Cytokines inflammatory processes (Lucas et al., 2004).
Like IL-1b, IL-6 is typically secreted from a variety of
The primary function of the inflammatory immune system is immune cells, particularly macrophages and T lymphocytes,
that of protecting the individual from bacterial and viral insults. activated by antigenic challenge. Moreover, IL-6 and its
Foreign particles ordinarily stimulate an orchestrated immune receptor are found on a variety of non-immune cells, including
response against specific antigens (foreign particles), and fibroblasts, synoviocytes, adipocytes, osteoblasts, endothelial
immunological factors develop a memory for them. Thus, upon cells, granulocytes, as well as microglia, astrocytes and even
subsequent encounters with these antigens, a rapid and robust neurons (Cichy et al., 1996; Helwig et al., 2007; Ringheim
immune response is mounted. Immune cells (T and B et al., 1995; Rosell et al., 2003), indicating its importance in the
lymphocytes, macrophages as well as endothelial cells) regulation of multiple physiological processes. The IL-6
synthesize and secrete signaling molecules, referred to as receptor is an 80 kDa transmembrane glycoprotein, which
cytokines, which also elicit growth and differentiation of exists in a membrane bound form and may also be shed through
lymphocytes. Thus, these cytokines are fundamental for the proteolysis to produce a 55 kDa soluble form (Mullberg et al.,
organism to mount an effective immune response against 1994). Binding of IL-6 to its receptor results in a relatively low
invading pathogens. affinity structure which requires subsequent recruitment of
The list of polypeptides that comprise the rapidly increasing glycoprotein-130 (gp130) to create a functional receptor
family of immunotransmitters (cytokines) include the inter- complex, thereby promoting a signaling cascade involving
leukins (IL), tumor necrosis factors (TNF), interferons (IFN), the stimulation of various protein kinases (Taga, 1996).
chemokines, and growth and cell stimulating factors (Rothwell, Signaling through the IL-6 receptor-gp 130 complex primarily
1999; Elenkov et al., 2005). Classification of the cytokines has involves activation of the Janus kinase (JAK)- signal and signal
been based upon their molecular structure, as well as common transducer and transcription activator (STAT) pathway. Indeed,
physiological actions they possess, including the production of activation of the IL-6 receptor complex triggers conformational
fever (pyrogenicity) or inflammation (i.e. swelling and changes in the cytoplasmic domain of the receptor that results
irritation resulting from leukocyte (white blood cell) infiltra- in JAK aggregation and subsequent phosphorylation of various
tion). IL-1b, TNF-a and IL-6 (all released from activated STATs. Thereafter, nuclear translocation of STAT homo- or
macrophages) are potent proinflammatory cytokines, whereas heterodimers provokes the transcription of a number of immune
IL-4 and IL-10, which are released from T-cells, have anti- and CNS regulatory proteins (Murray, 2007).
24 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Interferons (IFNs) are broadly divided into either Type 1 cellular proliferation, immune and inflammatory responses, and
IFNs, including the IFN-a and IFN-b isoforms, which behavioral states (e.g., those associated with sickness). Under
originated from a common ancestral gene, or the structurally normal physiological conditions, systemic cytokines are
unrelated Type 2 form, IFN-g, which has many functional maintained at a low level, and they usually act in an autocrine
actions in common with the aforementioned IFNs (De Maeyer (local cellular actions in a feedback capacity) or paracrine (acts
and De Meyer-Guignard, 1998). The a and b isoforms of IFN upon cells in proximity of where they are produced) manner at
bind to a 66 kDa receptor chain that is associated with several primary or secondary lymphoid organs, such as the spleen,
accessory proteins (Maher et al., 2007). Likewise, a 90 kDa lymph nodes or liver (Alkharfy et al., 2000). Under homeostatic
receptor binds IFN-g; however, to be functionally active the conditions, cytokines rarely have actions on cells located some
receptor requires the presence of an accessory protein (Farrar distance away, and hence their endocrine actions are limited.
et al., 1987). Like IL-6, the main signaling pathway utilized by Typically, any cytokine spillover into the circulation is rapidly
the IFNs is the JAK-STAT pathway, resulting in sequential diluted (Bocci, 1991). However, during severe immunological
phosphorylation steps by intracellular protein kinases (Stark challenge, such as septic shock, circulating levels of pro-
et al., 1998; Maher et al., 2007). However, the Type 1 inflammatory cytokines are markedly increased (Damas et al.,
interferons can also stimulate phosphatidylinositol 3-kinase 1997; Oberholzer et al., 2005). The elevated systemic cytokines
(PI3K) and mitogen-activated protein kinase (MAPK) path- can provoke the liberation of acute phase proteins and
ways, which may be responsible for their ability to interfere augmented utilization of steroidal hormones and central
with viral replication and promote the apoptotis of tumor and monoamines (Raison et al., 2006). In effect, upon presentation
other cells (Pokrovskaja et al., 2005). of a sufficient challenge, the role of cytokines may switch from
Similar to the aforementioned cytokines, TNF-a is a that of a locally acting paracrine or self-regulating autocrine to
pleiotropic cytokine, having physiological actions upon mono- that of an endocrine effector.
nuclear and polymorphonuclear blood cells, fibroblasts, kera-
tinocytes, and insulin sensitive adipocytes, as well as brain 11.3. Cytokines influence CNS processes
neurons and glial cells (Eigler et al., 1997). The major producers
of TNF-a are macrophages (and microglia in brain). TNF-a Given that the blood brain barrier (BBB) is relatively
receptors are often shed from the cell membrane and circulate impermeable to cytokines, owing to their size and hydro-
freely in a soluble form, binding any excess ligand (Rigby, 2007). philicity, the mechanism by which circulating cytokines
Endogenous antagonists for TNF-a, including a-melanocyte influence brain function is still a matter of debate. However,
stimulating hormone (a-MSH), which blocks the actions of cytokines may enter the brain through sites where the BBB is
TNF-a after both central and peripheral administration, also somewhat compromised, namely at circumventricular organs
contribute to cytokine regulation (Rajora et al., 1997). (median eminence, subfornical organ, area postrema, organum
The actions of TNF-a are mediated by two distinct receptor vasculosum) (Goehler et al., 2006). As well, saturable transport
subtypes, which are termed p55 and p75, with molecular mechanisms capable of moving IL-1b and TNF-a may allow
weights of 55 and 75 kDa, respectively (Neta et al., 1992; for limited penetration of cytokines into the brain (Banks, 1999;
Dinarello, 1987). The p55 receptor is generally regarded as Banks et al., 1991; Gutierrez et al., 1993). In addition to routes
playing an important role in apoptosis and cellular necrosis, dependent upon the circulatory system, cytokines may also
whereas the p75 receptor may be more important for nutritive influence brain processes by fibers projecting from the
functions, such as the promotion of thymocyte proliferation periphery to the CNS. Indeed, IL-1b and TNF-a stimulate
(Rigby, 2007). Although TNF-a shares several intracellular visceral branches of the vagus nerve to modulate many
signaling pathways with IL-1, it possesses a unique apoptotic neuroendocrine and neurochemical states, and hence behavior
signaling mechanism involving a death domain region (Maier and Watkins, 1998; Wieczorek et al., 2005).
intracellularly attached to its p55 receptor. Activation of this It is noteworthy that stressful events or certain immunologic
death domain triggers the recruitment of caspases 1 and 8, challenges may also compromise the integrity of the BBB,
which may engender cytotoxic consequences (Ashkenazi and thereby permitting greater cytokine action within the CNS. In
Dixit, 1998). As well, activation of second messenger systems this regard, it has been proposed that CRH release promotes
may result in stimulation of phospholipase A2 and the brain mast cell activation, which in turn increases the release of
production of prostaglandins or leukotrines via an arachidonic IL-6, IL-8 and vascular endothelial growth factor (Theoharides
acid substrate (Woo et al., 2000). Many of the intracellular and Konstantinidou, 2007; Esposito et al., 2001). Likewise,
actions of p55 may be counterbalanced by activation of the p75 pro-inflammatory cytokines themselves enhance permeability
receptor, which promotes stimulation of the transcription factor of the BBB and hence up-regulate their own production at
NFkB and under certain circumstances may have inhibitory vascular sites (de Vries et al., 1996; Saija et al., 1995).
effects on apoptosis (Rothe et al., 1994; Dopp et al., 2002). Furthermore, it seems that chemokines (chemo-attractant
cytokines) may also disrupt BBB functioning, promoting the
11.2. Paracrine and autocrine actions of cytokines formation of vasogenic brain edema that is associated with
various neuropathological conditions, including brain trauma,
Cytokines exert a range of effects on numerous cell types, ischemia, CNS infection, presence of brain tumors, and
and thus may affect several physiological processes, such as recurrent MS flares (Stamatovic et al., 2006).
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 25

Once cytokines gain entry to the brain, they interact with 11.4. Neurochemical effects of cytokines: implications for
receptors on cells lining the BBB, around the meninges as well depressive illness
as vascular areas of the brain (Rivest et al., 2000). Through
volume diffusion, cytokines may ultimately reach nuclei deep Cytokine activation elicits several neurochemical and
within the brain parenchyma, where they interact with behavioral responses reminiscent of those provoked by
receptors located at hypothalamic, amygdaloid and brain stressors as well as those of depressive illness. As will be
stem nuclei (Cunningham and De Souza, 1993; Kinouchi et al., discussed shortly, studies in animals revealed that pro-
1991; Konsman and Dantzer, 2001; Nadeau and Rivest, 1999; inflammatory cytokines, like stressors, increased HPA func-
Schobitz et al., 1994). In this respect, it seems that cytokines tioning, and influenced a range of monoaminergic and
may actually have several functions similar to those of peptidergic neurotransmitter changes at hypothalamic and
classical neurotransmitters by modulating neuronal Ca2+ extra-hypothalamic sites (Anisman and Merali, 1999; Hayley
channels, activation of intracellular second messenger et al., 2004b; Kronfol and Remick, 2000). Paralleling these
systems, and stimulation of the MAP kinase pathways effects, rodents treated with certain cytokines (or immune
(Tancredi, 1992). activating agents) displayed a behavioral profile characteristic
Induction of secondary mediators, such as prostaglandins, of depressive-like states comparable to those elicited by
may also be responsible for some of the central neurochemical stressors (Anisman and Merali, 1999). Furthermore, humans
effects of cytokines. Evidence supporting this proposition undergoing immunotherapy (e.g., using IFN-a) display
comes from studies showing that pretreatment with the neurovegetative changes and pronounced depressive symp-
cyclooxygenase inhibitor indomethacin (inhibiting conversion toms.
of arachidonic acid into prostaglandins) attenuated at least The mechanisms responsible for the cytokine-elicited
some of the central actions of IL-1b, such as the altered NE depressive features have yet to be identified, but several
variations observed within the hypothalamus (Masana et al., possible candidate systems have been proposed. Pro-inflam-
1990). matory cytokines influence central neurotransmitter and growth
Beyond their actions on CNS processes, cytokines and factors that have been thought to be related to depression (e.g.,
their receptors are endogenously expressed in the brain CRH, 5-HT and BDNF), and also affect enzymatic pathways
(Cunningham and De Souza, 1993; Buttini and Boddeke, involved in the production of oxidative species and other
1995), likely reflecting de novo synthesis by glia and possibly neurodegenerative factors (Anisman and Merali, 1999;
by neurons. Moreover, pro- and anti-inflammatory cytokine Churchill et al., 2006; Ericsson et al., 1994; Hopkins and
levels are markedly increased by traumatic insults (Johnson Rothwell, 1995; Quan and Herkenham, 2002). However, in
et al., 2006; Kamm et al., 2006; Zhu et al., 2006), endotoxin considering the involvement of cytokines in depressive illness
treatments (Ledeboer et al., 2002; Rivest, 2001; Turrin et al., there are several fundamentally important issues that need to be
2001), and treatment with several inflammatory cytokines emphasized from the start. Although about 50% of patients
(Churchill et al., 2006) as well as by neurogenic and treated with interferon-a (for certain cancers and hepatitis-C)
psychogenic stressors (Maier et al., 1999; Miyahara et al., develop marked symptoms of depression (Capuron et al.,
2000; Nguyen et al., 1998; Zhu et al., 2006). In this regard, 2002a; Raison et al., 2006), rodents treated with this cytokine
endothelial cells that line the interior surface of blood vessels (unlike IL-1b or TNF-a) exhibit few noticeable behavioral
and that of the ventricles produce IL-1b and IL-6, and their changes, and certainly none that bring to mind depressive
concentrations are increased by infection or injury (Banks, features (Anisman et al., in press, 2007a,b). Reconciling the
2001). Further, microglia, which serve as the brains disjunction between the effects seen in humans and those seen
specialized immune cells, are primary cytokine producers, in animals is possible by considering the broader context in
and the synthesis of these cytokines were augmented by head which IFN-a is administered. Specifically, patients receiving
injury, stroke and by neurotoxins (Kamm et al., 2006; Sriram immunotherapy are already under considerable strain given
et al., 2006; Zhu et al., 2006). their current disease state. As stressors and cytokines share
Cytokine–peptide interactions play an important role in multiple similarities, it is not unreasonable to suppose that the
shaping both peripheral immunity, as well as neuroinflamma- stressor background on which the treatment is applied would
tory cascades within brain regions important for several moderate the effects of cytokines on neurochemical processes.
neurological and neuropsychiatric conditions. Indeed, most From this perspective, synergies between cytokines and
centrally active peptides and cytokines also have co-existing traditional stressors might increase the strain placed on
receptors present on peripheral immune cells, and may biologically adaptive processes, particularly when these occur
influence immune functioning through activation of common on a chronic basis, thus leading to allostatic overload, and hence
molecular signaling mechanisms. Moreover, immune cells increased vulnerability to psychological disturbances.
(e.g., T lymphocytes) produce and respond to DA and 5-HT
(Blalock, 1984, 1994; Cosentino et al., 2007). Thus, cytokines 11.5. Neurochemical consequences of IL-1b, IL-6 and
and peptides may be viewed as messengers between the TNF-a
immune system and the brain, and may either augment or
antagonize one another’s actions, depending upon the state of Given that systemic cytokine administration promotes a
their microenvironment. cascade of neuroendocrine and brain neurochemical changes
26 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

reminiscent of those elicited by psychogenic and neurogenic of acute bolus treatments, and the data concerning the effects of
stressors, it has been suggested that the brain interprets repeated or sustained cytokine administration are sparse. This is
inflammatory immune activation, much as if these challenges somewhat puzzling, as inflammation, viral insults and cytokine
constituted stressors (Anisman and Merali, 1999; Dunn et al., immunotherapy all involve persistent cytokine elevations.
1999; Herman and Cullinan, 1997; Herman et al., 2003). Of Moreover, there is reason to believe that chronically increased
course, the effects of cytokines and traditional stressors are not cytokine levels may engender more profound effects than acute
identical (Buller et al., 2001). Moreover, when individuals are treatment, possibly owing to the potential for compromised
exposed to neurogenic or psychogenic insults they engage in BBB functioning. As well, chronic cytokine elevations may
the appraisal or processing of information related to the exert excessive strain on physiological systems, just as chronic
stressor, leading to coping efforts to deal with the challenge stressor exposure might have such an effect (McEwen, 2000).
confronting them. Bacterial or viral infections certainly do not In fact, sustained systemic or intracerebroventricular IL-1b
promote comparable effects with respect to cognitive informa- administration promoted persistent HPA activation, including
tion processing. Yet, this does not belie the possibility that protracted variations of CRH, CRH receptor and proopiome-
cytokine activation might result in appraisal or interpretive lanocortin gene expression, as well as elevated secretion of
processes being altered. Furthermore, as indicated earlier, ACTH, beta-endorphin and corticosterone (Parsadaniantz et al.,
multiple neural pathways are excited in response to different 2000; van der Meer et al., 1996). Further, chronic central or
types of psychogenic stressors (Merali et al., 2004a; Morrow intravenous IL-1b treatment increased immediate early gene
et al., 2000), and it is likewise possible that cytokines and expression in the PVN, CeA and supraoptic nucleus (Niimi
stressors might induce their actions through different neural et al., 1997).
circuits. It appears that in contrast to the HPA activation elicited Beyond their independent effects on neurochemical pro-
by psychogenic stressors that involve amygdala activation, cesses, certain cytokine combinations (e.g., IL-1b plus IL-6, or
systemic stressors may do so through non-limbic circuits IL-1b plus TNF-a) may synergistically increase the behavioral
(Buller and Day, 2002; Buller et al., 2001; Herman and and neuroendocrine alterations (Bluthe et al., 1994; Brebner
Cullinan, 1997). et al., 2000; van der Meer et al., 1995; Zhou et al., 1996).
It has been maintained that multidirectional communication Likewise, the combination of IL-1b and a stressor synergis-
exists between the immune system and other systems within the tically increased the in vivo release of hippocampal 5-HT
brain and periphery. There are, indeed, ample data indicating (Lacosta et al., 1998; Merali et al., 1997; Song et al., 1999).
that inflammatory immune activation (or administration of pro- More recently, it was observed that the synergistic effects on
inflammatory cytokines), like processive stressors, increases cortisol and central monoamine turnover were pronounced
HPA activity as well as the utilization of 5-HT and NE within when a cytokine (IFN-a), viral analogue (poly I:C) or bacterial
the PVN, mPFC, and central amygdala (Anisman and Merali, endotoxin treatment (lipopolysaccharide: LPS) was super-
1999; Day et al., 1999; Hayley et al., 1999a, 2002b). In imposed on a backdrop of psychosocial distress (Anisman et al.,
addition, systemically or centrally administered IL-1b stimu- 2007a; Gandhi et al., 2007; Gibb et al., 2005; Quan et al., 2003).
lated in vivo hypothalamic and hippocampal NE, DA and 5-HT These findings are consistent with the perspective described
release (Linthorst et al., 1995; Mohankumar and Quadri, 1993; earlier, namely that in considering the potential neurochemical
Zhang et al., 2001), as well as that of 5-HT at several limbic or behavioral consequences of cytokine treatments in humans,
sites (Linthorst et al., 1995; Merali et al., 1997; Shintani et al., it is essential to consider the backdrop upon which these
1993, 1995; Song et al., 1999). Moreover, inhibition of IL-1b treatments are superimposed.
attenuated the neurochemical consequences of an immobiliza- In contrast to IL-1b and TNF-a, the central neurochemical
tion stressor, suggesting a role for this cytokine in mediating the and behavioral effects of IFN-a (and IFN-g) have received
central effects of the stressor (Shintani et al., 1995). More limited attention, despite the fact that this cytokine has been
recently, it was reported that the behavioral and neuroendocrine used in clinical conditions, and as already indicated, it is known
effects of a chronic mild stressor was prevented among IL-1b to engender depression in treated patients. In vitro studies
type 1 receptor knockout mice (Goshen et al., 2007). As well, demonstrated that IFN-a stimulated CRH release within the
mice lacking the IL-1b receptor also failed to display the usual amygdala and hypothalamus (Raber et al., 1997), and
reduction of neuroplasticity (hippocampal neurogenesis) that administration of this cytokine provoked several in vivo brain
was evident in wild-type mice that received this stressor neurochemical and hormonal changes (Schaefer et al., 2003),
treatment (Goshen et al., 2007). In contrast, genetic ablation of although the magnitude of the effects were considerably less
the TNF-a p55 receptor did not influence the HPA response than those elicited by IL-1b. As the non-steroidal anti-
provoked by restraint or LPS, although, as predicted, the inflammatory (NSAID) diclofenac attenuated the HPA hormo-
corticoid elevation elicited by TNF-a was greatly attenuated nal effects, it is likely that the inflammatory cyclo-oxygenase
(Hayley et al., 2004b). Hence, there is reason to suppose that pathway (or other inflammatory factors) plays a pivotal role in
selective IL-1b changes that foster the emergence depressive- the elicitation of the HPA response to IFN-a.
like consequences might occur following exposure to ceratin In humans, acute IFN-a treatment in healthy volunteers and
stressors. in hepatitis patients increased ACTH, cortisol and circulating
Most studies that assessed the influence of cytokines on IL-6, but did not appreciably influence other cytokines (Cassidy
neurochemical and behavioral processes focused on the impact et al., 2002; Shimizu et al., 1995). With repeated treatment,
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 27

however, the elevated HPA activity elicited by IFN-a (when effects secondary to CRH release (Hayley et al., 2005). An
administered acutely) was no longer apparent (Gisslinger et al., alternative view that is not exclusive of other processes is that
1993). In rodents, however, a similar profile was not apparent. It cytokines, such as IFN-a, may stimulate indoleamine-2,3-
will be recalled that in contrast to the potent effects of IFN-a in dioxygenase (IDO) and GTP-cyclohydrolase activity, which
humans, the effects of this cytokine on circulating ACTH and foster degradation of the 5-HT precursor tryptophan (Menkes
corticosterone is modest in rats and mice (Anisman et al., in and MacDonald, 2000) thereby reducing 5-HT functioning and
press, 2007a,b; Menzies et al., 1996). It was even reported that leading to depressive illness (Capuron et al., 2002a,b,c).
neither 1 nor 5 days of treatment increased corticosterone levels Relatedly, IDO may have neuroprotective functions by limiting
(De La Garza and Asnis, 2003). In contrast, Anisman et al. (in inflammatory processes associated with infectious or traumatic
press, 2007a,b) observed that IFN-a increased corticosterone insults (Kwidzinski and Bechmann, 2007). However, IFN-a
levels (although not to the extent that IL-1b had such effects), also promotes metabolism of tryptophan into kynurenine and
and although the sickness associated with the cytokine subsequently into the oxidative metabolites 3-hydroxy-kynur-
diminished over five successive days of treatment, the enine and quinolinic acid (which themselves are increased in
corticosterone elevations were not reduced and, in fact, were depression) that may have neurotoxic actions (Wichers and
moderately increased relative to a single day of treatment Maes, 2004; Wichers et al., 2005). These kynurenine
(Anisman et al., in press, 2007a,b). metabolites synergistically induce free radical generation and
The dose-related effects of IFN-a have not been thoroughly have been implicated in neurodegenerative diseases, including
examined with respect to most biological processes, including Huntington’s, Parkinson’s and AIDS dementia (Hartai et al.,
brain functioning. However, it was reported that at low doses 2005).
IFN-a reduced corticosterone, but when administered directly
into brain it increased circulating corticosterone levels 11.6. Cytokine-induced behavioral changes
(Saphier, 1995). Interestingly, in the latter studies IFN-a
inhibited the activity of PVN neurons that are fundamental for Although the immunological, neurochemical and behavioral
HPA activation (Saphier, 1995). alterations elicited by pathogens may represent adaptive
Systemic IFN-a administration influences central neuro- changes to meet environmental demands (Dantzer et al.,
chemical functioning, but, once again, these effects were 1999a, 2000; Dantzer, 2001a,b; Hart, 1988; Konsman et al.,
modest compared to those evident following IL-1b adminis- 2002; Maier and Watkins, 1998), they may also elicit adverse
tration. By example, IFN-a treatment stimulated activity of the psychological consequences (Hopkins and Rothwell, 1995;
amino acid transmitters GABA and glutamate within limbic Quan and Herkenham, 2002; Rothwell and Hopkins, 1995).
and hypothalamic regions (Yokoyama et al., 2005), modestly Immune activation as well as administration of IL-1b or TNF-a
increased hypothalamic and hippocampal NE utilization typically engenders an array of behavioral symptoms (soporific
(Anisman et al., in press, 2007a,b) and reduced DA effects, ptosis, anorexia, fever, fatigue, reduced motor activity,
concentrations within the amygdala (Kitagami et al., 2003). curled body posture) referred to as ‘‘sickness behaviors’’ (Kent
With repeated treatment, IFN-a diminished DA utilization et al., 1992). These symptoms appear to be mediated, in part, by
(Shuto et al., 1997) and 5-HT levels in PFC, increased 5-HT central mechanisms, as low doses of IL-1b or TNF-a
turnover within the amygdala (De La Garza et al., 2005), and administered directly into the brain elicited these behaviors
also increased low-affinity 5-HT1A receptor sites (Abe et al., (Dantzer, 2001a,b; Konsman et al., 2002; Maier and Watkins,
1999). In line with inflammatory involvement in depression, 1998). The behavioral expression of sickness is contextually
IFN-a reduced 5-HT turnover in PFC, and the depressive dependent in the sense that the signs of malaise are suppressed
effects of the treatment were attenuated by pretreatment with an under conditions where sickness would be a disadvantage (e.g.,
NSAID (Asnis et al., 2003; De La Garza et al., 2005). in a novel or threatening environment; Kusnecov et al., 1999).
When IFN-a was administered directly into brain, fever, It was suggested that the sickness behaviors reflect
anorexia and analgesia were elicited, as were changes of depressive symptoms as the behaviors (Yirmiya et al., 1999,
hypothalamic neuronal firing (Hori et al., 1998). This treatment 2001) and elevated Fos expression (Castanon et al., 2002, 2003)
also reduced 5-HT concentrations in the PFC (De La Garza and elicited by endotoxins were attenuated by antidepressant
Asnis, 2003), increased hippocampal DA activity (De La Garza treatments. Furthermore, cytokines disrupt operant responding
and Asnis, 2003; Morikawa et al., 1998), and increased 5-HTT for food reward (Bret-Dibat et al., 1995; Kent et al., 1996) as
mRNA expression, which ought to promote reduced 5-HT well as exploration and social interaction (Bluthe et al., 1997),
availability (Morikawa et al., 1998). It is noteworthy that IFN-a which would also be expected if cytokines elicit a depressive-
administration promoted 5-HT2C receptor mRNA editing, like state. Some of these behaviors are, indeed, reminiscent of
resulting in receptor down-regulation (Yang et al., 2004). This neurovegetative features associated with major depressive
outcome may be particularly relevant, as it will be recalled that illness (e.g., reduced libido, diminished food intake, motor
5-HT2C receptor functioning has been associated with anxiety retardation), but it can be argued that they are simply a sign of
and depressive symptoms (Merali et al., 2006a), and altered 5- general malaise, rather than a reflection of depression per se.
HT2C editing was associated with depression/suicide. Of particular relevance to depression are reports that
There are several processes by which pro-inflammatory cytokines and bacterial endotoxins may elicit anhedonia, a key
cytokines may come to affect 5-HT functioning, including symptom of depressive illness, independent of any effects of
28 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

anorexia (Anisman and Merali, 1999; Anisman et al., 1998, IL-1b and TNF-a provoked a time-dependent and persistent
2002; Merali et al., 2002a). By example, endotoxin treatment increase in the co-expression of CRH and AVP within the
(lipopolysaccharide: LPS) disrupted responding for rewarding external zone of the median eminence. That is, although the co-
brain stimulation during the ascending portion of a current expression was not evident at short intervals following the
titration schedule (Borowski et al., 1998). Likewise, we have cytokine challenge, it developed over several weeks following
shown that the anorexic and motivational (anhedonic) effects of the cytokine treatment, and once evident the co-expression
cytokines (such as IL-1b) are distinguishable from one another, persisted for an extended period of time. It will be recalled that
and only the latter is attenuated by chronic antidepressant the co-release of these peptides synergistically stimulate ACTH
treatment (Merali et al., 2002a; see also Larson and Dunn, secretion from the anterior pituitary, and thus it was suggested
2001; Larson et al., 2002; as well as Sammut et al., 2001, 2002 that the development of the CRH-AVP co-expression might
for effects of interferon-a). Using a progressive ratio (PR) contribute to the depressive symptoms elicited in response to
schedule for a tasty treat, namely sucrose reinforcement (i.e., later stressor or cytokine challenges (Hayley et al., 2001b,
progressively increasing the number of operant responses that 2002; Schmidt et al., 1995; Tilders and Schmidt, 1999). This
are required for a fixed amount of sucrose, thereby providing an proposition is in line with the view described earlier (Post,
index of the motivation to work for reward), we found that IL- 1992), wherein it was suggested that the sensitization of
1b reduced both PR performance and free-chow consumption. neuronal processes, including those governing neuropeptide
However, whereas chronic antidepressant (fluoxetine) treat- release, may increase over time and/or with repeated illness
ment attenuated the PR performance deficit, it did not affect the episodes, and may play a fundamental role in the emergence of
diminished free chow consumption provoked by the cytokine, depression (Post, 1992). In this instance, however, rather than
thus indicating that the cytokine may promote anhedonia, quite being instigated by a traditional stressor, the stimulus eliciting
apart from its anorexic effects. These varied findings using the co-expression comprises an inflammatory challenge.
animal models of depression, coupled with the effects of The studies that have assessed the sensitizing effects of
antidepressants on sickness behaviors and other behavioral cytokines have typically done so in adult animals. Yet, given
disturbances associated with cytokine challenges, are consis- that early life stressors, including impoverished dam–pup
tent with a role for inflammatory factors in the provocation of a interactions, increase the stress response measured during
depressive-like state. subsequent adulthood (Meaney, 2001), the possibility was
Although cytokines may elicit effects pertinent to mood/ considered that early life endotoxin treatment might similarly
anxiety disorders (Anisman and Merali, 1999; Dantzer, 2001a; influence the adult stress response. Indeed, rat pups that had
Dantzer et al., 2000; Hayley et al., 2005, 2002a; Kent et al., been exposed to a bacterial endotoxin, when subsequently
1992; Lyte et al., 1998; Maier and Watkins, 1998; Musselman tested as adults, exhibited increased stressor-elicited corti-
et al., 2001; Reichenberg et al., 2001), and appear to involve costerone reactivity, elevated lymphocyte sensitivity to
CNS mechanisms (Bluthe et al., 1997; Dantzer, 2001a; De suppression by stressors, and protection against adjuvant-
Sarro et al., 1997; Kent et al., 1996), the pleiotropic effects of induced arthritis (Shanks et al., 2000). Whether the effects of
these cytokines (such as sickness) often complicate analysis of early life stressors and early life cytokine treatments involve
these effects. It is questionable, however, whether analyses of similar neural circuits is uncertain. Nevertheless, these
sickness itself could be considered as an analogue of depressive findings raise the possibility that early life immunological
illness. Assessment of the effects of cytokines on behavioral insults, like other stressors, might by virtue of greater stressor
outputs, with the intent of simulating depressive illness, would sensitivity or reactivity, increase vulnerability to pathological
be better positioned if they focused on specific features that, in states.
fact, bore greater face validity than sickness behaviors.
11.8. Cytokines in relation to depression in humans
11.7. Proactive effects of cytokines and stressors
Human studies have provided considerable evidence in favor
Paralleling the effects of stressors described earlier, cytokine of the view that cytokines contribute to the evolution of major
administration increased the response to later challenge with depression. Correlational studies have indicated that depressive
either a stressor or the cytokine treatment (Anisman et al., 2003; symptoms occurred fairly frequently in physical illnesses
Hayley et al., 1999b, 2001c, 2002b; Schmidt et al., 1995), and, associated with inflammation and with elevated IL-1 levels
conversely, stressor exposure augmented the neuroendocrine (Dantzer et al., 1999b). As well, there have been reports that
response elicited by later LPS administration (Johnson et al., depression was accompanied by aspects of an acute phase
2002, 2003). For instance, stressors increased central IL-1b response, such as elevated complement protein levels (C3 and
levels and also enhanced the effects of later central LPS C4) as well as positive acute phase proteins, haptoglobin, a1-
administration. However, if rats were pretreated with the IL-1 antitrypsin, a1 and a2 macroglobulin and C-reactive protein
antagonist, IL-1ra, then the effects of the subsequent stressor (CRP), whereas negative acute phase proteins were reduced
treatment were attenuated (Johnson et al., 2004). (Danner et al., 2003; Miller et al., 2002). Furthermore,
As described earlier, variations of CRH and AVP may be depression was associated with an increase of activated T cells
fundamental to the sensitization associated with cytokine (CD25+ and HLA-DR+), and secretion of neopterin, prosta-
treatment. Specifically, it was demonstrated that like stressors, glandin E2 and thromboxane (Maes, 1995, 1999).
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 29

In addition to these indices of inflammation, it has been features (reduced eating and weight loss, reduced sleep) evident
reported that circulating and mitogen-provoked cytokine in typical major depression could promote altered cytokine
concentrations were elevated in major depressive disorder, functioning. In fact, it has been known for some time that sleep
especially in relatively severe (melancholic) illness (Maes, disturbances are associated with cytokine variations (Moldofsky,
1999). The cytokine elevations included IL-2 and its soluble 1995), and disturbances of circadian cycles that may be evident
receptors (sIL-2R), IL-1b, IL-1 receptor antagonist (IL-1Ra), among depressed individuals may similarly influence cytokine
IL-6, sIL-6R, and IFN-g. As well, it was reported that the production (Cover and Irwin, 1994). Finally, any of the many
normal IL-6 circadian rhythm was disturbed in individuals comorbid features of depression, such as anxiety, heart disease,
diagnosed with major depressive disorder (Alesci et al., 2005). diabetes and Parkinson’s disease (McDonald et al., 2003), could
Moreover, there have been indications that the elevated levels potentially influence circulating cytokine levels.
of IL-1b, IL-6, TNF-a, and IFN-g normalized with anti-
depressant medication (Basterzi et al., 2005; Sluzewska et al., 11.9. Immune activation provokes depressive mood state
1995; Tuglu et al., 2003). In contrast to these reports, however,
it was also found that the elevated production of sIL-2R, IL-6, The studies showing that depressive illness is accompanied
IL-10 and sIL-6R in severe depression (Maes, 1999; Sluzewska by variations of inflammatory processes comprise correlational
et al., 1995) and the elevated IL-1b associated with chronic low data and do not speak to the causal role of cytokines or acute
grade depression (dysthymia) (Anisman et al., 1999a,b) were phase reactants to the provocation or exacerbation of
not attenuated with antidepressant treatments. The fact that depression. However, two lines of research have suggested
most cytokines did not normalize with attenuation of clinical that cytokines (or other inflammatory factors) are causally
symptoms suggests that these cytokines are not causally linked related to this illness: (1) immune activating agents (e.g.,
to depression, but might serve as trait markers of the illness. Of vaccines or endotoxin treatment) administered to healthy adults
course, the possibility should not be dismissed that cytokine may elicit depressive-like behaviors in direct relation to TNF-a
normalization might only be evident with sustained treatment, and IL-6 secretion associated with the treatment (Reichenberg
or that cytokines normalize in only a subset of individuals, with et al., 2001; Wright et al., 2005) and (2) immunotherapy (using
elevated levels being a harbinger for illness recurrence. IFN-a in the treatment of some forms of cancer and hepatitis C)
Few studies have considered that subtypes of the illness, may be associated with the induction of depressive-like states.
particularly typical vs. atypical depression, may be associated There have now been an appreciable number of studies
with different cytokine profiles, despite the possibility that showing that individuals treated with IFN-a (and IL-2 in earlier
these illness subtypes may involve different underlying studies; Denicoff et al., 1987) displayed cognitive disturbances,
processes. It was reported, however, that T cell proliferation retarded cognitive processing and impaired executive function-
was diminished in typical relative to atypical depression ing, impaired concentration and memory, irritability, and anxiety,
(Zaharia et al., 2000), but no such effects were apparent in IL- as well as manic periods (Constant et al., 2005; Dieperink et al.,
1b production (Anisman et al., 1999a). Nevertheless, it has 2003; Hilsabeck et al., 2005; Lieb et al., 2006; Meyers, 1999;
been suggested that cytokine activation might contribute Meyers and Valentine, 1995; Pavol et al., 1995; Sceibel et al.,
selectively to some of the symptoms that comprise the 2004; Valentine et al., 1995; Valentine and Meyers, 2005). There
depressive profile (e.g., IL-1b contributes to impaired appetite have also been studies that failed to detect such outcomes
associated with depression; Andreasson et al., 2007). (Caraceni et al., 1998; Mapou et al., 1996), possibly reflecting
The finding that elevated cytokine levels accompany between study differences in the dosage and treatment regimens
depressive illness has been taken as support for some form of employed. The latter studies notwithstanding, it has been
relation (although not necessarily causal) between these factors. reported that IFN-a reduced frontal lobe electrophysiological
Yet, as described by Irwin (1999), processes not directly related activity (Rohatiner et al., 1983; Smedley et al., 1983), and the
to the mood alterations associated with major depression may increase of depressive symptoms elicited by the cytokine were
have promoted the altered cytokine levels. Among other things, it accompanied by reduced frontal cortical functioning measured
was suggested that depression may be associated with several by PET analysis (Juengling et al., 2000).
antecedent conditions, including recent stressful experiences, Particularly troublesome from a clinical perspective was that
stressors encountered early in life, and altered appraisal and IFN-a engendered pronounced depressive symptoms (Capuron
coping with respect to ongoing stressors, any one of which could and Ravaud, 1999; Capuron et al., 2000, 2001, 2002a,b,c,
have influenced cytokine levels. Further, institutionalization (and 2003a,b, 2005; Caraceni et al., 1992; Maes and Bonaccorso,
the accompanying social and environmental change) of 2004; Maes et al., 2001; Miyaoka et al., 1999; Musselman et al.,
depressed patients may itself have contributed to cytokine 2001; Scalori et al., 2005; Wichers and Maes, 2002, 2004). An
variations, as would an increase of drug use (including nicotine, appreciable portion of patients reported suicidal ideation
alcohol, as well as illicit drugs). Indeed, Haack et al. (1999) (Dieperink et al., 2000) and, in some, the symptom severity
reported that when they statistically controlled for age, body necessitated discontinuation of therapy (Valentine and Meyers,
mass index, gender, smoking habits, recent infection or prior 2005). Although, depressive symptoms ordinarily resolved with
medication, differences of circulating cytokine levels between treatment cessation (Loftis and Hauser, 2004), patients were
controls and depressed patients were absent. Further to this issue, sensitized to the treatment, becoming highly reactive to further
it is not unreasonable to hypothesize that neurovegetative cytokine administration.
30 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Just as MDD may comprise both neurovegetative (e.g., improve tolerability to the immunotherapeutic side effects, these
lethargy, somnolence, psychomotor retardation) and cognitive/ findings are of practical as well as theoretical importance.
mood symptoms, IFN-a elicits each of these symptom In addition to altered 5-HT functioning, other processes,
dimensions, and high doses may provoke a confusional state including actions on neuropeptide and growth factor as well as
characterized by disorientation, as well as psychotic-like on inflammatory processes, are also likely involved in
features (Asnis and De La Garza, 2006; Raison et al., 2005a,b). determining the effects of SSRIs. It was reported, for instance,
Over the course of treatment, depressive symptoms and that fluoxetine reduced expression of IL-1b, IL-6, IFN-g and
cognitive impairments (e.g., disturbances of immediate recall) TNF-a, and increased the circulating levels of the inhibitory
may become increasingly pronounced (Capuron et al., 2005; cytokine IL-10 (Chiou et al., 2006). It was similarly observed
Lieb et al., 2006; Raison et al., 2005b). In addition, IFN-a that amitriptyline, clomiprimine, imiprimine, and bupropion,
immunotherapy may initially engender disturbed vigilance, all diminished IL-1b and TNF-a release provoked by LPS
alertness and some memory problems, which diminish with (Brustolim et al., 2006; Obuchowicz et al., 2006) and had potent
treatment cessation (Kraus et al., 2005a,b,c,d). Despite the link anti-inflammatory effects, including suppression of macro-
with depression and immunotherapy, given the non-specificity phage migration (Bianchi et al., 1994). These actions were not
of symptoms (reflected by the wide array of disturbances restricted to SSRIs, as another class of compounds with
associated with IFN-a immunotherapy), the obvious question antidepressant effects, namely the phosphodiesterase IV
arises as to whether the effects observed are a genuine inhibitors (e.g., rolipram), also diminished cytokine production
manifestation of neurochemical changes that underlie depres- in response to LPS (Griswold et al., 1998; Zhu et al., 2001).
sion, or whether the constellation of symptoms is more a Centrally, hippocampal and brainstem levels of TNF-a were
reflection of general malaise or toxicity. reduced by desimipramine (Reynolds et al., 2004, 2005), and
Contrary to the latter view, however, it seems that the the clinical efficacy of this compound was linked to its ability to
development of depression following IFN-a therapy is linked to alter the sensitivity of NE neurons to TNF-a (Reynolds et al.,
some of the same characteristics that predict MDD. Specifi- 2005). Paralleling the effects of antidepressants upon cytokines,
cally, the presence of subsyndromal levels of depression prior to administration of 5-HT itself reduced TNF-a and IFN-g
immunotherapy predicted major depressive symptoms in expression, and it is significant that this outcome could be
response to the IFN-a (Beratis et al., 2005; Capuron et al., reversed by CRH treatment (Maes et al., 2002). As such, these
2001, 2004) as did the early signs, such as sadness, pessimistic findings suggest that cytokines and 5-HT neuronal activity, as
thoughts and sleep disturbances (Capuron and Ravaud, 1999). well as CRH, might reciprocally influence one another, and
As well, poor social support at baseline was also related to the might thus come to influence depressive mood.
emergence of depressive symptoms following IFN-a treatment If inflammatory factors contribute to depressive sympto-
(Capuron et al., 2004). Interestingly, like the mood changes matology, it would be expected that anti-inflammatory
associated with depletion of the 5-HT precursor tryptophan compounds might themselves possess antidepressant proper-
(Neumeister et al., 2004; Young and Leyton, 2002), suscept- ties, or at least enhance the effects of conventional
ibility to the depressive symptoms elicited by IFN-a were antidepressants. In this regard, cyclooxygenase-2 (COX-2)
greater among women and among those with a history of antagonists (non-steroidal anti-inflammatory drugs) were found
depression (Capuron et al., 2003a). It also appeared that low to attenuate the depressive-like behavioral and neurochemical
levels of tryptophan were predictive of depressive illness alterations associated with both LPS and IFN-a treatment in
(Capuron et al., 2003a) as were the initial elevations of ACTH animal models of depression (Asnis et al., 2003; De La Garza
and cortisol in response to IFN-a (Capuron et al., 2003b). et al., 2004), and such treatments also diminished glucocorti-
Along these same lines, individuals with the highest baseline coid responses to social stressors (Bugajski et al., 2003). As
levels of sIL-2r, IL-6, and IL-10 were most prone to depressed well, pre-treatment with the COX-2 antagonist indomethacin
mood following immunotherapy (Wichers et al., 2006). prevented the inhibitory effects of LPS upon ambulatory
Together, these findings suggest that the depression that locomotion, rearing and grooming, and also diminished the
emerges following immunotherapy is, in fact, tied to 5-HT and endotoxin’s effect on Fos immunoreactivity within 5-HT
cytokine related processes. neurons (Hollis et al., 2006). As expected, a COX-2 inhibitor
Consistent with this perspective, several investigators enhanced the effectiveness of antidepressant medication in
reported that SSRIs, such as sertraline, paroxetine and treating depression (Muller et al., 2006), but the usefulness of
citalopram, attenuated the depressive symptoms provoked by combining NSAIDs with antidepressant medication are limited
IFN-a (Kraus et al., 2005c; Musselman et al., 2001; Maddock by the increased risk of gastrointestinal disturbances that may
et al., 2004; Schramm et al., 2000). Moreover, SSRI treatment come about (de Jong et al., 2003).
could be used prophylactically to limit the emergence of
depressive symptoms (Kraus et al., 2005b; Schaefer et al., 2005). 11.10. Mechanisms linking cytokine activation and
It seems that the neurovegetative and mood-related features depressive illness
introduced by IFN-a therapy are independent of one another, as
paroxetine primarily affected the mood-related symptoms, with The processes by which anti-inflammatory agents modulate
only minor effects on fatigue and anorexia (Musselman et al., affective processes is a matter of ongoing research. Simply
2001; Raison et al., 2005a,b). Given that antidepressants may because SSRIs attenuate the adverse effects of cytokines cannot
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 31

be taken as definitive evidence of direct 5-HT involvement, neurogenesis otherwise reduced by endotoxin treatment or
particularly given the multiple downstream effects of these by irradiation (Monje et al., 2003). Likewise, over-expression
treatments. Nevertheless, in view of the factors associated with of IL-6 or administration of LPS inhibited hippocampal
depression proneness in IFN-a treated patients (e.g., tryptophan neurogenesis, and several anti-inflammatory treatments,
levels), it is likely that 5-HT contributes directly or indirectly to including minocycline and indomethacin, prevented this
the outcomes observed. It was suggested (as depicted in Fig. 1) outcome (Monje et al., 2003; Vallieres et al., 2002). Together,
that these immunotransmitters, like stressors, provoke the these findings raise the possibility that cytokine actions on
release of hypothalamic CRH, which then stimulates ACTH depressive states stem from effects on neuroplasticity.
and corticosterone secretion. Concurrently, release of amygdala An alternative process by which cytokines may influence
CRH triggered by pro-inflammatory cytokines, alone or in depressive disorder is through NFkB. This transcription factor
combination with GABA activation, leads to forebrain and is fundamental in the mobilization of inflammatory chemokines
hippocampal 5-HT release, and either the CRH or the 5-HT and lymphocyte proliferative responses to both infection and
alterations may elicit anxiogenic and/or depressive effects. traumatic injury (Lucas et al., 2004; Poynter et al., 2004).
As alluded to earlier, cytokines may promote depressive Ordinarily, NFkB, which is composed of five subunits, is held
states through any of several different neuronal processes (and in an inactive state by an endogenous inhibitory factor, IkB.
these are not mutually exclusive). Among its other effects, it Inflammatory stimuli trigger the phosphorylation and degrada-
will be recalled that IFN-a increases IDO, which is responsible tion of IkB, and then the translocation of NFkB to the nucleus
for the catabolism of tryptophan to kynurenine (Menkes and where it promotes gene expression (Lucas et al., 2004). There is
MacDonald, 2000). Thus, the IFN-a elicited increase of IDO reason to believe that NFkB contributes to CNS processes that
may diminish tryptophan availability, thereby reducing 5-HT are fundamental for neuronal survival and plasticity, and may
synthesis. In line with this view, it was reported that depression be involved in memory processes (Mattson et al., 2004). In
induced by IFN-a was accompanied by reduced tryptophan particular, within the hippocampus, NFkB expression is
coupled with elevated levels of kynurenine (Bonaccorso et al., increased during activation of neuroplastic processes, such
2001, 2002). It is tempting to conclude that depression elicited as the induction of long-term potentiation, and it has been
by IFN-a was, indeed, related to diminished levels of suggested that this transcription factor may be involved in
tryptophan. However, as indicated by Wichers and Maes promoting neuronal survival following hippocampal injury
(2004), kynurenine metabolites, such as 3-hydroxy-kynurenine (Kassed and Herkenham, 2004; Freudenthal et al., 2004).
and quinolinic acid, through their effects on reactive oxygen The neuroprotective action of NFkB may stem from the
species or stimulation of hippocampal NMDA receptors, may induction of anti-apoptotic proteins, such as bcl-2, as well as the
engender neuronal cell loss that could favor the development of antioxidant enzyme manganese-superoxide dismutase (Matt-
depression. Finally, other neurotransmitters, including NE, may son et al., 2004). At the same time, it is possible that NFkB
play a fundamental role in subserving the mood regulatory signaling may also result in the synthesis of inflammatory
actions of cytokines. In fact, it was posited that increased cytokines, reactive oxygen species, and excitotoxins that
central TNF-a levels might provoke depressive illness by contribute to neurodegeneration (Mattson et al., 2004; Pahan
activation of adrenergic autoreceptors leading to a subsequent et al., 2001), and as such NFkB may be an important down-
reduction of NE neurotransmission (Reynolds et al., 2005). stream signaling factor for cytokines that contribute to
In addition to changes of monoamine activity, cytokines may pathologies, such as Parkinson’s disease (PD), MS and MDD
influence the development of depressive pathology by affecting (Barger et al., 2005). For instance, IL-1b infusion into the
processes aligned with neuroplasticity, such as neurogenesis lateral ventricles induced the translocation of NFkB to the
(Hayley et al., 2005). Indeed, among transgenics with elevated nucleus at several brain regions distal to the site of infusion,
IL-6 levels, neurogenesis was markedly reduced (Vallieres including the choroid plexus, ependymal cells, cerebral
et al., 2002). As well, when IFN-a was chronically vasculature and meninges as well as at the basolateral
administered to rodents (in a schedule mimicking an amygdala (Konsman et al., 2000). It is thought that some of
immunotherapeutic protocol), hippocampal neurogenesis was these changes, including the variations of NFkB, may be
diminished, and this outcome was attenuated by the IL-1 instrumental in promoting or sustaining depressive-like
antagonist IL-1ra (Kaneko et al., 2006). To be sure, this does symptoms (Miller and Raison, 2006; Raison et al., 2006). As
not necessarily imply a causal relation between cytokine well, there is reason to believe that NFkB might contribute to
immunotherapy, neurogenesis and depression. Nevertheless, anxiety, as mice deficient for the p50 subunit of NFkB
animal studies have shown that psychogenic stressors may (resulting in impaired NFkB activity) displayed reduced
promote death of cortical neurons through apoptotic mechan- anxiety (Kassed and Herkenham, 2004). In effect, immuno-
isms, and that IL-1b as well as the TNF-a family member Fas logical insults and the ensuing IL-1b and TNF-a activation may
were involved in this regard (Lee et al., 2006a,b). In line with initiate the NFkB cascade (Lucas et al., 2004), ultimately
this perspective, as will be discussed shortly, antidepressants affecting CNS processes.
elevated neurogenesis and prevented the effects otherwise Summarizing, although the primary role of the inflammatory
engendered by chronic stressor exposure (Chiou et al., 2006). immune system is that of protecting the individual from
Furthermore, attenuating the inflammatory response through bacterial and viral insults, there is considerable evidence
indomethacin administration had the effect of restoring supporting the view that cytokines may also be involved in the
32 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

provocation of depressed mood and anxiety. Like stressors, The morphological changes associated with MDD could
cytokines influence HPA functioning, as well as monoaminer- come about owing to genetic factors (Gass et al., 2000),
gic and peptidergic processes at hypothalamic and extra- distressing early life events, or chronic strain (McEwen, 1999;
hypothalamic sites, and may influence neurogenesis. The Brunson et al., 2001; MacQueen et al., 2003; McEwen, 2005;
notion that cytokines are directly or indirectly involved in MDD Vythilingam et al., 2002, 2004), but it is equally possible that
is reinforced by the findings that certain cytokines, notably structural changes may be secondary to illness. In this regard,
IFN-a, promote the appearance of MDD, and these effects are caution should be exercised in the interpretation of imaging
attenuated by antidepressant medication. As will be seen in studies given the observation that (a) large variations of
ensuing sections, MDD has also been linked to changes of hippocampal volume exist within the general population and
growth factors and neurogenesis, and the case will be made that (b) the difference in volume between individuals in the lowest
inflammatory factors may influence depressive illness by quartile compared to the mean of a sample of young healthy
modifying these processes. adults exceeded the difference observed between healthy adults
and depressed subjects (Lupien et al., 2007).
12. Morphological correlates of major depressive illness
12.1. Stressor and corticoid actions in relation to
Major depressive disorder has been associated with both morphological changes associated with MDD
structural and functional changes within discrete brain
regions, including limbic regions such as the hippocampus Clinical depression may be related to the direct actions of
and the amygdala (Drevets et al., 2007; Videbech and glucocorticoids, which are frequently elevated in relatively
Ravnkilde, 2004). It was reported that hippocampal volume severe cases of depression or in response to chronic stressors
was reduced in patients diagnosed with major depression, and (Sapolsky et al., 1985), and the proposition was advanced
postmortem analyses indicated that the extent of the that glucocorticoids might contribute to the reduced
hippocampal reduction was related to illness duration hippocampal volume associated with MDD. Consistent with
(Bremner et al., 2000; Campbell et al., 2004; Kempermann this view was the finding that impaired hippocampal
et al., 2003; Sheline et al., 2003; Videbech and Ravnkilde, neurogenesis occurred in rodents exposed to a chronic
2004). Consistent with the view that pre-existing morpho- corticosterone or stressor regimen (Coyle and Duman, 2003).
logical changes might contribute to MDD, it was reported As well, in monkeys, hippocampal implantation of cortisol
that the reduction of hippocampal volume was evident at the pellets for an extended period (1 year) engendered
time of the first depressive episode in relatively young morphological changes in the hippocampal CA3/CA2
individuals (Frodl et al., 2002). Yet, it has also been reported subfields, including irregular cell layers, soma shrinkage
that hippocampal reductions were most apparent after and dendritic atrophy (Sapolsky et al., 1990). Furthermore, in
repeated depressive episodes and that the magnitude of the rats, intermittent stressor (restraint) exposure promoted
reduction was positively associated with illness duration hippocampal neuronal loss, coupled with increased expres-
(MacQueen et al., 2003). Despite this strong evidence for sion of the degenerative marker tau protein (Stein-Behrens
morphological changes being associated with MDD, reduced et al., 1994). The hippocampal cell loss was not only
hippocampal volume was also associated with other apparent in response to physical stressors but was also
pathologies, such as schizophrenia (Boos et al., 2007), and elicited by psychosocial disturbances. That is, reduced apical
alterations of the right caudate nucleus were not only dendrite length of hippocampal pyramidal neurons and fewer
associated with depression, but were evident in association dendritic branching points were evident among subordinate
with anxiety, obsessive compulsive disorder (OCD) and male tree shrews chronically exposed to a dominant animal
paranoia (Starkman et al., 2007). Thus, it is possible that (Magarinos et al., 1996). Such effects were not confined to
these morphological alterations may be a predisposing the hippocampus, as a chronic restraint stressor regimen also
influence for pathological states in general, as opposed to reduced apical dendritic branching and length in pyramidal
being uniquely linked to depressive illness. PFC neurons (Cook and Wellman, 2004).
This caveat not withstanding, paralleling studies that There are several processes by which glucocorticoids could
implicated 5-HTT in depression, it was reported that reduced affect cell survival. For instance, corticoids could impair
hippocampal volume was most pronounced in MDD patients glucose transport into hippocampal pyramidal neurons, hence
carrying the long variant of the 5-HTT allele (Frodl et al., 2004). promoting an energetic compromise of these cells (McEwen,
Furthermore, the hippocampal volume was lower in non- 2000, 2005). It is similarly possible that glucocorticoids might
remitted patients than among those in remission (Frodl et al., potentiate elevations of free cytosolic calcium, and hence may
2004), and this was especially notable within the left either directly impair neuronal survival or render cells
hemisphere (Mervaala et al., 2000; MacMaster and Kusumakar, vulnerable to other insults. As well, excessive corticoid levels
2004; Vakili et al., 2000). Still, it is possible that the reduced may contribute to free radical accumulation, largely by
hippocampal volume is not a consequence of MDD, but instead reducing the capacity of antioxidant enzymes (Sapolsky,
represents a risk factor for the illness, just as small hippocampal 2000). Furthermore, neuronal survival could be influenced
volume may function in this capacity in relation to PTSD through excitotoxic processes involving elevations of NMDA
(Gilbertson et al., 2002; Pitman et al., 1999). and/or AMPA glutamate receptors (McEwen, 2000, 2005).
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 33

13. Growth factors in relation to stressors and anxiety was associated with reduced hippocampal BDNF
depression expression (Yee et al., 2007), and transgenic mice over-
expressing this neurotrophin displayed elevated anxiety
As already indicated, depression is likely mediated by (depression and anxiety are frequently co-morbid) (Govindar-
several concomitant neurochemical disturbances, and effective ajan et al., 2006). Moreover, BDNF infusion into the DRN
treatment strategies ought to consider the interconnected provoked an antidepressant-like effect while concomitantly
central pathways involved. There have been reports indicating promoting sprouting of central 5-HT neurons (Altar, 1999).
that in addition to monoamine and neuropeptide variations, Likewise, hippocampal infusion of BDNF or NT-3 produced
stressors may influence neurotrophic growth factors (Duman antidepressant-like effects in the learned helplessness and
and Monteggia, 2006; Duman et al., 1999; Manji and Duman, forced swim models of depression (Shirayama et al., 2002).
2001). Moreover, their functions may be related to one another Interestingly, antidepressant treatment was associated with
as well as to 5-HT activity (Hayley et al., 2005; Manji et al., increased hippocampal BDNF, whereas variations of neuro-
2003). As 5-HT and BDNF reciprocally innervate one another trophic factors in the amygdala appeared to be more closely
(Mattson et al., 2004), the possibility exists that this process aligned with anxiety (Govindarajan et al., 2006). In effect,
might influence depression. depression may be uniquely related to hippocampal BDNF
variations, whereas other features of the illness may involve
13.1. BDNF involvement in depression other brain regions or other processes. It will be noted that in
addition to being implicated in depression, it has been reported
Consistent with a role for BDNF in depressive illness, that BDNF present in DA neurons contributes to the normal
stressors were shown to reduce the expression of BDNF in expression of D3 receptors in the nucleus accumbens, and may
limbic regions that mediate mood states (Duman and be responsible for behavioral sensitization by triggering
Monteggia, 2006; Manji et al., 2001) and that BDNF variations overexpression of D3 receptors in striatum of hemiparkinsonian
may be responsible for the hippocampal atrophy associated rats (Guillin et al., 2001). As such, BDNF may contribute to
with depression (MacQueen et al., 2003). Conversely, treatment pathology involving altered DA functioning.
with antidepressant agents had the effect of increasing BDNF Studies concerning the impact of acute and chronic stressors
expression and attenuating the effects of stressors (Jacobs, (including chronic mild stressors) indicated that aversive events
2002; Manji et al., 2003). The present review focuses on the reduced hippocampal BDNF levels (Smith et al., 1995; Song
potential contribution of BDNF in depression, but this should et al., 2006), and that such effects could be induced in response
not be taken to exclude a role for other growth factors, including to a psychological stressor (explicit or contextual cues
FGF1 and FGF2, that are influenced by stressors and have also previously paired with footshock) (Rasmusson et al., 2002).
been implicated in the evolution of depressive states (Evans In addition to the variations evident within the hippocampus, a
et al., 2004; Newton and Duman, 2004; Turner et al., 2006). neurogenic stressor (tailshock) increased BDNF mRNA
Analyses of signaling processes involved in depression have expression within the anterior cingulate gyrus (Bland et al.,
increasingly focused upon alterations of trophic factors that 2007), a region important for the processing of emotionally
might lead to deficiencies of neuroplasticity and the subsequent salient events. However, following exposure to a predator
provocation of depressive pathology (Manji et al., 2001). In line stressor, BDNF mRNA was reduced within the CA1 region of
with a role for BDNF in depression, it was reported that BDNF the hippocampus and simultaneously up-regulated expression
expression and protein levels and that of its receptors (tyrosine of its TrkB receptor (Kozlovsky et al., 2007). It is interesting
kinase B, TrkB) were lower within the PFC and hippocampus of that with chronic exposure to a uniform (homotypic) stressor
suicides than in age- and sex-matched controls (Dwivedi et al., (restraint) the magnitude of the BDNF reduction in hippo-
2003). Moreover, antidepressant treatment was found to elevate campus was less pronounced than after acute treatment
cortical BDNF levels and enhance TrkB signaling and these (Murakami et al., 2005), suggesting some degree of adaptation.
effects were required for the induction of the behavioral effects Yet, exposure to a chronic unpredictable stressor over several
of the antidepressant (Saarelainen et al., 2003). As well, BDNF weeks reduced BDNF protein within the dentate gyrus but
levels were reduced in the serum of depressed patients and the surprisingly did not affect levels of the growth factor within the
reduction correlated negatively with the degree of clinical hippocampus proper (Grønli et al., 2006). It is uncertain
impairment, as well as reductions of hippocampal volume whether the between-laboratory differences were due to the
(Shimizu et al., 2003). As would be predicted, chronic chronicity of the stressors, the stressor schedule employed, or
antidepressant treatment normalized BDNF serum levels in effects that were unique to some types of stressful events.
parallel with clinical improvement (Piccinni et al., 2007). As would be expected, several clinically beneficial
However, it was also reported that reduced serum BDNF levels treatments, including SSRIs, tricyclics, and electroconvulsive
occurred in depressed females, but not males, and that chronic therapy, increased hippocampal BDNF expression (Shirayama
antidepressant treatment selectively elevated the growth factor et al., 2002; Castrén et al., 2007; Rogóz et al., 2007). Likewise,
in females (Huang et al., 2007), suggesting that the role of the reduced BDNF associated with restraint was prevented by
BDNF in depression may be gender specific. antidepressant treatment (Duman et al., 1999), and up-
Consistent with the contention that BDNF is involved in regulation of BDNF expression was also associated with
depression and anxiety, it has been reported that elevated improved performance among rats in the forced swim test (i.e.,
34 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

reduced immobility), a paradigm that has been used to screen did not influence the initial proliferation or differentiation of
antidepressant agents (Shirayama et al., 2002). More recently, new-born cells, but diminished their survival (Lee et al.,
the atypical antidepressant tianeptine was also shown to elevate 2006a,b).
BDNF expression within the amygdala, but curiously had no In keeping with the view that neurogenesis is associated with
such effect within the hippocampus (Reagan et al., 2007). depression, the antidepressant effects of electroconvulsive
There is evidence that the efficacy of antidepressant shock were accompanied by increased generation of new
treatments is related to their ability to promote central BDNF hippocampal neurons in adult non-human primates (Perera
signaling. For instance, inhibition of BDNF functioning, using et al., 2007). Similarly, several antidepressants enhanced
a conditional knockout manipulation, attenuated the antide- neurogenesis in rodents and it was suggested that these newly
pressant actions of desipramine in the forced swim test born neurons might contribute to affective and cognitive
(Monteggia et al., 2007). However, it seems that the impact of processes (Warner-Schmidt and Duman, 2006). Indeed,
antidepressants upon growth factor expression may be inhibition of neurogenesis by selective irradiation of the
dependent upon the physical or metabolic state of the animal. hippocampus also blocked the antidepressant effects of
Although fluoxetine elevated central BDNF expression in fluoxetine and imiprimine (Santarelli et al., 2003), suggesting
physically active rats, the antidepressant actually reduced that neurogenesis may be fundamental in determining the
mRNA for the growth factor in sedentary animals (Greenwood functional consequences of antidepressant treatment. In effect,
et al., 2007). It also appeared that BDNF expression was antidepressant treatments may elevate mood, at least in part,
dependent on estrus cycle, being reduced when estradiol levels through their pro-mitotic neuronal effects. It may be that the
were high, and could also be diminished by exogenous estradiol enhanced neurogenesis stems from activation of growth factor
treatment. Of particular interest, however, was the finding that signaling (e.g. BDNF) following antidepressant exposure, but it
estradiol could attenuate the up-regulation of BDNF elicited by has been suggested that enhanced 5-HT neurotransmission may
a 5-HT2A agonist treatment (Cavus and Dman, 2003). Together, also support such processes (Djavadian, 2004; Ueda et al.,
these findings suggest complex interactions between sex 2005).
hormones, 5-HT receptors and BDNF, and could potentially The role of 5-HT in supporting neurogenesis appears to be
help explain the sex differences that have often been noted in controversial as recent reports indicated that 5-HT depletion
illnesses, such as depression. alone did not alter neurogenesis, whereas co-depletion of both
5-HT and NE effectively reduced neurogenesis (Jha et al.,
13.2. Neurogenesis, growth factors and depression 2006). Moreover, hippocampal neurogenesis did not differ
between 5-HTT deficient mice, which display chronically
An increasing number of neurotrophic peptides, including elevated synaptic 5-HT levels, relative to non-transgenic mice
BDNF, glial derived neurotrophic factor (GDNF), FGF, and (Schmitt et al., 2007). Interestingly, neurotoxin induced
leukemia inhibitory factor (LIF), have been shown to promote depletion of 5-HT was found to reduce the immediate elevation
or influence neurogenesis (Bauer et al., 2007). For instance, of neurogenesis associated with environmental enrichment, but
intracerebroventricular infusion of GDNF enhanced neurogen- did not influence the number of new neurons measured weeks
esis within the hippocampus, and administration into the later (Ueda et al., 2005). Hence, 5-HT may be important for
striatum promoted neurogenesis within the sub-ventricular birth of new neurons stimulated by enrichment, but this amine
zone (Kobayashi et al., 2006). As well, administration of novel might not directly contribute to the survival and maturation of
agonists of the BDNF receptor TrkB enhanced neurogenesis these cells.
and promoted neuronal survival and neurite growth (Qian et al., Although stressor-induced glucocorticoid elevations can
2006). Likewise, the neurotrophic cytokine granulocyte potently inhibit neurogenesis (Manji et al., 2003), factors other
colony-stimulating factor (G-CSF) influenced both hippocam- than these hormones may also contribute to the observed effects
pal and sub-ventricular neurogenesis (Schneider et al., 2005). in depression. In this regard, chronic electroconvulsive shock
In contrast to these hippocampal effects, over-expression of LIF treatment was associated with increased neurogenesis, even in
caused self-renewal of neural stem cells, coupled with a the presence of elevated corticoid levels (Hellsten et al., 2002).
reduction of neurogenesis within the olfactory bulb and Moreover, the SSRI fluoxetine normalized hippocampal
subventricular zone (Bauer and Patterson, 2006). neurogenesis in animals exposed to inescapable electric
Disturbances of hippocampal neurogenesis may contribute footshock, independent of any actions upon corticosterone
to the primary or secondary symptoms associated with (Malberg and Duman, 2003). It may be that neuropeptides,
depression. As already indicated, several different types of rather than glucocorticoids, contribute to the modulation of
stressors may inhibit neurogenesis, and it seems that the neurogenesis in stressor associated pathologies. Indeed,
controllability of the stressor may be important in this respect. normalization of reduced neurogenesis associated with a
In fact, although uncontrollable tailshock reduced FGF levels chronic mild stressor was elicited by CRH1 or AVP1b receptor
and impaired hippocampal neurogenesis, when rodents had antagonists (Alonso et al., 2004). Other peptides, such as
some degree of control over termination of the shock, FGF pituitary adenylate cyclase-activating polypeptide (PACAP)
levels actually increased and neurogenesis was unaltered and vasoactive intestinal peptide (VIP), might also contribute to
(Bland et al., 2006). It may be that stressors influence specific processes similar to neurogenesis, including astrogenesis and
stages of the cell cycle, as exposure to a series of mild stressors dendritic branching (Nishimoto et al., 2007; Henle et al., 2006).
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 35

The appearance of growth factors following ischemic injury sion of the prototypical apoptotic initiators Fas, p53, Bax and
influenced hippocampal neurogenesis within the subventricular downstream caspases in clinical depression. Yet, animal studies
zone (Wang et al., 2004; Lee et al., 2007). By example, the indicated that stressor exposure reduced expression of the anti-
infusion of GDNF following ischemia stimulated the prolif- apoptotic factor bcl-2 (Manji and Chen, 2002; Manji et al.,
eration and maturation of subventricular progenitor cells, which 2003), and exposure to a severe stressor exacerbated infarct size
eventually migrate into the striatum and become mature in response to ischemia through the suppression of bcl-2 (Manji
neurons (Kobayashi et al., 2006). Similarly, in addition to et al., 2003). The suggestion has even been made that
attenuating neuronal degeneration following cerebral ischemia, experimental treatments modulating bcl-2 expression may be
G-CSF markedly increased hippocampal double labeling of the effective for the treatment of depressive disorder (Duman,
proliferative marker BrdU, together with the mature neuronal 2004). Although frank neurodegeneration is probably not the
marker NeuN (Sehara et al., 2007; Solaroglu et al., 2007). It is primary factor subserving clinical depression, as will be
likely that enhanced hippocampal or subventricular neurogen- discussed shortly, it might contribute to several comorbid
esis serve as an adaptive response to compensate for damaged features of this illness.
neurons or function in a protective capacity to limit future As alluded to earlier, depression is highly co-morbid with
injury. several neurological conditions, including PD, AD, stroke and
Just as antidepressants influence growth factor expression, MS (McDonald et al., 2003; Ramasubbu, 2000). In this context,
such treatments may limit the reduction of neurogenesis depressive illness may interact or co-occur with neurodegen-
ordinarily associated with stressor exposure or neural injury. In erative pathology because they share underlying mechanisms
parallel with the attenuation of the deficit of escape (e.g., inflammatory cytokines). Alternatively, the aetiological
performance provoked by the stressful treatment, fluoxetine processes leading to depression (e.g., stressor experiences)
blocked the down-regulation of hippocampal cell proliferation might influence neurodegenerative disorders, but these effects
ordinarily induced by inescapable (uncontrollable) shock could potentially occur through processes unrelated to
(Malberg and Duman, 2003). Similarly, hippocampal progeni- depression.
tor cell proliferation and BDNF levels were decreased in
chronically stressed mice, and repeated administration of the 13.4. Downstream growth factor signaling pathways in
antidepressant moclobemide normalized cellular proliferation depression
and BDNF concentration (Li et al., 2004a,b). As well,
decreased hippocampal cell proliferation elicited by chronic Reductions of BDNF and bcl-2 expression elicited by
restraint was prevented by administration of the SNRI stressors and neurodegenerative insults have been linked to
venlafaxine (Xu et al., 2006). altered levels of neurotrophic factors and induction of mitogen-
In contrast to the effects of stressors, enriched environmental activated protein (MAP) kinase pathways (Manji and Chen,
conditions promoted neurogenesis and such an outcome was 2002). These signaling pathways may be important for BDNF
mediated by the provocation of several growth factors. and associated growth factor functioning (Ying et al., 2002;
Specifically, environmental enrichment enhanced the produc- Eom et al., 2004; Wang et al., 2006; Duman et al., 2007).
tion of immature neurons and increased residual neurogenesis Growth factors have, indeed, been shown to stimulate
following irradiation. Concomitantly, enriched conditions also extracellular signal-regulated kinase (ERK), c-Jun N-terminal
improved spatial and motor learning in a water-maze and kinase (JNK) and p38 pathways. These three MAP kinase
rotorod test, indicating that the positive impact on neurogenesis pathways are believed to be fundamental in cellular responses
was associated with functional improvements (Fan et al., 2007). to environmental events through the transmission of synaptic
However, heterozygous BDNF knockout mice, which have low signals to the nucleus, thereby promoting synthesis of proteins
levels of the neurotrophin, failed to show the normal elevation important for cellular survival/death as well as inflammatory
of neurogenesis after exposure to enriched housing conditions processes (Curtis and Finkbeiner, 1999).
(Rossi et al., 2006), attesting to the fundamental role of this Recent in vitro work revealed that imiprimine induced
growth factor in neurogenesis. BDNF and bcl-2 expression and had anti-apoptotic conse-
quences that were prevented by the MAP kinase inhibitor
13.3. Neurodegeneration and depression U0126 (Peng et al., 2007). As such, these data raise the
possibility that MAP kinase signaling may be fundamental for
There is evidence that indirectly links activation of pro-death neuroprotective as well as affective regulatory consequences
pathways with depression as several studies hinted at the of antidepressants. Indeed, studies using animal models of
possibility that apoptotic, excitotoxic or oxidative stress factors depression, as well as those that assessed the impact of
may be altered over the course of depressive illness or antidepressants, supported the view that MAP kinase signaling
following exposure to chronic stressors. For instance, cascades may contribute to the modulation of depressive
postmortem analysis revealed modest DNA fragmentation in pathology (Duman et al., 2007; Qi et al., 2006). For instance,
11 of 15 depressed cases (Lucassen et al., 2001). Furthermore, systemic administration of the MAP kinase inhibitor PD184161
in vitro, antidepressants such as imiprimine and fluoxetine blocked the antidepressant-like effects of desipramine and
prevented apoptotic cell death (Peng et al., 2007; Nahon et al., sertraline in a forced swim test (Duman et al., 2007). However,
2005). However, data are unavailable concerning the expres- when administered directly into the amygdala, PD098059,
36 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

another MAP kinase antagonist, attenuated immobility in a factors and cytokines (e.g., TNF-a and IL-1b) that occur
forced swim test (Huang and Lin, 2006). Whether these effects following traumatic ischemic injury (Piao et al., 2003). In
reflect the route of administration or the unique properties of peripheral tissues, it was likewise observed that in response to
these inhibitors remains to be established. infection (Mycobacterium), p38 signaling mediates macro-
Olfactory bulbectomy (which elicits depressive-like beha- phage responses involving the synthesis of TNF-a and other
viors in rodents) also influenced MAP kinase signaling, as this cytokines (Roach et al., 2005). However, IL-1b administration
treatment elicited enhanced amygdaloid expression of the stimulated central expression of elements of all three MAP
immediate early gene c-Jun (Wrynn et al., 2000), which is a kinase pathways (Maher et al., 2004; Minogue et al., 2003; Wu
fundamental messenger of the JNK pathway. Following JNK et al., 2004). In this respect, IL-1b administration stimulated
activation, c-Jun dimerizes with c-Fos to control expression of ERK activity, as indicated by the enhanced phosphorylation of
several genes that promote either adaptive neuroplastic changes ERK-1 and -2 at the meninges, circumventricular organs, PVN,
or apoptotic responses to toxic insults, such as trophic factor arcuate nucleus and central amygdala (Nadjar et al., 2005). The
withdrawal, traumatic brain injury or neurotoxin exposure possibility exists that through MAP kinase pathways (e.g.,
(Cowan and Storey, 2003; Crocker et al., 2001; Hayley et al., ERK), IL-1b may inhibit neuroplastic processes. In fact,
2004a; Saporito et al., 2002). Augmented JNK expression selectively blocking these pathways also influenced the
within the amygdala and hippocampus was also provoked by establishment of long-term potentiation and the degree of
administration of electroconvulsive shock that involved a neuronal injury sustained following central insults (Minogue
protocol similar to that used in the clinical treatment of et al., 2003; Wang et al., 2004).
depression (Brecht et al., 1999). Although this treatment In addition to MAP kinases, the cAMP response element-
initially enhanced c-Jun activation, diminished expression of binding protein (CREB) may be important for controlling
the active immediate early gene was apparent with progressive affective and neuroplastic processes through its transcriptional
treatment (Brecht et al., 1999), suggesting neuroplastic changes regulation of stressor sensitive genes (D’Sa and Duman, 2002;
corresponding to the clinical efficacy of the treatment. Duman et al., 2007). In fact, CREB deficient mice displayed an
In addition to JNK signaling, the ERK pathway appears to be antidepressant phenotype in the forced swim and tail suspension
particularly sensitive to stressful manipulations. For instance, tests, which was accompanied by augmented hippocampal cell
chronic restraint enhanced ERK phosphorylation within proliferation (Gur et al., 2007). As well, mice lacking the CREB
serotonergic raphe neurons (Imbe et al., 2004). However, gene had a more rapid positive behavioral response following
ERK-2 expression and activational state (through phosphoryla- antidepressant treatment (Gur et al., 2007). Beyond these effects,
tion) was diminished in the hippocampus and prefrontal cortex CREB signaling within the hippocampus appeared to be involved
of rats exposed to 14 days of forced swim, a treatment that also in the regulation of long-term potentiation (Nguyen and Woo,
led to anhedonic-like behavioral consequences (Qi et al., 2006). 2003) and stressors may impair these processes by affecting
Paralleling the effects seen in animals, protein and mRNA CREB expression (Song et al., 2006).
expression of ERK was reduced in the hippocampus and PFC of Consistent with a role for CREB in depression, the
depressed suicides (Dwivedi et al., 2001, 2006). behavioral impairment elicited by repeated exposure to
Antidepressant treatments influenced the ERK pathway, inescapable footshock was associated with diminished
with chronic (but not acute) fluoxetine treatment promoting a [3H]cAMP binding, accompanied by alterations of several
long-lasting reduction of phosphorylated ERK-1 and -2 within intracellular kinases, including PKA, that are important for
the cytosol and nuclear compartments within the frontal cortex, CREB signaling (Dwivedi et al., 2004). Further, amygdala
but were differentially regulated within the hippocampus. CREB over-expression, which may be important for anxiety
Interestingly, however, imipramine increased ERK-1 in the associated with depression, was apparent with attenuation of
cytosol of frontal cortex neurons (Fumagalli et al., 2005). As behavioral impairments in the learned helplessness model of
such, these data raise the possibility that the differential 5-HT depression (Wallace et al., 2004). The reduced CREB
and NE functioning elicited by these two antidepressants (or the expression reported in rodents exposed to a chronic stressor
combined actions of imipramine) may be responsible for the was reversed by 5-HT acting antidepressant treatments (Shen
divergent ERK-1 changes reported. It might be noted, as well, et al., 2004), and antidepressants that influenced 5-HT and NE
that treatment with fluoxetine enhanced central GDNF uptake also provoked increased hippocampal and cortical
expression through the ERK and p38 kinase signaling pathways expression of phosphorylated CREB (Sairanen et al., 2007).
(Mercier et al., 2004). Essentially, these data raise the As in animals, reduced expression of CREB was observed
possibility that MAP kinases serve as an interface, bridging within the prefrontal and orbitofrontal cortices of depressed
neuronal plasticity with the emotional state elicited by various suicides (Dwivedi et al., 2003, 2004; Yamada et al., 2003a,b).
challenges. However, increased cortical CREB levels were also found in a
In addition to their modulation by stressful stimuli and mixed group of major depressive and bipolar disorder suicide
antidepressant treatments, MAP kinases are also influenced by victims (Odagaki et al., 2001). Moreover, Sibille et al. (2004)
cytokines, and conversely the downstream effects of cytokines failed to observe any CREB or BDNF changes in the
may involve activation of certain elements of MAP kinase dorsomedial and orbital prefrontal cortices of depressed
pathways. The p38 MAP kinase pathway appears to be suicides. Overall, it would appear that the role of CREB
necessary for microglial activation and release of oxidative signaling in depression is controversial and the discrepancies in
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 37

findings may reflect procedural differences, including varia- CRH-induced release of glucocorticoids has well-established
tions in subject populations examined. As indicated repeatedly, inhibitory effects on the production of numerous pro-
depression is a heterogeneous illness, in which the sub-types inflammatory cytokines and peripheral lymphocytes (Glazer
may be characterized by different signaling mechanisms as well and Rivest, 2004). Systemic administration of CRH or the
as co-morbid conditions that could potentially influence CRH2 agonist urocortin increased corticoid production,
neurotrophic factors. resulting in the attenuation of the normal rise of IL-1b and
TNF-a associated with LPS treatment (Agnello et al., 1998).
14. Neuropeptide and cytokine influences on Central CRH infusion likewise inhibited the inflammatory
neurodegeneration: Contribution of the inflammatory effects of LPS, including the induction of the intracellular
immune system adhesion molecule ICAM-1 on cerebral vessels, and this
treatment also impaired leukocyte infiltration into the brain
As indicated earlier, there is a high degree of comorbidity parenchyma (Casadevall et al., 1999). Furthermore, along with
between depression and several neurodegenerative illnesses, a marked reduction of plasma corticosterone, the levels of IL-4,
including Alzheimer’s disease (AD) and Parkinson’s disease IL-5, IL-13, IFN-g and the chemokine RANTES were all
(PD) as well as the aftermath of stroke. In addition to their increased in CRH knockout mice (Silverman et al., 2004),
potential effects on depressive illness, neuropeptide and suggesting that the balance of anti- and pro-inflammatory
cytokine factors have been implicated in neurodegenerative cytokines was altered in the absence of normal HPA drive.
disorders, and inflammatory processes might be a common Consistent with the animal studies, in a subset of depressed
denominator among these pathologies, and may contribute to patients, hyperactive basal HPA functioning was coupled with
the comorbidity that appears among them. reduced TNF-a levels (Himmerich et al., 2006), again
Accumulating evidence has pointed to an important role for suggesting that CRH-dependent corticoid activity was linked
neuroinflammation in the progression, and in some cases the to inflammatory cytokine functioning.
onset, of acute and chronic neurological conditions, such as AD, It seems that CRH may have anti-inflammatory effects
PD, stroke and MS (Mogi et al., 1996; Griffin et al., 2006; Owens, independent of glucocorticoids, as the peptide directly inhibited
2002; Sawada et al., 2006). Several peptides influenced the TNF-a production in primary macrophages by modulating
expression and activity of inflammatory factors that have been COX-2 dependent prostaglandin production (Tsatsanis et al.,
implicated in neuronal injury. In this regard, centrally acting 2007). Similarly, the corticosterone antagonist cyanoketone
peptides, including CRH, insulin-like growth factor I (IGF-I), prevented the inhibitory effects of urocortin on TNF-a
alpha-melanocyte-stimulating hormone (a-MSH), VIP, and production, but did not influence IL-1b expression, raising
PACAP, have potent anti-inflammatory actions under most the possibility that the peptide modulated IL-1b expression
conditions, whereas others, including calcitonin gene-related through processes independent of corticosterone (Agnello
peptide (CGRP) and substance P (SP) have pro-inflammatory et al., 1998).
actions (Gonzalez-Rey and Delgado, 2007; Gravanis and
Margioris, 2005; Calcagni and Elenkov, 2006). Some of these 14.2. Anti-inflammatory properties of a-MSH, VIP, PACAP
neuropeptides (e.g., CRH and angiotensin II (AGII)), may and IGF-1
simultaneously influence both pro- and anti-inflammatory
factors, resulting in shifts between these states, depending upon Like CRH, the centrally active peptide a-MSH has marked
the timing of exposure and concentration of these peptides. anti-inflammatory properties, and does so through melanocor-
The majority of neuropeptide-induced changes of immune tin receptors that are found throughout the periphery and at
cell activity and inflammatory cascades are mediated, at some several hypothalamic and extra-hypothalamic brain regions
level, by pro- and anti-inflammatory cytokines (Blalock, 1999; (Vulliémoz et al., 2006). In particular, a-MSH down-regulated
Ho et al., 1996; Ransjo et al., 1998). In fact, it seems that leukocyte activation and simultaneously blocked the elevation
reciprocal neuropeptide and cytokine interactions may be of corticosterone, as well as inducible nitric oxidase synthase
fundamental in the regulation of virtually all neuroinflamma- (iNOS) and COX-2 expression in response to several
tory processes. In the ensuing section we describe the role of immunogenic insults (Caruso et al., 2004). Overall, many of
cytokines in orchestrating inflammatory immune responses, the anti-inflammatory effects of a-MSH may be mediated by its
and the contribution of peptides in promoting these responses. ability to inhibit the transcription factor NFkB (Catania et al.,
Finally, the involvement of inflammatory cytokines in acute 1999). Indeed, the ability of a-MSH to inhibit TNF-a, IL-1b,
(stroke and head injury) and chronic (AD and PD) neurode- IL-6 and IFN-g secretion from olfactory and Schwann cells was
generation is presented, together with potential processes by dependent upon the down-regulation of NFkB signaling (Teare
which cytokines might interact with peptides to influence such et al., 2003, 2004).
pathology. Both VIP and PACAP belong to the secretin/glucagon
family of peptides and are produced by several immune cells,
14.1. Anti-inflammatory properties of CRH such as T lymphocytes, and share many common functions with
one another, including their anti-inflammatory effects (Gonza-
The anti-inflammatory actions of neuropeptide factors often lez-Rey and Delgado, 2007). VIP and PACAP inhibit pro-
stem from their ability to regulate HPA functioning. Indeed, inflammatory cytokine production and reduce the action of
38 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

dendritic and T cells, as well as microglia (Gonzalez-Rey and in macrophages (Agelaki et al., 2002), indicating the
Delgado, 2007; Hill et al., 2007). Administration of VIP alone importance of this receptor in the pro-inflammatory effects
elevated levels of the anti-inflammatory cytokine IL-10, and of the peptide. Conversely, IL-1b administration elevated CRH
when the peptide was co-administered with LPS, it limited the expression within the periphery, as well as within hypothalamic
elevation of IL-6 and plasma corticosterone ordinarily and amygdaloid brain nuclei (Nguyen et al., 1998; Day et al.,
provoked by the endotoxin (Bik et al., 2004). Similarly, 1999).
PACAP markedly reduced the ability of LPS to promote Following injury or immune challenges, peptides such as
corticosterone and TNF-a elevations and concomitantly CGRP, SP and AG II provoke potent pro-inflammatory actions
increased levels of T4 thyroid hormone (Baranowska-Bik on some cell types (Dunzendorfer et al., 1998). For instance,
et al., 2006). In addition to these peripheral actions, systemic central infusion of CGRP antagonists reduced IL-6 release from
VIP administration had central anti-inflammatory conse- injured neurons (Ma and Quirion, 2006) and CGRP itself
quences, as indicated by its ability to reduce microglial enhanced the production of IL-1b and TNF-a from herpes
activation following exposure to the DA neurotoxin MPTP simplex virus type-1 infected macrophages (Yaraee et al.,
(Gonzalez-Rey and Delgado, 2005). 2003). Similarly, SP provoked IL-1b, IL-6 and TNF-a release
Like several other peptides, IGF-I plays a fundamental from blood monocytes, and perpetuated activation of lympho-
role in the regulation of metabolism, proliferation and cytes and mast cells (Lotz et al., 1988; Kimball et al., 1988;
differentiation of most cell types. This growth factor is also Delgado, 2003). Following neurogenic inflammation provoked
essential for normal neuronal development and is critically by afferent nerve stimulation, application of SP sensitized
involved in the maturation of immunocompetent microglia central pain fibers, provoked alterations of cerebral blood vessel
within the CNS (Kaur et al., 2006). Like the previously constriction and recruited circulating lymphocytes and macro-
mentioned peptides, IGF-I stimulates several anti-inflamma- phages (Gendek-Kubiak, 2002). As well, SP may influence
tory cytokines, including both IL-4 and IL-10 (Kooijman and BBB permeability through mast cell activation elicited
Coppens, 2004). In response to acute tissue injury, such as following exposure to inflammatory and stressful insults
third degree burns or hematoma, IGF-I functions to (Annunziata et al., 2002; Singh et al., 1999).
equilibrate pro- to anti-inflammatory cytokine ratios, In the case of AG II, the osmoregulatory peptide may have
inhibiting IL-1b, IL-6 and TNF-a, while enhancing IL-4 some anti-inflammatory actions in addition to its potent pro-
and IL-10 production, thereby protecting against inflamma- inflammatory effects upon the cardiovascular and renal systems
tion-induced organ failure (Jeschke et al., 2002). Reciprocal (Schmieder et al., 2007; Ferder et al., 2006). Indeed, AG II
interactions exist between IGF-I and cytokines, as reflected inhibited the inflammatory effects of a cold stressor, causing a
by the finding that TNF-a administration reduced IGF-I reduction of stress-induced expression of TNF-a, prostaglandin
expression and the apoptotic effects of TNF-a were related to (PGE2) and adhesion protein ICAM-1, and prevented
its ability to down-regulate IGF-I (Anwar et al., 2002; neutrophil infiltration into the gastric mucosa (Bregonzio
Venters et al., 1999). et al., 2004). However, in the absence of a stressor, the peptide
Taken together, it seems that certain neuropeptides act on induced vascular expression of adhesion molecules, and
cytokines to promote anti-inflammatory actions or to limit the promoted cytokine and chemokine expression (Dandona
pro-inflammatory effects that would otherwise occur. As will et al., 2007; Ferder et al., 2006). Correspondingly, AG II
become evident shortly, these anti-inflammatory effects may be receptor blockers inhibited inflammatory mediators, including
fundamental in limiting pathology related to several neurolo- oxidative species and C-reactive protein (Dandona et al., 2007).
gical conditions. Certainly, in the case of cardiovascular insults, AG II acts in a
pro-inflammatory capacity since the peptide was found to
14.3. Proinflammatory effects of peptides contribute to cytokine and chemokine regulated inflammatory
damage following myocardial infarction (Schmieder et al.,
In addition to their anti-inflammatory actions, several 2007).
neuropeptides possess pro-inflammatory effects that may come Summarizing briefly, depending upon the peptide in
about by virtue of their influence on COX-2, oxidative question and the specific physiological context, neuropeptides
metabolism, induction of adhesion molecules, and production may have anti-inflammatory or pro-inflammatory effects.
of pro-inflammatory cytokines (Agelaki et al., 2002; McEvoy Clearly, in considering the influence of these peptides in the
et al., 2004). However, the nature of inflammatory reactions development of pathology, these divergent peptidergic actions
may change over the course of peptidergic activation, as well as ought to be considered. Although the present review focuses
the microenvironment in which these peptides act. For instance, only on a subset of such peptidergic factors (largely owing to a
although acute exposure to CRH ordinarily has anti-inflam- paucity of information), the possibility should not be ruled out
matory consequences, prolonged exposure to the peptide that a wide range of peptides could potentially influence
actually enhanced TNF-a expression within macrophages, neuronal survival and functioning. Moreover, as indicated
suggesting that the peptide had a time-dependent biphasic earlier, it ultimately may be important to consider the
effect in this regard (Tsatsanis et al., 2007). Moreover, interactive effects of different peptides or peptide-cytokine
administration of the CRH1 receptor antagonist antalarmin combinations in mediating or moderating the effects of various
suppressed LPS induced production of IL-1b, IL-6 and TNF-a challenges.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 39

15. Neuropeptide and cytokine interactions in the associated with stroke and head injury, CRH is thought to play a
modulation of neurodegeneration prominent role, and hence will be discussed in detail. In relation
to chronic conditions, specifically AD and PD, important roles
Neuroinflammatory factors, including both pro- and anti- for VIP and IGF-I have been suggested, and are thus discussed
inflammatory cytokines, are elevated in response to acute accordingly.
trauma as well as chronic disease states, including stroke, acute
head injury, AD, PD and MS (Hillhouse et al., 1998; Hopkins 15.1. Role of cytokines and CRH in stroke
and Rothwell, 1995; Mogi et al., 1994, 1995, 1996; Mrak et al.,
1995; Rothwell, 1999; Griffin et al., 1995). Certain disease Several lines of evidence favor a role for IL-1b in stroke,
states, such as cardiovascular conditions, are accompanied by a including those demonstrating that IL-1b and its receptor
rapid and persistent increase of TNF-a within the hypothala- antagonist, IL-1ra, were elevated by middle cerebral carotid
mus (Francis et al., 2004). As will be seen shortly, if one accepts artery occlusion (MCAO) and that central infusion of IL-1b
that cytokines contribute to depressive illness, then the long- exacerbated the neuronal damage provoked by ischemic insult,
term central cytokine changes might contribute to the as well as that induced by administration of excitotoxins such as
comorbidities that have been reported (e.g., the comorbidity kainic acid or glutamate analogues (Rothwell, 1999; Yamasaki
between cardiovascular illness and depression; Frasure-Smith et al., 1995). Conversely, inhibition of IL-1b reduced ischemic
and Lesperance, 2005). Yet, it ought to be underscored that infarct size, whereas immuno-neutralization of IL-1ra (i.e.,
cytokines may have both neuroprotective and neurodestructive attenuating the effects of the endogenous IL-1 receptor
factors, depending on the specific cytokine considered, the antagonist) markedly exacerbated neuronal damage (Loddick
concentration of the cytokine, and the specific constellation of et al., 1997; Lucas et al., 2006). The effects of IL-1b upon
cytokines that might be present (Stoll et al., 2000). ischemic neural damage may be mediated, at least in part, by
As already indicated, peptides may influence neurodegen- the well-established stimulatory actions of the cytokine on
erative processes through their impact on cytokines and CRH expression within the hypothalamus or cortical regions
neuroinflammatory mechanisms (Allan et al., 2005; Hayley (Allan et al., 2000). That said, a reciprocal relationship appears
et al., 2005; Niissalo et al., 2002), and peptide-cytokine to exist between IL-1b and CRH, as knockout mice lacking
interactions likely play a role in disturbed neuroimmune CRH displayed attenuated mRNA for the cytokine following
communication that typically characterizes these pathological LPS administration (Pournajafi Nazarloo et al., 2003),
conditions. Yet, it is uncertain whether the peptide-cytokine suggesting that endogenous CRH levels were necessary for
mediated inflammation primarily acts to promote neurotoxic production of the cytokine. Furthermore, as indicated earlier,
outcomes, or whether they actually serve in a neuroprotective IL-1b augments monoamine turnover and depolarization of
capacity. For instance, it may be that relatively low endogenous hypothalamic parvocellular soma in the PVN, resulting in
cytokine levels act protectively to buffer against damage related enhanced CRH release from terminals in the median eminence
to death processes, whereas chronically elevated levels (or very (Ferri et al., 2005; Mohankumar and MohanKumar, 2005). As
high levels) of these factors might contribute to neuronal well, cytokine-induced elevation of CRH expression within the
damage (Bruce et al., 1996). Indeed, low levels of cytokines can hypothalamus, coupled with enhanced sensitivity of these
provoke the release of beneficial trophic factors and free radical neurons to subsequent stressful challenges (Schmidt et al.,
scavengers, such as GDNF and manganese superoxide 1995; Tilders and Schmidt, 1998), may influence the response
dismutase (MnSOD), whereas elevated levels of cytokines, to later insults, such as cerebral ischemia and myocardial
including IL-1b and TNF-a, can provoke damaging oxidative infarction.
radicals and caspase-dependent apoptotic cascades (Rogers It is likely that the deleterious effects of CRH in stroke
et al., 2001; von Boyen et al., 2006; Saud et al., 2005). depend upon its modulation of one or more intermediate
Ultimately, the concentration and timing of release of these messengers, including IL-1b or other vascular factors, as direct
cytokines and the activation state of neighboring immune cells, in vitro application of CRH on primary cortical neurons did not
such as microglia, likely determine the nature of cytokine cause cell death nor did it modify the neurotoxic actions of
consequences within specific microenvironments. As such, it is NMDA (Craighead et al., 2000). Although CRH alone did not
likely that the ‘‘ebb and flow’’ of cytokine and peptide changes influence neuronal survival of cultured hippocampal neurons,
that occur over the course of neurodegeneration might in the presence of oxygen and glucose deprivation, CRH
differentially influence neuronal survival and functioning. It augmented hippocampal degeneration, and conversely, a CRH
is possible that factors that may be beneficial at early stages of a antagonist prevented such an outcome (Wang et al., 2004).
disease may assume a deleterious role later in disease As in the case of in vitro models, analyses of CRH and
progression. cytokine variations within in vivo models of stroke have
The conjoint and temporal patterns of action of multiple supported a link between these factors in the modulation of
cytokines (i.e., shifts of pro- and anti-inflammatory factors) neural injury. Moreover, ischemia stemming from MCAO
likely determine the course of neurological diseases. At the resulted in a time-dependent increase of CRH mRNA
same time, however, the neuropeptide factors associated with expression together with elevation of several cytokines,
one form of illness may be distinct from those associated with a including IL-1b and TNF-a (Block et al., 2005; Loddick
second disease state. In the case of acute neurodegeneration et al., 1997; Minami et al., 1992), and it appears likely that like
40 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

IL-1b, TNF-a may play an important role in mediating some of neuronal degeneration through up-regulation of the pro-
the effects associated with stroke (Heldmann et al., 2005; apoptotic TNF-a family member Fas (Wang et al., 2004).
Martin-Villalba et al., 2001; Zaremba and Losy, 2001). In this Moreover, it seems that the impact of CRH on TNF-a release
regard, TNF-a was rapidly induced following MCAO (Intiso from microglia may have stemmed from activation of MAP
et al., 2004; Sairanen et al., 2001; Vila et al., 2000) and may act kinase signaling (Wang et al., 2003). Thus, it appears that MAP
upon microglia to promote glial-dependent neural injury. kinase signaling may be fundamental in the promotion of
Indeed, inhibition of TNF-a, using either genetic or pharma- neurodegenerative processes, just as they appeared to play a role
cological manipulations, attenuated infarct volume following in subserving depression. In both instances, CRH and cytokines
ischemia (Dawson et al., 1996; Esposito et al., 2007; Martin- might contribute to stressor- or trauma-induced pathology by
Villalba et al., 2001). Parenthetically, a genetic link between virtue of their effects on MAP kinase signaling.
TNF-a and stroke was suggested by the significant association Although the BBB ordinarily protects the brain from large
found between a polymorphism of the TNF-a type 1 and 2 molecules that might disturb neural integrity, it seems that with
receptors and increased susceptibility to stroke within a Korean traumatic head injury there is a breakdown of the BBB (at least
population (Lee et al., 2004). temporarily), coupled with activation of resident glial cells, and
Interestingly, under some conditions, TNF-a may also have a the consequent induction of pro-inflammatory cytokines
neuroprotective action with respect to particular aspects of (Feuerstein et al., 1997; Jensen et al., 1997; Sullivan et al.,
stroke. For instance, the cytokine was found to be fundamental 1999). Electron microscopy studies indicated that TNF-a was
for the establishment of ischemic tolerance, wherein a mild non- elevated in lysosomes of glial cells at the site of injury 30 min
degenerative ischemic or inflammatory insult protected neurons after a percussive insult, and levels of this cytokine were
from the damaging effects of a subsequent more severe ischemic elevated within the hippocampus and parasagittal cortex 1 h
challenge (Castillo et al., 2003; Pradillo et al., 2005). In effect, following the injury (Kita et al., 1997). The severity of
after an ischemic preconditioning challenge the cytokine that is traumatic brain injury influences the nature of cytokine changes
released may function in a neuroprotective capacity, but in the observed, with moderate injury provoking IL-6 and TNF-a,
absence of this precondition it may contribute to neural injury whereas a relatively severe insult was required to elevate IL-1b
(Rosenzweig et al., 2004). Finally, it is also important to consider expression (Ahn et al., 2004).
that other cytokines might, either independently or in conjunc- It is thought that up-regulated cytokine expression might be
tion with IL-1b and TNF-a, influence the neurodegenerative fundamental in the provocation of the brain damage observed
course of cerebral ischemia. In this regard, the elevated central after traumatic head injury as well as in association with the
expression of IL-6 provoked by stroke may act in a edema seen after percussion injury (Knoblach et al., 1999;
neuroprotective fashion. Indeed, the cytokine was reported to Morganti-Kossmann et al., 1992). Such an outcome may occur
protect neurons from the NMDA mediated neuronal excitotoxi- through alterations of growth factors, metabolic turnover or brain
city associated with an ischemic event (Ali et al., 2000). temperature, especially as hypothermic treatments not only
reduced severity of injury, but also diminished TNF-a expression
15.2. Role of cytokines and CRH in sequelae of acute head within the injured hippocampus (Vitarbo et al., 2004).
injury Several studies have begun to focus on lesser known
cytokines, such as IL-23 and IL-18 (which have also been
In addition to playing a role in moderating the response to implicated in multiple sclerosis; Wheeler and Owens, 2005), in
stroke, it seems that other forms of brain injury are also the modulation of neural injury following traumatic insults. For
associated with marked cytokine and CRH changes. Once again, instance, inhibition of IL-18, a member of the IL-1 family,
it is not entirely certain whether the elevated cytokine levels significantly improved functional recovery following a closed
reflect an effort to act in a protective capacity or whether the head injury (Yatsiv et al., 2002). Similarly, greatly attenuated
cytokines, in fact, contribute to pathology. Acute traumatic head infarct volume following cerebral ischemia was apparent in IL-
injury induced CRH expression within the amygdala and 18 deficient mice (Hedtjärn et al., 2002). It has been suggested
hypothalamus, but surprisingly not in the cortical area that that IL-18 and it’s family member IL-1b might contribute to
sustained direct damage (Roe et al., 1998). Although elevated different phases of degeneration following cerebral ischemia,
hypothalamic CRH mRNA was provoked by traumatic head as IL-1b was elevated in microglia within hours following the
injury, levels of hypothalamic AVP and circulating corticoster- insult, whereas IL-18 expression only became apparent after
one were not affected by the insult (Grundy et al., 2001). several days (Jander et al., 2002). It may be that IL-18
Likewise, fluid percussion injury increased hypothalamic CRH contributes to later occurring neuronal loss following cerebral
mRNA, even in the absence of increased circulating corticoster- damage, possibly by influencing TNF-a, Fas or IFN-g signaling
one, suggesting that the peptide might moderate the con- (Prinz and Hanisch, 1999; Zhang et al., 2007).
sequences of traumatic injury, independent of glucocorticoid
activation (Grundy et al., 2001). It is possible that CRH 15.3. Role of cytokines, VIP, and PACAP in Parkinson’s
contributes to neural injury through its effects upon cytokines and disease
inflammatory microglia. In support of this contention, in vitro
application of CRH alone had no effect on hippocampal neurons, Epidemiological studies have implicated several environ-
but when they were co-cultured with microglia, CRH promoted mental toxins in the development of PD, with the incidence of
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 41

the disease being high in areas with heavy usage of certain levels of several pro-inflammatory cytokines (IL-1, IL-2, IL-6
pesticides, such as rotenone and paraquat (Betarbet et al., 2000; and TNF-a), as well as expression of elements of the
Jenner, 2001; Liu et al., 2003; Thiruchelvam et al., 2000; Tsui complement cascade important in mediating antibody depen-
et al., 1999). These pesticides also promote a loss of substantia dent cytotoxicity (Czlonkowska et al., 2002, 2005; Sawada
nigra pars compacta (SNc) DA neurons in rodents and induce et al., 2006; McGeer et al., 2002, 2003). However, unlike the
the activation of microglia and production of neuroinflamma- data regarding multiple sclerosis and stroke, definitive evidence
tory factors (Gao et al., 2003a; Liu et al., 2003; Peng et al., of T lymphocytes within the PD brain is lacking. Yet, reduced
2004). Additionally, repeated exposure to certain viral or circulating levels of T cells and impaired mitogen induced
bacterial infections was associated with increased risk of proliferative responses in PD patients indicate some degree of
developing PD. In fact, a viral hypothesis for PD suggests that altered peripheral immunity (Czlonkowska et al., 2002).
prenatal or early in life infection with latent virus(es) (having a Further support for an immune abnormality in PD stems from
long incubation period) may instigate this disease state the finding that circulating natural killer cell (NK) levels were
(Ringheim and Conant, 2004; Takahashi and Yamada, 1999). elevated in patients and positively correlated with disease
For instance, cases of PD associated with von Economo severity (Bokor et al., 1993). Moreover, auto-antibodies
encephalitis, influenza epidemics and HIV have been reported reactive against basal ganglia neurons were present in a subset
years after infection (Dickman, 2001; Koutsilieri et al., of PD patients (30%) (Abramsky and Litvin, 1978),
2002a,b). Exposure to a bacterial pathogen may also play a suggesting a humoral immune response that was directed
role in PD, as prenatal LPS exposure provoked SNc DA neuron against the CNS. Likewise, examination of cases of parkinson-
loss coupled with elevated nigrostriatal TNF-a in adulthood ism related to encephalitis revealed that 95% of patients had
(Carvey et al., 2003). Similarly, early endotoxin treatment auto-antibodies reactive against basal ganglia antigens (Dale
elicited protracted effects upon emotional and ischemic et al., 2004).
responses in adulthood (Perry et al., 2003; Spencer et al., Ordinarily IFN-g and TNF-a participate in immune
2005, 2006). Thus, early immunogenic exposure may provoke surveillance mechanisms responsible for protecting the
mild neuroinflammation that, over time, causes neurodegen- organism from tumor and viral invasion. Yet, these factors
eration or renders DA neurons vulnerable to the effects of may be inappropriately activated in neurological disease states,
normally low-grade insults (Thiruchelvam et al., 2000). and might actually damage neurons by promoting inflammatory
In addition to their independent consequences, immune and oxidative factors. Several studies involving TNF-a receptor
infection may interact with environmental insults (pesticides) knockout mice reported that neutralization of TNF-a signaling
that were related to exaggerated neuroinflammatory, neurode- attenuated the degeneration of SNc neurons and the accom-
generative and behavioral changes in neurological patients panying striatal DA alterations associated with MPTP treatment
(Perry et al., 2003). For instance, co-administration of LPS with (Ferger et al., 2004; Sriram et al., 2002). As well, among IFN-g
the pesticide rotenone synergistically augmented DA loss in knockout mice attenuated DA neuronal loss occurred following
midbrain-microglial co-cultures (Gao et al., 2003a,b). In vivo in vivo MPTP treatment, and in vitro neutralization of IFN-g
studies also indicated that although the pesticide maneb had no attenuated the impact of rotenone upon mesencephalic DA and
effect on DA neurons, when co-administered with paraquat it microglial co-cultures (Mount et al., 2007).
synergistically enhanced nigrostriatal damage primarily Like IFN-g the administration of IFN-a was found to
through microglial-dependent reactivity (Thiruchelvam et al., influence processes that might be related to PD. By example, in
2000). Evidently, different classes of environmental insults may rodents IFN-a depressed motor activity, coupled with reduced
synergistically activate neurodestructive mechanisms that DA levels (Shuto et al., 1997) Likewise, in rhesus monkeys
involve inflammatory processes. IFN-a altered DA functioning in association with elevated
Inflammatory insults may also sensitize DA neurons to the huddling (Felger et al., 2007), and in normal human volunteers,
neurodegenerative effects of subsequent paraquat treatment. IFN-a disrupted psychomotor responding (Smith et al., 1988).
Specifically, infusion of LPS into the SNc sensitized DA Moreover, in patients undergoing immunotherapy, the cytokine
neurons, such that the neurodegenerative effects of the pesticide elicited symptoms reminiscent of PD that could be attenuated
were greatly increased when administered 2 days following the by l-DOPA (Sunami et al., 2000). Together these data are
endotoxin (Mangano and Hayley, in press). It is possible that consistent with the perspective that IFN-a may influence brain
activation of neuroinflammatory cascades within the SNc regions relevant to Parkinson’s disease, namely the basal
enhanced DA neuronal vulnerability by promoting microglial- ganglia and substantia nigra.
dependent damaging processes, while concomitantly down- In contrast to the deleterious consequences of IFN-g and
regulating astrocyte protective responses. This perspective is TNF-a, certain interleukins, including IL-6 and the potent anti-
consistent with previous reports that the anti-inflammatory drug inflammatory cytokine IL-10 may have neuroprotective actions
3-hydroxymorphinan protected DA neurons from the effects of in PD. Indeed, IL-6 knockout mice exhibited enhanced
LPS by stimulating astrocytes and inhibiting the actions of degeneration of SNc DA neurons and striatal terminals
microglia (Zhang et al., 2005). following MPTP, suggesting augmented sensitivity in the
Consistent with a role for pro-inflammatory cytokines in PD, absence of endogenous levels of the cytokine (Bolin et al.,
postmortem analyses of brain tissue revealed numerous signs of 2002). The IL-6 deficient mice also displayed attenuated
inflammation, including microglial activation and increased microglial and inducible nitric oxide responses, but normal
42 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

astrocyte levels after MPTP (Cardenas and Bolin, 2003).


Support for a potential role for IL-10 in PD also comes from the
finding that a polymorphism of the IL-10 gene promoter was
associated with an earlier age of disease onset (Hakansson
et al., 2005). Moreover, IL-10 administration protected SNc
DA neurons from the damaging consequences of the
inflammatory endotoxin LPS (Arimoto et al., 2006; Qian
et al., 2006).
Although IL-6 can have pro-inflammatory actions, including
the provocation of an acute phase response, this cytokine also
shares many anti-inflammatory immune functions with IL-10,
including the inhibition of macrophage activation and release of
TNF-a (Niemand et al., 2003; Petersen and Pedersen, 2005;
Riley et al., 1999). Many of the overlapping effects of IL-6 and
IL-10 may stem from similar signaling mechanisms primarily
involving activation of JAK and STAT proteins. Essentially,
STATs are phosphorylated by JAK kinases, resulting in their
dimerization and subsequent translocation to the nucleus to Fig. 2. Schematic representation of the route by which cytokines might
influence degeneration of DA neurons as occurs in Parkinson’s disease.
initiate the transcription of several target genes that regulate
Following toxin exposure (e.g. paraquat), IFN-g together with TNF-a (both
inflammatory and neurodegenerative processes. Particular of which may stem from peripheral immune cells or from local glial production)
attention has focused upon STAT3 in modulating many actions may promote microglial reactivity and drive production of inflammatory
of IL-6 and IL-10 (Niemand et al., 2003; Riley et al., 1999). prostaglandins (PGEs), reactive oxidative species (O2 , NO), and neuronal
Moreover, several other cytokines with neurotrophic properties, apoptotic factors (JNK, caspases). These, in turn, induce behavioural variations
and may contribute to neurodegeneration. In addition, the lower portion of the
including G-CSF, ciliary neurotrophic factor and erythropoie-
figure depicts the proposition that IL-6 and IL-10 might act upon astrocytes to
tin, had neuroprotective effects through STAT3 activation in promote activation of JAK kinases resulting in STAT3 phosphorylation and the
models of stroke and retinal degeneration (Ji et al., 2004; Kretz production of neuroprotective factors, such as the anti-oxidant, MnSOD or other
et al., 2005; Schabitz et al., 2003). growth and anti-apoptotic factors (e.g. oncostatin M, Bcl-2). Essentially,
It has been our contention that through JNK signaling, astrocyte activation by these cytokines may have neuroprotective consequences
by inhibiting microglial activity or directly blocking pro-death cascades at
cytokines might promote neurodegerative processes, whereas
nigrostriatal DA neurons (dashed lines). Of course, IL-6 and IL-10 may also
activation of JAK-STAT pathways might actually act in a have anti-inflammatory or neuroprotective actions independent of astrocytes,
neuroprotective capacity. In this respect, IFN-g and TNF-a are but these are not considered in detail in the figure.
proposed as having primarily neurodestructive actions, whereas
IL-6 and IL-10 serve in a protective fashion. Fig. 2 depicts
proposed neurodestructive pathways involving microglial 1b, TNF-a and reactive oxygen species (Delgado and Ganea,
activation by IFN-g and TNF-a, and the view is offered that 2003). These neuroprotective effects likely stemmed from their
IL-6 and IL-10 may act through astrocytes to attenuate inhibitory actions on the transcription factor, NFkB (Hulley
neurodegenerative processes that may be fundamental in et al., 1995). It will be recalled that NFkB is a well-established
promoting toxin-induced PD-like pathology. transcriptional regulator of pro-inflammatory cytokine produc-
Although limited evidence is available concerning neuro- tion, and inhibition of its expression prevented LPS-induced
peptide alterations in PD, several reports suggested that certain cytokine release from microglia (Laflamme and Rivest, 1999;
peptide factors, through their anti-inflammatory properties, Lee et al., 2005; Takatsuna et al., 2005). Moreover, NFkB was
may be neuroprotective in animal models of this disorder. Of shown to modulate microglial expression of other transcription
these, VIP has recently emerged as having potent neuropro- factors (e.g., STAT1) that are associated with IFN-g and IL-6
tective consequences in several models of PD, including signaling (Lee et al., 2005). Hence, VIP and PACAP may have
exposure to DA toxins, such as MPTP (Delgado and Ganea, their positive actions on neurodegenerative processes by acting
2003). As well, it has been shown that leptin administration, on neuroinflammatory NFkB signaling pathways (as described
through activation of the ERK pathway, attenuated the in Fig. 2).
neurodegerative effects of 6-OHDA (Weng et al., 2007). Besides microglial NFkB signaling, the neuroprotective
Both VIP and PACAP (which share 60% homology), but not consequences VIP may have involved modulation of brain mast
other members of this peptide family, including secretin and cell activity (Tunçel et al., 2005). In fact, VIP prevented the
glucagons, greatly attenuated the loss of DA neurons associated behavioral disturbances normally associated with the DA
with MPTP treatment (Delgado and Ganea, 2003). As well, VIP neurotoxin 6-OHDA, and this effect was related to changes in
prevented the neurotoxic effects of 6-OHDA upon cultured brain mast cell expression without affecting the inflammatory
neurons, possibly by buffering the resistance of these cells to exocytosis associated with these cells (Tunçel et al., 2005). It
oxidative stress (Offen et al., 2000). It appeared that the seems, as well, that VIP may influence DA neuronal survival by
neuroprotective effects of VIP and PACAP were dependent promoting the activity of protective growth factors. Specifi-
upon their inhibition of microglial activation and release of IL- cally, the peptide imparted neuroprotective actions by
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 43

stimulating astrocytes to secrete activity-dependent neuro- from co-cultures of astrocytes and microglia, whereas
trophic factor (Dejda et al., 2005). Similarly, the orphan nuclear inhibition of IFN-g reduced b-amyloid accumulation and
receptor Nurr1, which is essential for proper development of gliois in a genetic model of AD involving mutation of the b-
DA neurons, may influence neuronal survival through its amyloid precursor protein (Yamamoto et al., 2007). In addition,
impact upon VIP (Luo et al., 2007). cytokines may promote AD pathology by interfering with the
production of growth factors critical for the proper maintenance
15.4. Role of IGF-I and cytokines in Alzheimer’s disease of neurons. For instance, IL-1b inhibited BDNF signaling in
(AD) cultured neurons by blocking activation of the ERK MAP
kinase pathway (Tong et al., 2007). Taken together, these data
Cytokines have been implicated in the formation of senile suggest that cytokines, through their actions on b-amyloid and
plaques, which represent a defining histological feature of AD. growth factor functioning, may influence the course of AD.
For instance, IL-1b and IL-6 have been associated with the Besides acting in a provocative capacity in b-amyloid
deposition of b-amyloid in neuritic plaques and with the induced neurodegeneration, cytokines may also function in a
development of neurofibrillary tangles (Cotter et al., 1999; protective manner. For instance, pretreatment of neuronal
Eikelenboom et al., 2006; Heneka and O’Banion, 2007). cultures with TNF-a reduced hippocampal neuronal death and
Activated microglia, together with IL-1b, IL-6 and TNF-a, hyperphosphorylation of tau protein provoked by b-amyloid
have frequently been found in conjunction with neuritic plaques (Orellana et al., 2007). However, as already mentioned, TNF-a,
(Cotter et al., 1999; Eikelenboom et al., 2006; Kim and Joh, together with IL-1b and other pro-inflammatory cytokines,
2006; Haga et al., 1989). These cytokines may interact with one served to enhance b-amyloid accumulation and augmented its
another or with endogenous b-amyloid to influence neurode- neurodegenerative effects (Chiarini et al., 2006). In fact, it was
generation. In this regard, TNF-a, IL-1b and IFN-g synergis- reported that activation of TNF-R1 and the subsequent
tically enhanced the release of the free radical nitric oxide from induction of apoptotic protease-activating factor (APAF) were
astrocytes, an effect that was further amplified by addition of required for b-amyloid induced neuronal death to occur (Li
soluble b-amyloid fragments (Chiarini et al., 2006). As well, et al., 2004a,b). The duality of functioning of TNF-a in
neuritic plaque-associated microglia expressing IL-1a were neuronal survival may stem from the differential activation of
more likely to be in an activated phagocytic state than those not its two receptor subtypes, TNF-R1 and TNF-R2. Whereas TNF-
associated with plaques (Sheng et al., 1997). R1 has pro-death effects that are mediated by its intra-cellular
Indirect evidence concerning inflammatory processes in AD death domain and associated apoptotic caspases, TNF-R2
comes from studies of arthritic patients, in which anti- mainly has pro-survival and proliferative functions (Dopp et al.,
inflammatory drugs that antagonized these cytokines were 2002; Neta et al., 1992). Indeed, TNF-R2 activation typically
associated with reduced risk of AD (Heneka and O’Banion, stimulates several intra-cellular adapter proteins that influence
2007; McGeer and McGeer, 1999; McGeer et al., 2002). The the production of a number of neuroprotective proteins by
mechanism by which anti-inflammatory treatments attenuate NFkB (Baud and Karin, 2001; Fernyhough et al., 2005). As
AD plaque formation is uncertain. Nevertheless, it was reported well, the degree of activation of microglial cells may influence
(Simard et al., 2006) that in contrast to resident microglia, those the neurodegenerative effects of TNF-a. Indeed, this cytokine
derived from bone marrow might actually play a role in limiting had neurotoxic effects in neuronal cultures exposed to b-
senile plaque formation. amyloid in the presence of microglia, but not in those treated
Not only are cytokines associated with the presence of with the glial suppressor Ara-C (Chao et al., 1993; Takeuchi
neuritic plaques, but they may also contribute to plaque et al., 2006; Orellana et al., 2007). These findings suggest that
maturation. Although the presence of neuritic plaques in AD the TNF-a exposed glia may be releasing toxic factors that
patients correlates with clinical dementia, the earlier, pre- counteract any effects that the cytokine may have on
sumably less mature, diffuse plaques do not correlate well with neuroprotective pathways.
cognitive decline (Ding et al., 2007; von Gunten et al., 2006). In addition to the direct central actions of cytokines,
One intriguing hypothesis that has been offered is that infectious agents may influence systemic cytokines and
cytokines, in fact, drive the evolution of the plaques into their immune cell functioning that may, in turn, contribute to AD
more mature dense core forms (Griffin et al., 2006; Liu et al., pathology. Indeed, LPS-provoked elevations of circulating IL-
2005). Thus, IL-1 and IL-6 may contribute to AD pathology by 1b and IL-6, coupled with that of several chemokines,
increasing b-amyloid deposition and dystrophic neurite augmented neuronal damage associated with reduced cerebral
formation (Griffin et al., 2006). Evidence in favor of this blood flow and was posited to contribute to the pathology
hypothesis is supported by reports indicating that IL-1b observed in cases of AD involving neurovascular changes
released from microglia may provoke the release of the (McColl et al., 2007). Variations of circulating cytokines may
neurotrophin S100b from adjacent astrocytes, which may then also contribute to the progression from pre-clinical states,
contribute to the development of dystrophic neurites and other characterized by mild cognitive impairment, to the AD
pathological alterations in neuronal elements associated with syndrome. Peripheral blood monocytes derived from elderly
the plaques (Liu et al., 2005). Other cytokines, including TNF- individuals experiencing mild cognitive impairments displayed
a and IFN-g, may also contribute to b-amyloid deposition. In higher levels of mitogen stimulated IL-6, IL-8 and IL-10
fact, both of these cytokines enhanced b-amyloid production compared to healthy age-matched controls (Magaki et al.,
44 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

2007). As well, serum TNF-a levels were elevated in AD and in and the difficulties in the choice of treatments given different
patients with mild cognitive impairment, and levels of the comorbidities (Goodnick and Hernandez, 2000), knowledge of
cytokine were negatively correlated with concentration of the the processes that lead to a given disturbance may facilitate the
protective growth factor IGF-1 (Alvarez et al., 2007). identification of the mechanisms that promote a second
Although several genes have been implicated in the disturbance, and might also be instrumental in the development
development of AD, it seems that APOe E4 may be particularly of more effective treatment strategies for both.
important in this respect (Mahley et al., 2006). Moreover, In some instances the development of comorbid depression
recent findings also suggested that cytokine gene polymorph- that occurs in conjunction with physical illness might be a result
isms may have predictive power with regard to AD. For of the distress attributable to the disease or it may be secondary
instance, a polymorphism in the promoter region of the IL-10 to the psychosocial stress associated with it. Yet, it has also been
gene was found in an Italian sample to predict increased risk of reported that at least some of these comorbidities cannot be
AD (Bagnoli et al., 2007). Additionally, a polymorphism of the attributed to such factors, but instead might reflect the
IL-18 gene ( 137 C/C) was associated with a faster rate of possibility that the illnesses involve similar underlying
cognitive decline in AD patients (Bossù et al., 2007). processes or etiologies (Leentjens et al., 2003; McDonald
In addition to cytokines, insulin and IGF-I may be involved et al., 2003). For instance, as mentioned earlier, stressor-
in AD, as the peptide was reported to regulate amyolid provoked elevations of corticosterone may instigate hippo-
precursor protein (APP) processing and hence central levels of campal neuronal damage (McEwen, 1994; Sapolsky et al.,
b-amyloid (Adlerz et al., 2007). A role for IGF-I in the disease 2000), as well as inhibition of neurogenesis (Kozorovitskiy
has also been suggested by the finding of a polymorphism of the et al., 2004; Montaron et al., 2006), and as such could contribute
IGF-I receptor in a subset of female AD patients (Garcia et al., to both depression and chronic neurodegenerative diseases. In
2006). Similarly, disturbances of the IGF-I receptor were addition to hippocampal variations, a chronic variable stressor
reported in transgenic animal models of AD, as well as in regimen was shown to provoke cortical cell loss, and this
diabetic mice that displayed signs of dementia (Wu et al., 2006; outcome was synergistically enhanced by cortical administra-
Li et al., 2007). As might be expected, administration of IGF-I tion of LPS, and attenuated by the glucocorticoid inhibitor RU-
prevented cognitive and vascular dysfunction, and increased 486 (de Pablos et al., 2006). Likewise, stressor-induced
central IGF-I receptor levels in AD-like mutant mice that over- alterations of glutamate and DA activity within the basal
express amyloid precursor protein/presenilin genes (Lopez- ganglia were suggested to have neurotoxic consequences that
Lopez et al., 2007). might be relevant for PD (Smith et al., 2002; Zigmond et al.,
Many of the functions of IGF-I in AD may be associated 1998). Conversely, antidepressants or other agents that mitigate
with the ability of the peptide to influence neuroinflammatory the deleterious effects of stressors or provoke beneficial growth
cascades. In this regard, IGF-I attenuated the behavioral effects factors may have neuroprotective actions (Hayley et al., 2005;
of LPS (Bluthe et al., 2006), and conversely, exposure to TNF-a Manji and Chen, 2002; Peng et al., 2007). In fact, in addition to
induced neuronal apoptosis by inhibiting IGF-I pro-survival pharmacological treatments, manipulations such as exercise,
signals (Venters et al., 1999). Similarly, reduced IGF-I which have positive effects on mood, had neuroprotective
expression was associated with increased IFN-g levels, along consequences through the activation of GDNF and subsequent
with deficits in hippocampal long-term potentiation (Maher signaling of ERK kinases (Smith and Zigmond, 2003).
et al., 2006). Thus, pro-inflammatory cytokine activity may be Comorbid illnesses may be promoted by similar experiential
negatively coupled to IGF-I expression, and hence interactions factors (e.g., protracted distress), or they may be subject to
between these factors may have important modulatory effects several common underlying biological features. The specific
upon cognitive neuronal processes. Moreover, the insulin pathology that develops in response to a chronic stressor might
receptor substrates 1 and 2 (IRS-1 and -2), which are adaptor largely be determined by genetic dispositions, coupled with a
proteins essential for IGF signaling, require the anti-inflam- constellation of experiential and environmental factors. As
matory cytokine, IL-4, in order to induce proliferation of B and depicted in Fig. 3, there is reason to believe that activation of the
T lymphocytes (Hartman et al., 2004). Together, these data inflammatory immune system may be pivotal for the expression
suggest that IGF-I may act to suppress the deleterious central of each of these pathologies discussed to this point. As shown,
consequences of pro-inflammatory stimuli (TNF-a, IFN-g), multiple overlapping CNS processes may act in a feed-forward
while interacting with anti-inflammatory elements (IL-4) to manner, in which one illness may influence other co-morbid
provide pro-cellular functions. conditions by acting upon common or parallel biological
processes. Of course, co-morbid psychiatric pathology may stem
16. Cytokines mediate the comorbidity between from the psychosocial or prognostic burden of coping with the
depression and neurodegenerative conditions aftermath of neurological disability (Sobel et al., 2005).

As indicated earlier, comorbidity between psychiatric 16.1. Depression and comorbid cardiovascular (heart and
illnesses and neurological conditions is fairly common, and stroke) disturbances
not surprisingly, the presence of depression may also hinder
alleviation of symptoms related to these disorders. Despite the Of the pathologies that have been linked to major depression,
complications associated with dealing with comorbid illnesses, particular attention has been devoted to cardiovascular diseases
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 45

immune system could serve as a therapeutic target in the


treatment of cardiovascular disease, and that anti-inflammatory
agents may play a positive role in the treatment of depression
(Brustolim et al., 2006).
Despite the impressive evidence suggesting that CRP may be
pertinent to the comorbidity that exists between cardiovascular
illness and depression (Frasure-Smith and Lesperance, 2005;
Miller et al., 2005), other reports indicated that the relation
between CRP and depression was generally weak or entirely
absent (Steptoe et al., 2003). This was especially the case when
other comorbid factors were controlled, such as body mass index
(Douglas et al., 2004). Thus, as tempting as it might be to focus
heavily on this acute phase protein, it may be a bit premature to
suggest a causal role for CRP in the evolution of depression.
As in the case of heart attack, depression is often co-morbid
with cerebral stroke following ischemia or hemorrhagic insult
(Nuyen et al., 2006). Although post-stroke depression has been
successfully treated with a variety of antidepressants (Goodnick
and Hernandez, 2000), treatment resistance is more frequent in
comparison to that evident in absence of co-morbidity
(Iosifescu et al., 2004). It appears, as well, that anxiety often
Fig. 3. Stressors induce depressive illness, which may be associated with
several comorbid conditions, namely vascular illness (e.g., stroke and myo-
co-occurs in stroke patients already experiencing depression.
cardial infarction), as well as neurodegenerative conditions, including Parkin- Indeed, generalized anxiety disorder co-existed with depression
son’s and Alzheimer’s diseases. Inflammatory processes activated in response to in 20% of patients assessed 3–4 months after stroke, and in
stressors may be a common denominator for these varied illnesses. Indeed, addition to affecting mood symptoms, antidepressant treatment
several common cytokines have been associated with these pathologies, (nortriptyline) significantly diminished anxiety, and enhanced
although in several respects they may involve different processes. Specifically,
these cytokines might contribute to comorbid pathologies through several
overall patient functioning (Kimura et al., 2003). It seems that
divergent routes, including activation of microglia (release of oxidative and co-morbid depression or anxiety may be related to the severity
inflammatory factors), the promotion of plaque/protein aggregate formation, of functional impairment and progression of pathology
and by altering vascular factors that contribute to arterial blockage (e.g., clot associated with stroke.
formation). Several common biological processes may contribute to
stroke and co-morbid depression, including variations of
(heart attack and stroke). There is, indeed, ample literature central monoamines and neuropeptides that are thought to
showing that depression was a predictor of later cardiovascular contribute to depressive illness and that may be altered by the
disease and illness recurrence (Frasure-Smith and Lesperance, neurological damage that occurs following stroke (Kato et al.,
2005; Lespérance et al., 2004; Pratt et al., 1996; Zellweger et al., 2000; Behan et al., 1996). Moreover, as shown in Fig. 3,
2004). Conversely, myocardial events were predictive of activation of neuroinflammatory processes occur in both stroke
subsequent depression (Frasure-Smith and Lesperance, 2005). and depression and as such, inflammatory signaling factors may
It is of interest, as well, that the covariation of depressive play a role in perpetuating the co-morbidity. In particular, IL-
symptoms and coronary artery disease may involve a common 1b, TNF-a and IL-6 are markedly altered within the brain and
genetic vulnerability (McCaffery et al., 2006). Inasmuch as C- periphery following an ischemic event, as well as in response to
reactive protein, and other inflammatory factors (e.g., IL-6 and psychological stressors (Dantzer, 2006; Minami et al., 1991,
soluble intracellular adhesion molecule: sICAM-1) are asso- 2006; Spalletta et al., 2006). In fact, it was proposed that stroke-
ciated with heart disease (Dibbs et al., 1999; Gullestad and induced elevations of IL-1b, TNF-a and IL-18 may enhance
Aukrust, 2001) and with depression, and are also increased in activity of enzymatic pathways, including IDO, resulting in 5-
response to chronic stressors (Black, 2002; Pasic et al., 2003), the HT depletion at limbic brain regions, hence promoting
possibility has been explored that CRP may be an underlying subsequent depressive symptoms (Spalletta et al., 2006).
factor for both these illnesses. Indeed, the presence of depression Conversely, stressors that normally provoke depressive-like
and elevated levels of CRP were predictors of major adverse pathology enhanced the damaging effects of an ischemic insult
cardiac events over a 2-year period, and the occurrence of these and this outcome was thought to stem from the actions of pro-
events were especially frequent when individuals displayed both inflammatory cytokines. In particular, stressor exposure
elevated CRP and depression (Frasure-Smith et al., 2007). increased infarct size caused by later MCAO, whereas
Furthermore, it has been reported that antidepressants (SSRIs) inhibition of IL-1b or TNF-a prevented the augmenting
are not only effective in the treatment of depression among effects of the stressor (Caso et al., 2006, 2007). In effect, these
myocardial infarction patients, but that this treatment may findings suggest that stressors might contribute to both
facilitate management of coronary artery disease (Parissis et al., depressive and neurodegenerative pathology through their
2007). Likewise, there is reason to believe that the inflammatory actions on cytokines.
46 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

16.2. Depression and comorbid Parkinson’s disease depressive symptoms among individuals with mild cognitive
impairment greatly increased the probability that they would
Depressive illness is co-morbid with 40–50% of PD cases develop AD symptoms measured 3 years later (Palmer et al.,
and often precedes the development of motor disturbances or 2007). Similarly, co-morbid anxiety and depression additively
PD diagnosis. Beyond motor disabilities that characterize PD, increased the severity of cognitive decline in AD patients
the disease is frequently associated with neuropsychiatric (Starkstein et al., 2007). In general, elderly individuals that
symptoms, including depression, anxiety and memory dis- experience depressive illness appear to be particularly sensitive
turbances (McDonald et al., 2003). Given the temporal relations to the cognitive aspects of the disorder, such that it may be
between these disease states, depression has even been difficult to disentangle the affective disturbances from
suggested as a risk factor for PD development (Leentjens symptoms of AD or dementia. In fact, underlying clinical
et al., 2003; McDonald et al., 2003). Although PD is typically depression might be masked by a primary AD syndrome, hence
considered a disease that involves substantia nigra DA neurons, resulting in under-diagnosis of affective disturbances in the
degeneration or dysfunction of other brain regions (e.g., those elderly (Starkstein and Mizrahi, 2006).
associated with depression, such as the locus coeruleus, As we have repeatedly emphasized, related neurobiological
hippocampus, prefrontal cortex) have indeed been reported and psychological (e.g., exposure to stressors) mechanisms
(Chan-Palay, 1993; Leentjens et al., 2003). Given that may give rise to depression and neurodegenerative processes. In
inflammatory factors might influence both depressive and this respect, an increased number of hypothalamic CRH
PD syndromes, cytokines may contribute to the high degree of neurons were reported in both AD and depression (Meynen
co-morbidity that exists between these conditions (Hayley and et al., 2007) and it was postulated that a hyperactive HPA axis,
Anisman, 2005). characteristic of certain depressive subtypes, may engender
As already indicated, pro-inflammatory cytokines, such as vulnerability to AD pathology. Conversely, disturbances of
TNF-a and IFN-g, may contribute to PD pathology, whereas CRH signaling stemming from loss of neurons in AD might
peptides, including VIP and PACAP, may have neuroprotective promote negative mood states (Raadsheer et al., 1994; Meynen
actions. Just as peptide-induced anti-inflammatory effects may et al., 2007). Similarly, widespread degeneration within the AD
protect DA neurons, several antidepressants may gain some of brain has been associated with monoamine deficiencies in
their beneficial actions by promoting anti-inflammatory out- several brain regions (including the hippocampus, locus
comes. Indeed, in addition to their mood elevating properties, coeruleus and brainstem raphe) that have been linked to
antidepressants such as rolipram and buproprion, enhanced the depressive symptoms (Engelborghs and De Deyn, 1997). As
production of the anti-inflammatory cytokine IL-10 while such, treatments that influence common neurochemical
reducing levels of the pro-inflammatory cytokines TNF-a and processes may be beneficial for both depressive and AD co-
IFN-g (Brustolim et al., 2006; Zhu et al., 2001). As well, the morbid states and may even be used in a prophylactic capacity.
tricyclic antidepressant imiprimine protected hippocampal For instance, pretreatment with the SSRI paroxetine improved
neurons from the apoptotic consequences of high doses of cognitive performance and reduced the accumulation of b-
LPS and these effects were related to the anti-inflammatory amyloid and abnormal tau protein in a transgenic rodent model
actions of the antidepressant (Peng et al., 2007). Consistent of AD (Nelson et al., 2007). As well, the proposition was
with the view expressed here, cytokine inhibitors used in advanced that new therapies aimed at targeting cytokines and
treating inflammatory disease, diminished comorbid depressive growth factors (e.g., BDNF), together with their downstream
symptoms (Kulmatycki and Jamali, 2006). Thus, it seems that effectors, might be important for treatment of AD and co-
anti-inflammatory treatments might not only be useful in morbid depression (Kuipers and Bramham, 2006).
mitigating neural injury and offset the development of In addition to playing an important role in various pathologies,
neurodegenerative conditions in already depressed individuals, inflammatory processes likely contribute to the co-evolution of
they might also limit the onset of depressive symptoms in AD and depression. For instance, as seen in Fig. 3, augmented
neurological patients. For instance, glatiramer acetate, a pro-inflammatory cytokine levels may influence neuronal
synthetic polypeptide used to treat multiple sclerosis, was functioning in brain regions (e.g., prefrontal cortex and
hypothesized to have antidepressant effects through its hippocampus) that are normally aligned with both MDD and
stimulatory actions on the anti-inflammatory cytokine IL-10, AD. Likewise, neurotransmitters and inflammatory factors, such
as well as on BDNF (Tsai, 2007). From the available evidence, as oxidative radicals and COX-2 products, might act as triggers
it would seem likely that anti-inflammatory treatments might be for the co-evolution of these illnesses (Hussain and Mitra, 2000;
useful in the management of co-morbid neurodegenerative and Melnikova et al., 2006). In fact, COX-2 activation was shown to
depressive pathologies, as occurs in PD. enhance b-amyloid toxicity and, as in the case of depression,
inhibition of COX-2 signaling attenuated pathology in an animal
16.3. Depression and comorbid Alzheimer’s disease (AD) model of AD (Liang et al., 2005; Melnikova et al., 2006).
Moreover, altered cytokine (TNF-a, IL-1b) and histamine serum
AD patients frequently (25–40%) present with co-morbid levels were correlated with the incidence of depression among
depression (Caballero et al., 2006), and a recent meta-analysis AD patients (Alvarez et al., 1996). Finally, it was reported that
indicated that depression may be a significant risk factor for AD co-morbid AD and depression were associated with common
(Ownby et al., 2006). Moreover, the presence of anxiety and genetic polymorphisms of cytokine genes (McCulley et al.,
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 47

2004). Indeed, AD patients that were heterozygous for a Acknowledgements


polymorphism in the promoter region of IL-1b (-511 variant)
were much more likely to develop depression relative to This work was supported by the Canadian Institutes of
individuals that were homozygous for the genetic variant Health Research and by the Natural Sciences and Engineering
(McCulley et al., 2004). Ultimately, it may be that certain genetic Research Council of Canada. HA and SH hold Canada
factors engender inflammatory responses to stressful or Research Chairs in Behavioral Neuroscience and Neu-
neurotoxic insults that may favor the evolution of pathology roscience, respectively. SH also holds a NARSAD Young
within multiple CNS regions. Researchers Award. None of the authors have any direct or
indirect conflicts of interests of a financial nature relevant to the
17. Summary and conclusion present work.

In this review, we have emphasized the importance of


considering depressive disorder in terms of endophenotypes, References
rather than as a uniform illness. We indicated, as have others,
Abe, S., Hori, T., Suzuki, T., Baba, A., Shiraishi, H., Yamamoto, T., 1999.
that major depression represents a cluster of features, and in Effects of chronic administration of interferon alpha A/D on serotonergic
analyzing the neurochemical disturbances that characterize receptors in rat brain. Neurochem. Res. 24, 359–363.
depression, it is important to distinguish not only on the basis Abercrombie, E.D., Jacobs, B.L., 1988. Systemic naloxone administration
of illness severity and chronicity but also as a function of the potentiates locus coeruleus noradrenergic neuronal activity under stressful
but not nonstressful conditions. Brain Res. 441, 362–366.
symptoms that characterize the disorder. To be sure, a lengthy
Abercrombie, E.D., Keefe, K.A., DiFrischia, D.S., Zigmond, M.J., 1989.
list of neurochemical processes (and genes governing these Differential effects of stress on in vivo dopamine release in striatum,
processes) have been identified in relation to MDD, and in nucleus accumbens, and medial frontal cortex. J. Neurochem. 52, 1655–
some instances these could be tied to specific features of the 1658.
disorder. However, tying behavioral and neurochemical Abizaid, A., Liu, Z.W., Andrews, Z.B., Shanabrough, M., Borok, E., Elsworth,
endophenotypes to one another may prove exceedingly J.D., Roth, R.H., Sleeman, M.W., Picciotto, M.R., Tschöp, M.H., Gao, X.B.,
Horvath, T.L., 2006. Ghrelin modulates the activity and synaptic input
difficult given that neurochemical processes may interact organization of midbrain dopamine neurons while promoting appetite. J.
with one another, as well as with environmental and Clin. Invest. 116, 3229–3239.
experiential factors, in promoting a given symptom (or set Abramsky, O., Litvin, Y., 1978. Automimmune response to dopamine-receptor
of symptoms). as a possible mechanism in the pathogenesis of Parkinson’s disease and
The neurochemical features associated with the MDD schizophrenia. Perspect. Biol. Med. 22, 104–114.
Adlerz, L., Holback, S., Multhaup, G., Iverfeldt, K., 2007. IGF-1-induced
symptom profile span neurotransmitters, peptidergic factors, processing of the amyloid precursor protein family is mediated by different
inflammatory processes, and factors that influence neuronal signaling pathways. J. Biol. Chem. 282, 10203–10209.
plasticity and neurogenesis. Some of these same features are Agelaki, S., Tsatsanis, C., Gravanis, A., Margioris, A.N., 2002. Corticotropin-
also prominent in illnesses that are comorbid with MDD, releasing hormone augments proinflammatory cytokine production from
macrophages in vitro and in lipopolysaccharide-induced endotoxin shock in
most notably cardiovascular related events (including stroke
mice. Infect. Immun. 70, 6068–6074.
and its aftermath), PD and AD. In effect, it seems that Agnello, D., Bertini, R., Sacco, S., Meazza, C., Villa, P., Ghezzi, P., 1998.
inflammatory processes are a common denominator for these Corticosteroid-independent inhibition of tumor necrosis factor production
disorders, and it may well be that they contribute to the by the neuropeptide urocortin. Am. J. Physiol. 275, 757–762.
provocation of these different pathological states. This is not Aguilera, G., Rabadan-Diehl, C., Nikodemova, M., 2001. Regulation of pitui-
to say that cytokines necessarily subserve all instances of tary corticotropin releasing hormone receptors. Peptides 22, 769–774.
Ahn, M.J., Sherwood, E.R., Prough, D.S., Lin, C.Y., DeWitt, D.S., 2004. The
depression (or other pathologies), but rather that immune effects of traumatic brain injury on cerebral blood flow and brain
activation that involves inflammatory processes may give rise tissue nitric oxide levels and cytokine expression. J. Neurotrauma 21,
to depressive features, and that these effects might be 1431–1442.
particularly pronounced when superimposed on a backdrop of Akinci, M.K., Johnston, G.A., 1993. Sex differences in acute swim stress-
induced changes in the binding of MK-801 to the NMDA subclass of
stressful experiences. Likewise, it seems that both stressful
glutamate receptors in mouse forebrain. J. Neurochem. 61, 2290–2293.
events and activation of inflammatory processes may promote Alesci, S., Martinez, P.E., Kelkar, S., Ilias, I., Ronsaville, D.S., Listwak, S.,
the sensitization of neuronal functioning so that subsequent et al., 2005. Major depression is associated with significant diurnal eleva-
similar experiences give rise to more rapid and more tions in plasma interleukin-6 levels, a shift of its circadian rhythm, and loss
pronounced neurochemical changes, thereby favoring the of physiological complexity in its secretion: clinical implications. J. Clin.
emergence of MDD. Whether sensitization of neurochemical Endocrinol. Metab. 90, 2522–2530.
Alfonso, J., Frasch, A.C., Flugge, G., 2005. Chronic stress, depression and
processes and synergies between stressors and inflammatory antidepressants: effects on gene transcription in the hippocampus. Rev.
factors contribute to the emergence of neurodegenerative Neurosci. 16, 43–56.
disorders and cardiovascular illness has not been examined Ali, C., Nicole, O., Docagne, F., Lesne, S., MacKenzie, E.T., Nouvelot, A.,
thoroughly. Yet, the data that are available appear to support Buisson, A., Vivien, D., 2000. Ischemia-induced interleukin-6 as a potential
this view. Indeed, there is reason to believe that attenuating endogenous neuroprotective cytokine against NMDA receptor-mediated
excitotoxicity in the brain. J. Cereb. Blood Flow Metab. 20, 956–966.
depression, or the stress associated with depression, may have Alkharfy, K.M., Kellum, J.A., Matzke, G.R., 2000. Unintended immunomo-
positive effects on the course of illnesses comorbid with dulation. Part I. Effects of common clinical conditions on cytokine bio-
MDD. synthesis. Shock 13, 333–345.
48 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Allan, S.M., Parker, L.C., Collins, B., Davies, R., Luheshi, G.N., Rothwell, N.J., Anisman, H., Ravindran, A.V., Griffiths, J., Merali, Z., 1999a. Endocrine and
2000. Cortical cell death induced by IL-1 is mediated via actions in the cytokine correlates of major depression and dysthymia with typical or
hypothalamus of the rat. Proc. Natl. Acad. Sci. 97, 5580–5585. atypical features. Mol. Psychiatry 4, 182–188.
Allan, S.M., Tyrrell, P.J., Rothwell, N.J., 2005. Interleukin-1 and neuronal Anisman, H., Ravindran, A.V., Griffiths, J., Merali, Z., 1999b. Interleukin-1
injury. Nat. Rev. Immunol. 5, 629–640. beta production in dysthymia before and after pharmacotherapy. Biol.
Alonso, R., Griebel, G., Pavone, G., Stemmelin, J., Le Fur, G., Soubrié, P., 2004. Psychiatry 46, 1649–1655.
Blockade of CRF(1) or V(1b) receptors reverses stress-induced suppression Anisman, H., Hayley, S., Kelly, O., Borowski, T., Merali, Z., 2001. Psycho-
of neurogenesis in a mouse model of depression. Mol. Psychiatry 9, 278– genic, neurogenic and systemic stressor effects on plasma corticosterone
286. and behaviors: mouse strain-dependent outcomes. Behav. Neurosci. 115,
Altar, C.A., 1999. Neurotrophins and depression. Trends Pharamacol. Sci. 20, 443–454.
59–61. Anisman, H., Kokkinidis, L., Merali, Z., 2002. Further evidence for the
Alvarez, A., Cacabelos, R., Sanpedro, C., Garcı́a-Fantini, M., Aleixandre, M., depressive effects of cytokines: anhedonia and neurochemical changes.
2007. Serum TNF-alpha levels are increased and correlate negatively with Brain Behav. Immun. 16, 544–556.
free IGF-I in Alzheimer disease. Neurobiol. Aging 28, 533–536. Anisman, H., Hayley, S., Merali, Z., 2003. Cytokines and stress: sensitization
Alvarez, X.A., Franco, A., Fernández-Novoa, L., Cacabelos, R., 1996. Blood and cross-sensitization. Brain Behav. Immun. 17, 86–93.
levels of histamine, IL-1 beta, and TNF-alpha in patients with mild to Anisman, H., Du, L., Palkovits, M., Faludi, G., Kovacs, G.G., Szontagh-
moderate Alzheimer disease. Mol. Chem. Neuropathol. 29, 237–252. Kishazi, P., Merali, Z., Poulter, M.O., in press. Serotonin receptor subtype
Alves, S.H., Pinheiro, G., Motta, V., Landeira-Fernandez, J., Cruz, A.P., 2004. and p11 mRNA expression in stress-relevant brain regions of suicides and
Anxiogenic effects in the rat elevated plus-maze of 5-HT(2C) agonists into controls. J. Psychiatry Neurosci.
ventral but not dorsal hippocampus. Behav. Pharmacol. 15, 37–43. Anisman, H., Poulter, M.O., Gandhi, R., Merali, Z., Hayley, S., 2007a. Inter-
Amat, J., Matus-Amat, P., Watkins, L.R., Maier, S.F., 1998a. Escapable and feron-alpha effects are exaggerated when administered on a psychosocial
inescapable stress differentially alter extracellular levels of 5-HT in the stressor backdrop: cytokine, corticosterone and brain monoamine varia-
basolateral amygdala of the rat. Brain Res. 812, 113–120. tions. J. Neuroimmunol. 186, 45–53.
Amat, J., Matus-Amat, P., Watkins, L.R., Maier, S.F., 1998b. Escapable and Anisman, H., Prakash, P., Merali, Z., Poulter, M.O., 2007b. Corticotropin
inescapable stress differentially and selectively alter extracellular levels of releasing hormone receptor alterations elicited by acute and chronic unpre-
5-HT in the ventral hippocampus and dorsal periaqueductal gray of the rat. dictable stressor challenges in stressor-susceptible and resilient strains of
Brain Res. 797, 12–22. mice. Behav. Brain Res. 181, 180–190.
Amat, J., Tamblyn, J.P., Paul, E.D., Bland, S.T., Amat, P., Foster, A.C., Watkins, Annunziata, P., Cioni, C., Santonini, R., Paccagnini, E., 2002. Substance P
L.R., Maier, S.F., 2004. Microinjection of urocortin 2 into the dorsal raphe antagonist blocks leakage and reduces activation of cytokine-stimulated rat
nucleus activates serotonergic neurons and increases extracellular serotonin brain endothelium. J. Neuroimmunol. 131, 41–49.
in the basolateral amygdala. Neuroscience 129, 509–519. Antelman, S., Chiodo, L.A., 1984. Amphetamuine as a stressor. In: Iversen,
Amat, J., Baratta, M.V., Paul, E., Bland, S.T., Watlins, L.R., Maier, S.F., 2005. L.L., Iversen, S.D., Snyder, S.H. (Eds.), Handbook of Psychopharama-
Medial prefrontal cortex determines how stressor controllability affects cology. Plenum, New York.
behavior and dorsal raphe nucleus. Nat. Neurosci. 8, 365–371. Antoni, F.A., 1993. Vasopressinergic control of pituitary adrenocorticotropin
Amat, J., Paul, E., Zarza, C., Watlins, L.R., Maier, S.F., 2006. Previous secretion comes of age. Front. Neuroendocrinol. 14, 76–122.
experience with behavioral control over stress blocks the behavioral and Antonijevic, I.A., Murck, H., Frieboes, R.M., Horn, R., Brabant, G., Steiger, A.,
dorsal raphe nucleus activating effects of later uncontrollable stress: role of 1998. Elevated nocturnal profiles of serum leptin in patients with depres-
the ventral medial prefrontal cortex. J. Neurosci. 26, 13264–13272. sion. J. Psychiatry Res. 32, 403–440.
Andreasson, A., Arborelius, L., Erlanson-Albertsson, C., Lekander, M.A., 2007. Anwar, A., Zahid, A.A., Scheidegger, K.J., Brink, M., Delafontaine, P., 2002.
Putative role for cytokines in the impaired appetite in depression. Brain Tumor necrosis factor-alpha regulates insulin-like growth factor-1 and
Behav. Immun. 21, 147–152. insulin-like growth factor binding protein-3 expression in vascular smooth
Anguelova, M., Benkelfat, C., Turecki, G., 2003a. A systematic review of muscle. Circulation 105, 1220–1225.
association studies investigating genes coding for serotonin receptors and Arango, V., Underwood, M.D., Gubbi, A.V., Mann, J.J., 1995. Localized
the serotonin transporter. I. Affective disorders. Mol. Psychiatry 8, 574–591. alterations in pre- and postsynaptic serotonin binding sites in the
Anguelova, M., Benkelfat, C., Turecki, G., 2003b. A systematic review of ventrolateral prefrontal cortex of suicide victims. Brain Res. 688,
association studies investigating genes coding for serotonin receptors and 121–133.
the serotonin transporter. II. Suicidal behaviour. Mol. Psychiatry 8, 646– Arango, V., Underwood, M.D., Boldrini, M., Tamir, H., Kassir, S.A., Hsiung, S.,
653. et al., 2001. Serotonin 1A receptors, serotonin transporter binding and
Anisman, H., Matheson, K., 2005. Stress, anhedonia and depression: caveats serotonin transporter mRNA expression in the brainstem of depressed
concerning animal models. Neurosci. Biobehav. Rev. 29, 525–546. suicide victims. Neuropsychopharmacology 25, 892–903.
Anisman, H., Merali, Z., 1999. Anhedonic and anxiogenic effects of cytokine Arango, V., Huang, Y.Y., Underwood, M.D., Mann, J.J., 2003. Genetics
exposure. Adv. Exp. Med. Biol. 461, 199–233. of the serotonergic system in suicidal behavior. J. Psychiatry Res. 37,
Anisman, H., Sklar, L.S., 1979. Catecholamine depletion upon reexposure to 375–386.
stress: mediation of the escape deficits produced by inescapable shock. J. Arato, M., Banki, C.M., Bissette, G., Nemeroff, C.B., 1989. Elevated CSF CRF
Comp. Physiol. Psychol. 93, 610–625. in suicide victims. Biol. Psychiatry 25, 355–359.
Anisman, H., Zacharko, R.M., 1991. Multiple neurochemical and behavioral Arborelius, L., Owens, M.J., Plotsky, P.M., Nemeroff, C.B., 1999. The role of
consequences of stressors. Implications for depression. In: File, S.E. corticotropin-releasing factor in depression and anxiety disorders. J. Endo-
(Ed.), Psychopharmacology of Anxiolytics and Antidepressants. Perga- crinol. 160, 1–12.
mon Press, New York, pp. 57–82. Arimoto, T., Choi, D.Y., Lu, X., Liu, M., Nguyen, X.V., Zheng, N., Stewart,
Anisman, H., deCatanzaro, D., Remington, G., 1978. Escape performance C.A., Kim, H.C., Bing, G., 2006. Interleukin-10 protects against inflamma-
following exposure to inescapable shock; deficits in response initiation tion-mediated degeneration of dopaminergic neurons in substantia nigra.
and maintenance. J. Exp. Psychol: Anim. Behav. Proc. 4, 197–218. Neurobiol. Aging 28, 894–906.
Anisman, H., Zalcman, S., Zacharko, R.M., 1993. The impact of stressors on Arora, R.C., Meltzer, H.Y., 1989. Serotonergic measures in the brains of suicide
immune and central neurotransmitter activity: bidirectional communication. victims: 5-HT2 binding sites in the frontal cortex of suicide victims and
Rev. Neurosci. 4, 147–180. control subjects. Am. J. Psychiatry 146, 730–736.
Anisman, H., Kokkinidis, L., Borowski, T., Merali, Z., 1998. Differential effects Arranz, B., Cowburn, R., Eriksson, A., Vestling, M., Marcusson, J., 1992.
of IL-1, IL-2 and IL-6 on responding for rewarding lateral hypothalamic Gamma-aminobutyric acid-B (GABAB) binding sites in postmortem sui-
stimulation. Brain Res. 779, 177–187. cide brains. Neuropsychobiology 26, 33–36.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 49

Arranz, B., Eriksson, A., Mellerup, E., Plenge, P., Marcusson, J., 1994. Brain 5- Basterzi, A.D., Aydemir, C., Kisa, C., Aksaray, S., Tuzer, V., Yazici, K., Goka,
HT1A, 5-HT1D, and 5-HT2 receptors in suicide victims. Biol. Psychiatry E., 2005. IL-6 levels decrease with SSRI treatment in patients with major
35, 457–463. depression. Hum. Psychopharmacol. 20, 473–476.
Ashkenazi, A., Dixit, V.M., 1998. Death receptors: signaling and modulation. Battey, J., Wada, E., 1991. Two distinct receptor subtypes for mammalian
Science 281, 1305–1308. bombesin-like peptides. Trends Neurosci. 14, 524–528.
Asnis, G.M., De La Garza, 2006. Interferon-induced depression in chronic Baud, V., Karin, M., 2001. Signal transduction by tumor necrosis factor and its
hepatitis C: a review of its prevalence, risk factors, biology, and treatment relatives. Trends Cell Biol. 11, 372–377.
approaches. J. Clin. Gastroenterol. 40, 322–335. Bauer, S., Patterson, P.H., 2006. Leukemia inhibitory factor promotes neural
Asnis, G.M., McGinn, L.K., Sanderson, W.C., 1995. Atypical depression: stem cell self-renewal in the adult brain. J. Neurosci. 26, 12089–12099.
clinical aspects and noradrenergic function. Am. J. Psychiatry 152, 31–36. Bauer, S., Kerr, B.J., Patterson, P.H., 2007. The neuropoietic cytokine family in
Asnis, G.M., De La Garza, R., Kohn, S.R., Reinus, J.F., Henderson, M., Shah, J., development, plasticity, disease and injury. Nat. Rev. Neurosci. 8, 221–232.
2003. IFN-induced depression: a role for NSAIDs. Psychopharmacol. Bull. Bedard, T., Mountney, C., Kent, P., Anisman, H., Merali, Z., 2007. Role of
37, 29–50. gastrin-releasing peptide and neuromedin B in anxiety and fear-related
Atmaca, M., Kuloglu, M., Tezcan, E., Ustundag, B., Gecici, O., Firidin, B., behavior. Behav. Brain Res. 179, 133–140.
2002. Serum leptin and cholesterol values in suicide attempters. Neurop- Behan, D.P., Grigoriadis, D.E., Lovenberg, T., Chalmers, D., Heinrichs, S.,
sychobiology 45, 124–127. Liaw, C., De Souza, E.B., 1996. Neurobiology of corticotropin releasing
Attar-Levy, D., Martinot, J.L., Blin, J., Dao-Castellana, M.H., Crouzel, C., factor (CRF) receptors and CRF-binding protein: implications for the
Mazoyer, B., Poirier, M.F., Bourdel, M.C., Aymard, N., Syrota, A., Feline, treatment of CNS disorders. Mol. Psychiatry 1, 265–277.
A., 1999. The cortical serotonin2 receptors studied with positron-emis- Beitia, G., Garmendia, L., Azpiroz, A., Vegas, O., Brain, P.F., Arregi, A., 2005.
sion tomography and [18F]-setoperone during depressive illness and Time-dependent behavioral, neurochemical, and immune consequences of
antidepressant treatment with clomipramine. Biol. Psychiatry 45, 180– repeated experiences of social defeat stress in male mice and the ameli-
186. orative effects of fluoxetine. Brain Behav. Immun. 19, 530–539.
Austin, M.C., Janosky, J.E., Murphy, H.A., 2003. Increased corticotropin- Belanoff, J.K., Flores, B.H., Kalezhan, M., Sund, B., Schatzberg, A.F., 2001.
releasing hormone immunoreactivity in monoamine-containing pontine Rapid reversal of psychotic depression using mifepristone. J. Clin. Psy-
nuclei of depressed suicide men. Mol. Psychiatry 8, 324–332. chopharmacol. 21, 516–521.
Bagnoli, S., Cellini, E., Tedde, A., Nacmias, B., Piacentini, S., Bessi, V., Bracco, Benke, D., Michel, C., Mohler, H., 1997. GABA(A) receptors containing the
L., Sorbi, S., 2007. Association of IL10 promoter polymorphism in Italian alpha4-subunit: prevalence, distribution, pharmacology, and subunit archi-
Alzheimer’s disease. Neurosci. Lett. 418, 262–265. tecture in situ. J. Neurochem. 69, 806–814.
Bale, T.L., Vale, W.W., 2003. Increased depression like behaviors in cortico- Beratis, S., Katrivanou, A., Georgiou, S., Monastirli, A., Pasmatzi, E., Gourzis,
tropin-releasing factor receptor-2-deficient mice: sexually dichotomous P., Tsambaos, D., 2005. Major depression and risk of depressive sympto-
responses. Neuroscience 23, 5295–5301. matology associated with short-term and low-dose interferon-alpha treat-
Bale, T.L., Contarino, A.B., Smith, G.W., Chan, R., Gold, L.H., Sawchenko, ment. J. Psychosom. Res. 58, 15–18.
P.E., Koob, G.F., Vale, W.W., Lee, K.F., 2000. Mice deficient for cortico- Bertini, L.T., Kiss, J.Z., 1991. Hypophysiotrophic neurons are capable of
tropin-releasing hormone receptor-2 display anxiety-like behaviour and are altering the ratio of co-packaged neurohormones. Neuroscience 42, 237–
hypersensitive to stress. Nat. Genet. 24, 410–414. 244.
Bale, T.L., Picetti, R., Contarino, A., Koob, G.F., Vale, W.W., Lee, K.F., 2002. Betarbet, R., Sherer, T.B., MacKenzie, G., Garcia-Osuna, M., Panov, A.V.,
Mice deficient for both corticotropin-releasing factor receptor 1 (CRFR1) Greenamyre, J.T., 2000. Chronic systemic pesticide exposure reproduces
and CRFR2 have an impaired stress response and display sexually dichot- features of Parkinson’s disease. Nat. Neurosci. 3, 1301–1306.
omous anxiety-like behavior. J. Neurosci. 22, 193–199. Bhansali, P., Dunning, J., Singer, S.E., David, L., Schmauss, C., 2007. Early life
Bali, B., Kovacs, K.G., 2003. GABAergic control of neuropeptide gene stress alters adult serotonin 2C receptor pre-mRNA editing and expression
expression in parvocellular neurons of the hypothalamic paraventricular of the alpha subunit of the heterotrimeric G-protein G q. J. Neurosci. 27,
nucleus. Eur. J. Neurosci. 18, 1518–1526. 1467–1473.
Banks, W.A., 1999. Physiology and pathology of the blood–brain barrier: Bianchi, M., Sacerdote, P., Panerai, A.E., 1994. Fluoxetine reduces inflamma-
implications for microbial pathogenesis, drug delivery and neurodegenera- tory edema in the rat: involvement of the pituitary-adrenal axis. Eur. J.
tive disorders. J. Neurovirol. 5, 538–555. Pharmacol. 263, 81–84.
Banks, W.A., 2001. Cytokines, CVSs, and the blood–brain-barrier. In: Ader, Bik, W., Wolinska-Witort, E., Chmielowska, M., Baranowska-Bik, A.,
R., Felten, D.L., Cohen, N. (Eds.), Psychoneuroimmunology, vol. 2. Rusiecka-Kuczalek, E., Baranowska, B., 2004. Vasoactive intestinal peptide
Academic Press, New York, pp. 483–498. can modulate immune and endocrine responses during lipopolysaccharide-
Banks, W.A., 2006. The blood–brain barrier as a regulatory interface in the gut– induced acute inflammation. Neuroimmunomodulation 11, 358–364.
brain axes. Physiol. Behav. 89, 472–476. Billings, A.G., Moos, R.H., 1985. Life stressors and social resources affect
Banks, W.A., Ortz, L., Plotkin, S.R., Kasten, A.J., 1991. Human interleukin (IL) posttreatment outcomes among depressed patients. J. Abnorm. Psychol. 94,
1 alpha, murine IL-1 alpha and murine IL-2 beta are transported from blood 140–153.
to brain in the mouse by a shared saturable mechanism. J. Pharmacol. Exp. Birzniece, V., Backstrom, T., Johansson, I.M., Lindblad, C., Lundgren, P.,
Ther. 259, 988–996. Lofgren, M., Olsson, T., Ragagnin, G., Taube, M., Turkmen, S., Wahlstrom,
Bao, A.M., Meynen, G., Swaab, D.F., 2007. The stress system in depression and G., Wang, M.D., Wihlback, A.C., Zhu, D., 2006. Neuroactive steroid effects
neurodegeneration: focus on the human hypothalamus. Brain Res. Rev. 27 on cognitive functions with a focus on the serotonin and GABA systems.
(Epub ahead of print). Brain Res. Rev. 51, 212–239.
Baranowska-Bik, A., Bik, W., Wolinska-Witort, E., Chmielowska, M., Mar- Bissette, G., Klimek, V., Pan, J., Stockmeier, C., Ordway, G., 2003. Elevated
tynska, L., Baranowska, B., 2006. Can PACAP-38 modulate immune and concentrations of CRF in the locus coeruleus of depressed subjects.
endocrine responses during lipopolysaccharide (LPS)-induced acute Neuropsychopharmacology 28, 1328–1335.
inflammation? Ann. NY Acad. Sci. 1070, 156–160. Biver, F., Wikler, D., Lotstra, F., Damhaut, P., Goldman, S., Mendlewicz, J.,
Barger, S.W., Moerman, A.M., Mao, X., 2005. Molecular mechanisms of 1997. Serotonin 5-HT2 receptor imaging in major depression: focal changes
cytokine-induced neuroprotection: NFkappaB and neuroplasticity. Curr. in orbito-insular cortex. Br. J. Psychiatry 171, 444–448.
Pharm. Des. 11, 985–998. Black, P.H., 2002. Stress and the inflammatory response: a review of neurogenic
Bartanusz, V., Jezova, D., Bertini, L.T., Tilders, F.J., Aubry, J.M., Kiss, J.Z., inflammation. Brain Behav. Immun. 16, 622–653.
1993. Stress-induced increase in vasopressin and corticotropin-releasing Black, P.H., 2006. The inflammatory consequences of psychologic stress:
factor expression in hypophysiotrophic paraventricular neurons. Endocri- relationship to insulin resistance, obesity, atherosclerosis and diabetes
nology 132, 895–902. mellitus, type II. Med. Hypotheses 67, 879–891.
50 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Blalock, J.E., 1984. The immune system as a sensory organ. J. Immunol. 132, behavior in rats that is prevented by chronic antidepressant drug treatment.
1067–1070. Neuropsychopharmacology 33, 320–331.
Blalock, J.E., 1994. The syntax of immune-neuroendocrine communication. Bondy, B., 2007. Common genetic factors for depression and cardiovascular
Immun. Today 15, 504–511. disease. Dialogues Clin. Neurosci. 9, 19–28.
Blalock, J.E., 1999. Proopiomelanocortin and the immune-neuroendocrine Boos, H.B., Aleman, A., Cahn, W., Pol, H.H., Kahn, R.S., 2007. Brain volumes
connection. Ann. NY Acad. Sci. 885, 161–172. in relatives of patients with schizophrenia: a meta-analysis. Arch. Gen.
Blanc, G., Herve, D., Simon, H., Lisoprawski, A., Glowinski, J., Tassin, J.P., Psychiatry 64, 297–304.
1980. Response to stress of mesocortico-frontal dopaminergic neurones in Boothman, L., Raley, J., Denk, F., Hirani, E., Sharp, T., 2006. In vivo evidence
rats after long-term isolation. Nature 284, 265–267. that 5-HT(2C) receptors inhibit 5-HT neuronal activity via a GABAergic
Bland, S.T., Hargrave, D., Pepin, J.L., Amat, J., Watkins, L.R., Maier, S.F., mechanism. Br. J. Pharmacol. 149, 861–869.
2003a. Stressor controllability modulates stress-induced dopamine and Borowski, T., Kokkinidis, L., Merali, Z., Anisman, H., 1998. Lipopolysacchar-
serotonin efflux and morphine-induced serotonin efflux in the medial ide, central in vivo biogenic amine variations, and anhedonia. Neuroreport
prefrontal cortex. Neuropsychopharmacology 28, 1589–1596. 9, 3797–3802.
Bland, S.T., Twining, C., Watkins, L.R., Maier, S.F., 2003b. Stressor controll- Bossù, P., Ciaramella, A., Moro, M.L., Bellincampi, L., Bernardini, S., Federici,
ability modulates stress-induced serotonin but not dopamine efflux in the G., Trequattrini, A., Macciardi, F., Spoletini, I., Di Iulio, F., Caltagirone, C.,
nucleus accumbens shell. Synapse 49, 206–208. Spalletta, G., 2007. Interleukin 18 gene polymorphisms predict risk and
Bland, S.T., Schmid, M.J., Greenwood, B.N., Watkins, L.R., Maier, S.F., 2006. outcome of Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry 78, 807–
Behavioral control of the stressor modulates stress-induced changes in 811.
neurogenesis and fibroblast growth factor-2. Neuroreport 17, 593–597. Brambilla, P., Perez, J., Barale, F., Schettini, G., Soares, J.C., 2003. GABAergic
Bland, S.T., Tamlyn, J.P., Barrientos, R.M., Greenwood, B.N., Watkins, L.R., dysfunction in mood disorders. Mol. Psychiatry 8, 721–737.
Campeau, S., Day, H.E., Maier, S.F., 2007. Expression of fibroblast growth Brebner, K., Hayley, S., Merali, Z., Anisman, H., 2000. Synergistic effects of
factor-2 and brain-derived neurotrophic factor mRNA in the medial pre- interleukin-1, interleukin-6, and TNF-a: neuroendocrine, neurochemical
frontal cortex and hippocampus after uncontrollable or controllable stress. and behavioral changes. Neuropsychopharmacology 22, 566–580.
Neuroscience 144, 1219–1228. Brecht, S., Simler, S., Vergnes, M., Mielke, K., Marescaux, C., Herdegen, T.,
Bligh-Glover, W., Kolli, T.N., Shapiro-Kulnane, L., Dilley, G.E., Friedman, L., 1999. Repetitive electroconvulsive seizures induce activity of c-Jun N-
Balraj, E., et al., 2000. Serotonin transporter in the midbrain of suicide terminal kinase and compartment-specific desensitization of c-Jun phos-
victims with major depression. Biol. Psychiatry 15, 1015–1024. phorylation in the rat brain. Brain Res. Mol. Brain Res. 68, 101–108.
Block, F., Dihné, M., Loos, M., 2005. Inflammation in areas of remote changes Bregonzio, C., Armando, I., Ando, H., Jezova, M., Baiardi, G., Saavedra, J.M.,
following focal brain lesion. Prog. Neurobiol. 75, 342–365. 2004. Angiotensin II AT1 receptor blockade prevents gastric ulcers during
Bluthe, R.M., Pawlowski, M., Suarez, S., Parnet, P., Pittman, Q., Kelley, K.W., cold-restraint stress. Ann. NY Acad. Sci. 1018, 351–355.
Dantzer, R., 1994. Synergy between tumor necrosis factor alpha and Bremner, J.D., Narayan, M., Anderson, E.R., Staib, L.H., Miller, H.L., Charney,
interleukin-1 in the induction of sickness behavior in mice. Psychoneur- D.S., 2000. Hippocampal volume reduction in major depression. Am. J.
oendocrinology 19, 197–207. Psychiatry 157, 115–118.
Bluthe, R.M., Dantzer, R., Kelley, K.W., 1997. Central mediation of the effects Bret-Dibat, J.L., Bluthe, R.M., Kent, S., Kelley, K.W., Dantzer, R., 1995.
of interleukin-1 on social explorationand body weight in mice. Psycho- Lipopolysaccharide and interleukin-1 depress food-motivated behavior in
neuroendocrinology 22, 1–11. mice by a vagal-mediated mechanism. Brain Behav. Immun. 9, 242–246.
Bluthe, R.M., Kelley, K.W., Dantzer, R., 2006. Effects of insulin-like growth Briones-Aranda, A., Rocha, L., Picazo, O., 2005. Influence of forced swimming
factor-I on cytokine-induced sickness behavior in mice. Brain Behav. stress on 5-HT1A receptors and serotonin levels in mouse brain. Prog.
Immun. 20, 57–63. Neuropsychopharmacol. Biol. Psychiatry 29, 275–281.
Bocci, V., 1991. Interleukins: clinical pharmacokinetics and practical implica- Brooks-Kayal, A.R., Shumate, M.D., Jin, H., Lin, D.D., Rikhter, T.Y.,
tions. Clin. Pharmacokinet. 21, 274–284. Holloway, K.L., Coulter, D.A., 1999. Human neuronal gamma-aminobu-
Bokor, M., Farago, A., Garam, T., Malatinszky, G., Schnabel, R., 1993. Anti- tyric acid(A) receptors: coordinated subunit mRNA expression and
body-dependent cell-mediated cytotoxicity (ADCC) in Parkinson’s disease. functional correlates in individual dentate granule cells. J. Neurosci.
J. Neurol. Sci. 115, 47–50. 19, 8312–8318.
Bolanos-Jimenez, F., Manhaes de Casatro, R., Seguin, L., Cloez-Tayarani, I., Bruce, A.J., Boling, W., Kindy, M.S., Peschon, J., Kraemer, P.J., Carpenter,
Monneret, V., Drieu, K., Fillion, G., 1995. Effects of stress on the functional M.K., Holtsberg, F.W., Mattson, M.P., 1996. Altered neuronal and micro-
properties of pre- and postsynaptic 5-HT1B receptors in the brain. Eur. J. glial responses to excitotoxic and ischemic brain injury in mice lacking TNF
Pharmacol. 294, 531–540. receptors. Nat. Med. 2, 788–794.
Boldrini, M., Underwood, M.D., Mann, J.J., Arango, V., 2007. Serotonin-1A Brunson, K.L., Avishai-Eliner, S., Hatalski, C.G., Baram, T.Z., 2001. Neuro-
autoreceptor binding in the dorsal raphe nucleus of depressed suicides. J. biology of the stress response early in life: evolution of a concept and the
Psychiatry Res. 42, 433–442. role of corticotropin releasing hormone. Mol. Psychiatry 6, 647–656.
Bolin, L.M., Strycharska-Orczyk, I., Murray, R., Langston, J.W., Di Monte, D., Brustolim, D., Ribeiro-dos-Santos, R., Kast, R.E., Altschuler, E.L., Soares,
2002. Increased vulnerability of dopaminergic neurons in MPTP-lesioned M.B., 2006. A new chapter opens in anti-inflammatory treatments: the
interleukin-6 deficient mice. J. Neurochem. 83, 167–175. antidepressant bupropion lowers production of tumor necrosis factor-alpha
Bonaccorso, S., Puzella, A., Marino, V., Pasquini, M., Biondi, M., Artini, M., and interferon-gamma in mice. Int. Immunopharmacol. 6, 903–907.
Almerighi, C., Levrero, M., Egyed, B., Bosmans, E., Meltzer, H.Y., Maes, Bugajski, J., Gadek-Michalska, A., Bugajski, A.J., 2003. Effect of cycloox-
M., 2001. Immunotherapy with interferon-alpha in patients affected by ygenase inhibitors on the CRH-induced pituitary–adrenocortical activity
chronic hepatitis C induces an intercorrelated stimulation of the cytokine during crowding stress. J. Physiol. Pharmacol. 54, 99–108.
network and an increase in depressive and anxiety symptoms. Psychiatry Buller, K.M., Day, T.A., 2002. Systemic administration of interleukin-1beta
Res. 105, 45–55. activates select populations of central amygdala afferents. J. Comp. Neurol.
Bonaccorso, S., Puzella, A., Marino, V., Pasquini, M., Biondi, M., Artini, M., 452, 288–296.
Almerighi, C., Levrero, M., Egyed, B., Bosmans, E., Meltzer, H.Y., Maes, Buller, K., Xu, Y., Dayas, C., Day, T., 2001. Dorsal and ventral medullary
M., 2002. Increased depressive ratings in patients with hepatitis C receiving catecholamine cell groups contribute differentially to systemic interleukin-
interferon-alpha-based immunotherapy are related to interferon-alpha- 1beta-induced hypothalamic pituitary adrenal axis responses. Neuroendo-
induced changes in the serotonergic system. J. Clin. Psychopharmacol. crinology 73, 129–138.
22, 86–90. Buttini, M., Boddeke, H., 1995. Peripheral lipopolysaccharide stimulation
Bondi, C.O., Rodriguez, G., Gould, G.G., Frazer, A., Morilak, D.A., 2007. induces interleukin-1b messenger RNA in rat brain microglial cells. Neu-
Chronic unpredictable stress induces a cognitive deficit and anxiety-like roscience 65, 523–530.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 51

Buwalda, B., Kole, M.H., Veenema, A.H., Huininga, M., de Boer, S.F., Korte, Cardenas, H., Bolin, L.M., 2003. Compromised reactive microgliosis in MPTP-
S.M., Koolhaas, J.M., 2005. Long-term effects of social stress on brain and lesioned IL-6 KO mice. Brain Res. 985, 89–97.
behavior: a focus on hippocampal functioning. Neurosci. Biobehav. Rev. 29, Carroll, J.C., Boyce-Rustay, J.M., Millstein, R., Yang, R., Wiedholz, L.M.,
83–97 Epub 2004 Dec 8. Murphy, D.L., Holmes, A., 2007. Effects of mild early life stress on
Caballero, J., Hitchcock, M., Beversdorf, D., Scharre, D., Nahata, M., 2006. abnormal emotion-related behaviors in 5-HTT knockout mice. Behav.
Long-term effects of antidepressants on cognition in patients with Alzhei- Genet. 37, 214–222.
mer’s disease. J. Clin. Pharm. Ther. 31, 593–598. Caruso, C., Mohn, C., Karara, A.L., Rettori, V., Watanobe, H., Schiöth, H.B.,
Calcagni, E., Elenkov, I., 2006. Stress system activity, innate and T helper Seilicovich, A., Lasaga, M., 2004. Alpha-melanocyte-stimulating hormone
cytokines, and susceptibility to immune-related diseases. Ann. NY Acad. through melanocortin-4 receptor inhibits nitric oxide synthase and cycloox-
Sci. 1069, 62–76. ygenase expression in the hypothalamus of male rats. Neuroendocrinology
Caldji, C., Francis, D., Sharma, S., Plotsky, P.M., Meaney, M.J., 2000. The 79, 278–286.
effects of early rearing environment on the development of GABAA and Carver, C.S., Scheier, M.F., Weintraub, J.K., 1989. Assessing coping strategies:
central benzodiazepine receptor levels and novelty-induced fearfulness in a theoretically based approach. J. Pers. Soc. Psychol. 59, 73–81.
the rat. Neuropsychopharmacology 22, 219–229. Carvey, P.M., Chang, Q., Lipton, J.W., Ling, Z., 2003. Prenatal exposure to the
Caldji, C., Diorio, J., Anisman, H., Meaney, M.J., 2004. Maternal behavior bacteriotoxin lipopolysaccharide leads to long-term losses of dopamine
regulates benzodiazepine/GABAA receptor subunit expression in brain neurons in offspring: a potential, new model of Parkinson’s disease. Front.
regions associated with fear in BALB/cByJ and C57BL/6ByJ mice. Neu- Biosci. 8, 826–837.
ropsychopharmacology 29, 1344–1352. Casadevall, M., Saperas, E., Panés, J., Salas, A., Anderson, D.C., Malagelada,
Campbell, S., Marriott, M., Nahmias, C., MacQueen, G.M., 2004. Lower J.R., Piqué, J.M., 1999. Mechanisms underlying the anti-inflammatory
hippocampal volume in patients suffering from depression: a meta-analysis. actions of central corticotropin-releasing factor. Am. J. Physiol. 276,
Am. J. Psychiatry 161, 598–607. 1016–1026.
Canli, T., Qiu, M., Omura, K., Congdon, E., Haas, B.W., Amin, Z., Herrmann, Caso, J.R., Lizasoain, I., Lorenzo, P., Moro, M.A., Leza, J.C., 2006. The role of
M.J., Constable, R.T., Lesch, K.P., 2006. Neural correlates of epigenesis. tumor necrosis factor-alpha in stress-induced worsening of cerebral ische-
Proc. Natl. Acad. Sci. U.S.A. 103, 16033–16038. mia in rats. Neuroscience 142, 59–69.
Capuron, L., Ravaud, A., 1999. Prediction of the depressive effects of interferon Caso, J.R., Moro, M.A., Lorenzo, P., Lizasoain, I., Leza, J.C., 2007. Involve-
alfa therapy by patient’s initial affective state. N. Engl. J. Med. 340, 1370. ment of IL-1beta in acute stress-induced worsening of cerebral ischaemia in
Capuron, L., Ravaud, A., Dantzer, R., 2000. Early depressive symptoms in rats. Eur. Neuropsychopharmacol. 17, 600–607.
cancer patients receiving interleukin 2 and/or interferon alfa-2b therapy. J. Caspi, A., Sugden, K., Moffitt, T.E., Taylor, A., Craig, I.W., Harrington, H.,
Clin. Oncol. 18, 2143–2151. McClay, J., Mill, J., Martin, J., Braithwaite, A., Poulton, R., 2003. Influence
Capuron, L., Ravaud, A., Gualde, N., Bosmans, E., Dantzer, R., Maes, M., of life stress on depression: moderation by a polymorphism in the 5-HTT
Neveu, P.J., 2001. Association between immune activation and early gene. Science 301, 386–389.
depressive symptoms in cancer patients treated with interleukin-2-based Cassens, G., Roffman, M., Kuruc, A., Orsulak, P.J., Schildkraut, J.J., 1980.
therapy. Psychoneuroendocrinology 26, 797–808. Alterations in brain norepinephrine metabolism induced by environmental
Capuron, L., Hauser, P., Hinze-Selch, D., Miller, A.H., Neveu, P.J., 2002a. stimuli previously paired with inescapable shock. Science 209, 1138–1140.
Treatment of cytokine-induced depression. Brain Behav. Immun. 16, 575– Cassidy, E.M., Manning, D., Byrne, S., Bolger, E., Murray, F., Sharifi, N., et al.,
580. 2002. Acute effects of low-dose interferon-alpha on serum cortisol and
Capuron, L., Ravaud, A., Neveu, P.J., Miller, A.H., Maes, M., Dantzer, R., plasma interleukin-6. J. Psychopharmacol. 16, 230–234.
2002b. Association between decreased serum tryptophan concentrations Castanon, N., Leonard, B.E., Neveu, P.J., Yirmiya, R., 2002. Effects of
and depressive symptoms in cancer patients undergoing cytokine therapy. antidepressants on cytokine production and actions. Brain Behav. Immun.
Mol. Psychiatry 7, 468–473. 16, 569–574.
Capuron, L., Gumnick, J.F., Musselman, D.L., Lawson, D.H., Reemsnyder, A., Castanon, N., Konsman, J.P., Medina, C., Chauvet, N., Dantzer, R., 2003.
Nemeroff, C.B., Miller, A.H., 2002c. Neurobehavioral effects of interferon- Chronic treatment with the antidepressant tianeptine attenuates lipopoly-
alpha in cancer patients: phenomenology and paroxetine responsiveness of saccharide-induced Fos expression in the rat paraventricular nucleus and
symptom dimensions. Neuropsychopharmacology 26, 643–652. HPA axis activation. Psychoneuroendocrinology 28, 19–34.
Capuron, L., Neurauter, G., Musselman, D.L., Lawson, D.H., Nemeroff, C.B., Castillo, J., Moro, M.A., Blanco, M., Leira, R., Serena, J., Lizasoain, I.,
Fuchs, D., Miller, A.H., 2003a. Interferon-alpha-induced changes in tryp- Dávalos, A., 2003. The release of tumor necrosis factor-alpha is associated
tophan metabolism. relationship to depression and paroxetine treatment. with ischemic tolerance in human stroke. Ann. Neurol. 54, 811–819.
Biol. Psychiatry 54, 906–914. Castrén, E., Võikar, V., Rantamäki, T., 2007. Role of neurotrophic factors in
Capuron, L., Raison, C.L., Musselman, D.L., Lawson, D.H., Nemeroff, C.B., depression. Curr. Opin. Pharmacol. 7, 18–21.
Miller, A.H., 2003b. Association of exaggerated HPA axis response to the Catania, A., Delgado, R., Airaghi, L., Cutuli, M., Garofalo, L., Carlin, A.,
initial injection of interferon-alpha with development of depression during Demitri, M.T., Lipton, J.M., 1999. alpha-MSH in systemic inflammation.
interferon-alpha therapy. Am. J. Psychiatry 160, 1342–1345. Central and peripheral actions. Ann. NY Acad. Sci. 885, 183–187.
Capuron, L., Ravaud, A., Miller, A.H., Dantzer, R., 2004. Baseline mood and Cavus, I., Dman, R.S., 2003. Influence of estradiol, stress, and 5-HT2A agonist
psychosocial characteristics of patients developing depressive symptoms treatment on brain-derived neurotrophic factor expression in female rats.
during interleukin-2 and/or interferon-alpha cancer therapy. Brain Behav. Biol. Psychiatry 54, 59–69.
Immun. 18, 205–213. Cenci, M.A., Kalen, P., Mandel, R.J., Bjorklund, A., 1992. Regional differences
Capuron, L., Pagnoni, G., Demetrashvili, M., Woolwine, B.J., Nemeroff, in the regulation of dopamine and noradrenaline release in the medial frontal
C.B., Berns, G.S., Miller, A.H., 2005. Anterior cingulate activation and cortex, nucleus accumbens and caudate putamen: a microdialysis study in
error processing during interferon-alpha treatment. Biol. Psychiatry 58, the rat. Brain Res. 581, 217–228.
190–196. Chan-Palay, V., 1993. Depression and dementia in Parkinson’s disease. Cate-
Caraceni, A., Gangeri, L., Martini, C., Belli, F., Brunelli, C., Baldini, M., cholamine changes in the locus ceruleus, a basis for therapy. Adv. Neurol.
Mascheroni, L., Lenisa, L., Cascinelli, N., 1998. Neurotoxicity of inter- 60, 438–446.
feron-alpha in melanoma therapy: results from a randomized controlled Chao, C.C., Molitor, T.W., Hu, S., 1993. Neuroprotective role of IL-4 against
trial. Cancer 83, 482–489. activated microglia. J. Immunol. 151, 1473–1481.
Caraceni, A., Martini, C., Belli, F., Mascheroni, L., Rivoltini, L., Arienti, F., Charnay, Y., Cusin, I., Vallet, P.G., Muzzin, P., Rohner-Jeanrenaud, F., Bouras,
Cascinelli, N., 1992. Neuropsychological and neurophysiological assess- C., 2000. Intracerebroventricular infusion of leptin decreases serotonin
ment of the central effects of interleukin-2 administration. Eur. J. Cancer transporter binding sites in the frontal cortex of the rat. Neurosci. Lett.
29A, 1266–1269. 283, 89–92.
52 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Charney, D.S., 2004. Psychobiological mechanisms of resilience and vulner- Coyle, J.T., Duman, R.S., 2003. Finding the intracellular signaling pathways
ability: implications for successful adaptation to extreme stress. Am. J. affected by mood disorder treatments. Neuron 38, 157–160.
Psychiatry 161, 195–216. Craighead, M.W., Boutin, H., Middlehurst, K.M., Allan, S.M., Brooks, N.,
Cheetham, S.C., Crompton, M.R., Katona, C.L., Parker, S.J., Horton, R.W., Kimber, I., Rothwell, N.J., 2000. Influence of corticotrophin releasing factor
1988. Brain GABAA/benzodiazepine binding sites and glutamic acid on neuronal cell death in vitro and in vivo. Brain Res. 881, 139–143.
decarboxylase activity in depressed suicide victims. Brain Res. 460, Cremers, T.I., Rea, K., Bosker, F.J., Wikstrom, H.V., Hogg, S., Mork, A.,
114–123. Westerink, B.H., 2007. Augmentation of SSRI effects on serotonin by 5-
Chiarini, A., Dal Pra, I., Whitfield, J.F., Armato, U., 2006. The killing of HT2C antagonists: mechanistic studies. Neuropsychopharmacology 32,
neurons by beta-amyloid peptides, prions, and pro-inflammatory cytokines. 1550–1557.
Ital. J. Anat. Embryol. 111, 221–246. Crocker, S.J., Lamba, W.R., Smith, P.D., Callaghan, S.M., Slack, R.S., Anis-
Chiou, S.H., Chen, S.J., Peng, C.H., Chang, Y.L., Ku, H.H., Hsu, W.M., Ho, man, H., Park, D.S., 2001. c-Jun mediates axotomy-induced dopamine
L.L., Lee, C.H., 2006. Fluoxetine up-regulates expression of cellular neuron death in vivo. Proc. Natl. Acad. Sci. 98, 13385–13390.
FLICE-inhibitory protein and inhibits LPS-induced apoptosis in hippo- Cross, J.A., Cheetham, S.C., Crompton, M.R., Katona, C.L., Horton, R.W.,
campus-derived neural stem cell. Biochem. Biophys. Res. Commun. 343, 1988. Brain GABA(B) binding sites in depressed suicide victims. Psychia-
391–400. try Res. 26, 119–129.
Chronwall, B.M., Pisano, J.J., Bishop, J.F., Moody, T.W., O’Donohue, T.L., Crow, T.J., Cross, A.J., Cooper, S.J., Deakin, J.F., Ferrier, I.N., Johnson, J.A.,
1985. Biochemical and histochemical characterization of ranatensin immu- Joseph, M.H., Owen, F., Poulter, M., Lofthouse, R., 1984. Neurotransmitter
noreactive peptides in rat brain: Lack of coexistence with bombesin/GRP. receptors and monoamine metabolites in the brains of patients with Alz-
Brain Res. 338, 97–113. heimer-type dementia and depression, and suicides. Neuropharmacology
Churchill, L., Taishi, P., Wang, M., Brandt, J., Cearley, C., Rehman, A., 23, 1561–1569.
Krueger, J.M., 2006. Brain distribution of cytokine mRNA induced by Cryan, J.F., Slattery, D.A., 2007. Animal models of mood disorders: recent
systemic administration of interleukin-1beta or tumor necrosis factor alpha. developments. Curr. Opin. Psychiatry 20, 1–7.
Brain Res. 1120, 64–73. Cullinan, W.E., 2000. GABA(A) receptor subunit expression within hypophy-
Cichy, J., Rose-John, S., Pryjma, J., Travis, J., 1996. Effect of soluble inter- siotropic CRH neurons: a dual hybridization histochemical study. J. Comp.
leukin-6 receptor on interleukin-6 synthesis in human skin fibroblasts. Neurol. 419, 344–351.
Biochem. Biophys. Res. Commun. 227, 318–321. Cullinan, W.E., Wolfe, T.J., 2000. Chronic stress regulates levels of mRNA
Cizza, G., Romagni, P., Lotsikas, A., Lam, G., Rosenthal, N.E., Chrousos, G.P., transcripts encoding beta subunits of the GABA(A) receptor in the rat stress
2005. Plasma leptin in men and women with seasonal affective disorder and axis. Brain Res. 887, 118–124.
in healthy matched controls. Horm. Metab. Res. 37, 45–48. Cunningham, E.T., De Souza, E.B., 1993. Interleukin 1 receptors in the brain
Claes, S.J., 2004. CRH, stress, and major depression: a psychobiological and endocrine tissues. Immunol. Today 14, 171–176.
interplay. Vitam. Horm. 69, 117–150. Curtis, J., Finkbeiner, S., 1999. Sending signals from the synapse to the
Coco, M.L., Kuhn, C.M., Ely, T.D., Kilts, C.D., 1992. Selective activation of nucleus: possible roles for CaMK, Ras/ERK, and SAPK pathways in the
mesoamygdaloid dopamine neurons by conditioned stress: attenuation by regulation of synaptic plasticity and neuronal growth. J. Neurosci. Res.
diazepam. Brain Res. 50, 39–47. 58, 88–95.
Cole, R.L., Sawchenko, P.E., 2002. Neurotransmitter regulation of cellular Czlonkowska, A., Ciesielska, A., Gromadzka, G., Kurkowska-Jastrzebska, I.,
activation and neuropeptide gene expression in the paraventricular nucleus 2005. Estrogen and cytokines production—the possible cause of gender
of the hypothalamus. J. Neurosci. 22, 959–969. differences in neurological diseases. Curr. Pharm. Des. 11, 1017–1030.
Collin, M., Håkansson-Ovesjö, M.L., Misane, I., Ogren, S.O., Meister, B., 2000. Czlonkowska, A., Kurkowska-Jastrzebska, I., Czlonkowski, A., Peter, D.,
Decreased 5-HT transporter mRNA in neurons of the dorsal raphe nucleus Stefano, G., 2002. Immune processes in the pathogenesis of Parkinson’s
and behavioral depression in the obese leptin-deficient ob/ob mouse. Res. disease: a potential role for microglia and nitric oxide. Med. Sci. Monit. 8,
Mol. Brain Res. 81, 51–61. 65–77.
Colotta, F., Dower, S.K., Sims, J.E., Mantovani, A., 1994. The type II ‘decoy’ Dale, R.C., Church, A.J., Surtees, R.A., Lees, A.J., Adcock, J.E., Harding, B.,
receptor: a novel regulatory pathway for interleukin 1. Immunol. Today 15, Nevill, B.G., Giovannoni, G., 2004. Encephalitis lethargica syndrome: 20
562–566. new cases and evidence of basal ganglia autoimmunity. Brain 127, 21–33.
Constant, A., Castera, L., Dantzer, R., Couzigou, P., de Ledinghen, V., Demotes- Dallman, M.F., Pecoraro, N., Akana, S.F., La Fleur, S.E., Gomez, F., Houshyar,
Mainard, J., Henry, C., 2005. Mood alterations during interferon-alfa H., Bell, M.E., Bhatnagar, S., Laugero, K.D., Manalo, S., 2003. Chronic
therapy in patients with chronic hepatitis C: evidence for an overlap stress and obesity: a new view of ‘‘comfort food’’. Proc. Natl. Acad. Sci.
between manic/hypomanic and depressive symptoms. J. Clin. Psychiatry 100, 11696–11701.
66, 1050–1057. Damas, P., Canivet, J.L., de Groote, D., Vrindts, Y., Albert, A., Franchimont, P.,
Contarino, A., Dellu, F., Koob, G.F., Smith, G.W., Lee, K.F., Vale, W.W., Gold, Lamy, M., 1997. Sepsis and serum cytokine concentrations. Crit. Care Med.
L.H., 1999. Reduced anxiety-like and cognitive performance in mice 25, 405–412.
lacking the corticotropin-releasing factor receptor 1. Brain Res. 835, 1–9. Dandona, P., Dhindsa, S., Ghanim, H., Chaudhuri, A., 2007. Angiotensin II and
Cook, S.C., Wellman, C.L., 2004. Chronic stress alters dendritic morphology in inflammation: the effect of angiotensin-converting enzyme inhibition and
rat medial prefrontal cortex. J. Neurobiol. 60, 236–248. angiotensin II receptor blockade. J. Hum. Hypertens. 21, 20–27.
Cosentino, M., Fietta, A.M., Ferrari, M., Rasini, E., Bombelli, R., Carcano, E., Danner, M., Kasl, S.V., Abramson, J.L., Vaccarino, V., 2003. Association
Saporiti, F., Meloni, F., Marino, F., Lecchini, S., 2007. Human CD4+CD25+ between depression and elevated C-reactive protein. Psychosom. Med.
regulatory T cells selectively express tyrosine hydroxylase and contain 65, 347–356.
endogenous catecholamines subserving an autocrine/paracrine inhibitory Dantzer, R., 2001a. Cytokine-induced sickness behavior: where do we stand.
functional loop. Blood 109, 632–642. Brain Behav. Immun. 15, 7–24.
Cotter, R.L., Burke, W.J., Thomas, V.S., Potter, J.F., Zheng, J., Gendelman, Dantzer, R., 2001b. Cytokine-induced sickness behavior: mechanisms and
H.E., 1999. Insights into the neurodegenerative process of Alzheimer’s implications. Ann. NY Acad. Sci. 933, 222–234.
disease: a role for mononuclear phagocyte-associated inflammation and Dantzer, R., 2006. Cytokine, sickness behavior, and depression. Neurol. Clin.
neurotoxicity. J. Leukoc. Biol. 65, 416–427. 24, 441–460.
Cover, H., Irwin, M., 1994. Immunity and depression: Insomnia, retardation and Dantzer, R., Bluthe, R.M., Aubert, A., Goodall, G., Bret-Dibat, J.-L., Kent, S.,
reduction of natural killer cell activity. J. Behav. Med. 17, 217–223. Goujon, E., Laye, S., Parnet, P., Kelley, K.W., 1999a. Mechanisms of the
Cowan, K.J., Storey, K.B., 2003. Mitogen-activated protein kinases: new behavioral effects of cytokines. Adv. Exp. Med. Biol. 461, 83–106.
signaling pathways functioning in cellular responses to environmental Dantzer, R., Wolman, E., Vitkovic, L., Yirmiya, 1999b. Cytokines and depres-
stress. J. Exp. Biol. 206, 1107–1115. sion: fortuitous or causative association? Mol. Psychiatry 4, 328–332.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 53

Dantzer, R., Konsman, J.-P., Bluthe, R.M., Kelley, K.W., 2000. Neural and retarded depression: relation with plasma vasopressin and cortisol. Neu-
humoral pathways of communication from the immune system to the brain: ropsychopharmacology 28, 140–147.
parallel or convergent? Auton. Neurosci. Basic Clin. 85, 60–65. Dejda, A., Sokołowska, P., Nowak, J.Z., 2005. Neuroprotective potential of
Davidson, J., Pelton, S., 1986. Forms of atypical depression and their response three neuropeptides PACAP, VIP and PHI. Pharmacol. Rep. 57, 307–320.
to antidepressant drugs. Psychiatry Res. 17, 87–95. Delgado, M., 2003. VIP: a very important peptide in T helper differentiation.
Davis, M., 1992. The role of the amygdala in fear and anxiety. Ann. Rev. Trends Immunol. 24, 221–224.
Neurosci. 15, 353–375. Delgado, M., Ganea, D., 2003. Vasoactive intestinal peptide prevents activated
Dawson, D.A., Martin, D., Hallenbeck, J.M., 1996. Inhibition of tumor necrosis microglia-induced neurodegeneration under inflammatory conditions:
factor-alpha reduces focal cerebral ischemic injury in the spontaneously potential therapeutic role in brain trauma. FASEB J. 17, 1922–1924.
hypertensive rat. Neurosci. Lett. 218, 41–44. Delongis, A., Holtzman, S., 2005. Coping in context: the role of stress, social
Day, H.E., Curran, E.J., Watson Jr., S.J., Akil, H., 1999. Distinct neurochemical support, and personality in coping. J. Pers. 73, 1633–1656.
populations in the rat central nucleus of the amygdala and bed nucleus of the Denicoff, K.D., Rubinow, D.R., Papa, M.Z., Simpson, L., Seipp, L.A., Lotze,
stria terminalis: evidence for their selective activation by interleukin-1beta. M.T., Chang, A.E., Rosenstein, D., Rosenberg, S.A., 1987. The neuropsy-
J. Comp. Neurol. 413, 113–128. chiatric effects of treatment with interleukin-2 and lymphokine-activated
Dayas, C.V., Buller, K.M., Day, T.A., 1999. Neuroendocrine responses to an killer cells. Ann. Int. Med. 107, 293–300.
emotional stressor: evidence for involvement of the medial but not the Deroche, D., Marinelli, M., Maccari, S., Le Moal, M., Simon, H., Piazza, P.V.,
central amygdala. Eur. J. Neurosci. 11, 2312–2322. 1995. Stress-induced sensitization and glucocorticoids. 1. Sensitization of
Dayas, C.V., Buller, K.M., Crane, J.W., Xu, Y., Day, T.A., 2001a. Stressor dopamine-dependent locomotor effects of amphetamine and morphine
categorization: acute physical and psychological stressors elicit distinctive depends on stress-induced corticosterone secretion. J. Neurosci. 15,
recruitment patterns in the amygdala and in medullary noradrenergic cell 7181–7188.
groups. Eur. J. Neurosci. 14, 1143–1152. Deuschle, M., Blum, W.F., Englaro, P., Schweiger, U., Weber, B., Pflaum, C.D.,
Dayas, C.V., Buller, K.M., Day, T.A., 2001b. Medullary neurones regulate Heuser, I., 1996. Plasma leptin in depressed patients and healthy controls.
hypothalamic corticotropin releasing factor cell responses to an emotional Horm. Metab. Res. 28, 714–717.
stressor. Neuroscience 105, 707–719. Deutch, A.Y., Roth, R.H., 1990. The determinants of stress-induced activation
De Bellis, M.D., Gold, P.W., Geracioti Jr., T.D., Listwak, S.J., Kling, M.A., of the prefrontal cortical dopamine system. Prog. Brain Res. 85,
1993. Association of fluoxetine treatment with reductions in CSF concen- 367–402.
trations of corticotropin-releasing hormone and arginine vasopressin in Deutch, A.Y., Tam, S.-Y., Roth, R.H., 1991. Footshock and conditioned stress
patients with major depression. Am. J. Psychiatry 150, 656–657. increase 3,4-dihydroxyphenylacetic acid (DOPAC) in the ventral tegmental
de Goeij, D.C., Kvetnansky, R., Whitnall, M.H., Jezova, D., Berkenbosch, F., area but not substantia nigra. Brain Res. 333, 143–146.
Tilders, F.J., 1991. Repeated stress-induced activation of corticotropin- Dhabhar, F.S., 2000. Acute stress enhances while chronic stress suppresses skin
releasing factor neurons enhances vasopressin stores and colocalization immunity. The role of stress hormones and leukocyte trafficking. Ann. NY
with corticotropin-releasing factor in the median eminence of rats. Neu- Acad. Sci. 917, 876–893.
roendocrinology 53, 150–159. Dibbs, Z., Kurrelmeyer, K., Kalra, D., Seta, Y., Wang, F., Bozkurt, B.,
de Jong, J.C., van den Berg, P.B., Tobi, H., de Jong-van den Berg, L.T., 2003. Baumgarten, G., Sivasubramanian, N., Mann, D.L., 1999. Cytokines in
Combined use of SSRIs and NSAIDs increases the risk of gastrointestinal heart failure: pathogenetic mechanisms and potential treatment. Proc.
adverse effects. Br. J. Clin. Pharmacol. 55, 591–595. Assoc. Am. Physicians 111, 423–428.
de Kloet, E.R., Oitzl, M.S., Joels, M., 1999. Stress and cognition: are corti- Dickerson, S.S., Kemeny, M.E., 2004. Acute stressors and cortisol responses: a
costeroids good or bad guys? Trends Neurosci. 22, 422–426. theoretical integration and synthesis of laboratory research. Psychol. Bull.
de Kloet, E.R., 2003. Hormones, brain and stress. Endocr. Regul. 37, 51–68. 130, 355–391.
de Kloet, E.R., Joëls, M., Holsboer, F., 2005. Stress and the brain: from Dickman, M.S., 2001. von Economo encephalitis. Arch. Neurol. 58, 1696–
adaptation to disease. Nat. Rev. Neurosci. 6, 463–475. 1698.
De La Garza, R., Asnis, G.M., 2003. The non-steroidal anti-inflammatory drug Dieperink, E., Willenbring, M., Ho, S.B., 2000. Neuropsychiatric symptoms
diclofenac sodium attenuates IFN-alpha induced alterations to monoamine associated with hepatitis C and interferon alpha: a review. Am. J. Psychiatry
turnover in prefrontal cortex and hippocampus. Brain Res. 977, 70–79. 157, 867–876.
De La Garza, R., Fabrizio, K.R., Radoi, G.E., Vlad, T., Asnis, G.M., 2004. Dieperink, E., Ho, S.B., Thuras, P., Willenbring, M.L., 2003. A prospective
Diclofenac sodium attenuates lipopolysaccharide-induced alterations to study of neuropsychiatric symptoms associated with interferon-alpha-2b
reward behavior and corticosterone release. Behav. Brain. Res. 149, 77–85. and ribavirin therapy for patients with chronic hepatitis C. Psychosomatics
De La Garza, R., Asnis, G.M., Pedrosa, E., Stearns, C., Migdal, A.L., Reinus, 44, 104–112.
J.F., Paladugu, R., Vemulapalli, S., 2005. Recombinant human interferon- Dinan, T.G., O’Brien, S., Lavelle, E., Scott, L.V., 2004. Further neuroendocrine
alpha does not alter reward behavior, or neuroimmune and neuroendocrine evidence of enhanced vasopressin V3 receptor responses in melancholic
activation in rats. Prog. Neuropsychopharmacol. Biol. Psychiatry 29, depression. Psychol. Med. 34, 169–172.
781–792. Dinarello, C.A., 1987. The biology of interleukin 1 and comparison to tumor
De Maeyer, E., De Meyer-Guignard, J., 1998. Interferons. In: Thomson, A. necrosis factor. Immunol. Lett. 16, 227–231.
(Ed.), The Cytokine Handbook. 3rd ed. Academic Press, San Diego, CA, Ding, Q., Dimayuga, E., Keller, J.N., 2007. Oxidative damage, protein synth-
USA. esis, and protein degradation in Alzheimer’s disease. Curr. Alzheimer Res.
de Pablos, R.M., Villarán, R.F., Argüelles, S., Herrera, A.J., Venero, J.L., Ayala, 4, 73–79.
A., Cano, J., Machado, A., 2006. Stress increases vulnerability to inflam- Djavadian, R.L., 2004. Serotonin and neurogenesis in the hippocampal dentate
mation in the rat prefrontal cortex. J. Neurosci. 26, 5709–5719. gyrus of adult mammals. Acta Neurobiol. Exp. 64, 189–200.
De Sarro, G., Gareri, P., Sinopoli, V.A., David, E., Rotiroti, D., 1997. Com- Doherty, M.D., Gratton, A., 1992. High-speed chronoamperometric measure-
parative, behavioural and electrocortical effects of tunor necrosis factor- ments of mesolimbic and nigostriatal dopamine release associated with
alpha and interleukin-1 microinjected into the locus coeruleus of rat. Life repeated daily stress. Brain Res. 586, 295–302.
Sci. 60, 555–564. Doherty, M.D., Gratton, A., 1996. Medial prefrontal cortical D1 receptor
de Vries, H.E., Blom-Roosemalen, M.C., van Oosten, M., de Boer, A.G., van modulation of the meso-accumbens dopamine responmse to stress: an
Berkel, T.J., Breimer, D.D., Kuiper, J., 1996. The influence of cytokines electrochemical study in freely-behaving rats. Brain Res. 715, 86–97.
on the integrity of the blood–brain barrier in vitro. J. Neuroimmunol. 64, Domin, J., Polak, J.M., Bloom, S.R., 1988. The distribution and biological
37–43. effects of neuromedins B and U. Ann. NY Acad. Sci. 547, 391–403.
de Winter, R.F., van Hemert, A.M., DeRijk, R.H., Zwinderman, K.H., Fran- Dopp, J.M., Sarafian, T.A., Spinella, F.M., Kahn, M.A., Shau, H., de Vellis, J.,
khuijzen-Sierevogel, A.C., Wiegant, V.M., Goekoop, J.G., 2003. Anxious- 2002. Expression of the p75 TNF receptor is linked to TNF-induced
54 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

NFkappaB translocation and oxyradical neutralization in glial cells. Neu- Dwivedi, Y., Rizavi, H.S., Pandey, G.N., 2006. Antidepressants reverse corti-
rochem. Res. 27, 1535–1542. costerone-mediated decrease in brain-derived neurotrophic factor expres-
Douglas, K.M., Taylor, A.J., O’Malley, P.G., 2004. Relationship between sion: differential regulation of specific exons by antidepressants and
depression and C-reactive protein in a screening population. Psychosom. corticosterone. Neuroscience 139, 1017–1029.
Med. 66, 679–683. Eigler, A., Sinha, B., Hartmann, G., Endres, S., 1997. Taming TNF: strategies to
Drevets, W.C., 2000a. Functional anatomical abnormalities in limbic and restrain this proinflammatory cytokine. Immunol. Today 18, 487–492.
prefrontal cortical structures in major depression. Prog. Brain Res. 126, Eikelenboom, P., Veerhuis, R., Scheper, W., Rozemuller, A.J., van Gool, W.A.,
413–431. Hoozemans, J.J., 2006. The significance of neuroinflammation in under-
Drevets, W.C., 2000b. Neuroimaging studies of mood disorders. Biol. Psy- standing Alzheimer’s disease. J. Neural Transm. 113, 1685–1695.
chiatry 48, 813–829. Eikelis, N., Esler, M., Barton, D., Dawood, T., Wiesner, G., Lambert, G., 2006.
Drevets, W.C., 2001. Neuroimaging and neuropathological studies of depres- Reduced brain leptin in patients with major depressive disorder and in
sion: implications for the cognitive-emotional features of mood disorders. suicide victims. Mol. Psychiatry 11, 800–801.
Curr. Opin. Neurobiol. 11, 240–249. Elenkov, I.J., Iezzoni, D.G., Daly, A., Harris, A.G., Chrousos, G.P., 2005.
Drevets, W.C., Thase, M.E., Moses-Kolko, E.L., Price, J., Frank, E., Kupfer, Cytokine dysregulation, inflammation and well-being. Neuroimmunomo-
D.J., Mathis, C., 2007. Serotonin-1A receptor imaging in recurrent depres- dulation 12, 255–269.
sion: replication and literature review. Nucl. Med. Biol. 34, 865–877. Emoto, H., Koga, C., Ishii, H., Yokoo, H., Yoshida, M., Tanaka, M., 1993a. A
Drevets, W.C., Frank, E., Price, J.C., Kupfer, D.J., Holt, D., Greer, P.J., Huang, CRF antagonist attenuates stress-induced increases in NA turnover in
Y., Gautier, C., Mathis, C., 1999. PET imaging of serotonin 1A receptor extended brain regions in rats. Brain Res. 627, 171–176.
binding in depression. Biol. Psychiatry 46, 1375–1387. Emoto, H., Tanaka, M., Koga, C., Yokoo, H., Tsuda, A., Yoshida, M., 1993b.
Drevets, W.C., Frank, E., Price, J.C., Kupfer, D.J., Greer, P.J., Mathis, C., 2000. Corticotropin-releasing factor activates the noradrenergic neuron system in
Serotonin type-1A receptor imaging in depression. Nucl. Med. Biol. 27, the rat brain. Pharmacol. Biochem. Behav. 45, 419–422.
499–507. Emoto, H., Yokoo, H., Yoshida, M., Tanaka, M., 1993c. Corticotropin-releasing
D’Sa, C., Duman, R.S., 2002. Antidepressants and neuroplasticity. Bipolar factor enhances noradrenaline release in the rat hypothalamus assessed by
Disord. 4, 183–194. intracerebral microdialysis. Brain Res. 601, 286–288.
Du, L., Faludi, G., Palkovits, M., Demeter, E., Bakish, D., Lapierre, Y.D., Endler, N.S., Parker, J.D., 1990. Multidimensional assessment of coping: a
Sotonyi, P., Hrdina, P.D., 1999. Frequency of long allele in serotonin critical evaluation. J. Pers. Soc. Psychol. 58, 844–854.
transporter gene is increased in depressed suicide victims. Biol. Psychiatry Engelborghs, S., De Deyn, P.P., 1997. The neurochemistry of Alzheimer’s
46, 196–201. disease. Acta Neurol. Belg. 97, 67–84.
Duman, R.S., 2004. Depression: a case of neuronal life and death? Biol. Eom, D.S., Choi, W.S., Oh, Y.J., 2004. Bcl-2 enhances neurite extension via
Psychiatry 56, 140–145. activation of c-Jun N-terminal kinase. Biochem. Biophys. Res. Commun.
Duman, R.S., Monteggia, L.M., 2006. A neurotrophic model for stress-related 314, 377–381.
mood disorders. Biol. Psychiatry 59, 1116–1127. Ericsson, A., Kovacs, K.J., Sawchenko, P.E., 1994. A functional anatomical
Duman, R.S., Malberg, J., Thome, J., 1999. Neural plasticity to stress and analysis of central pathways subserving the effects of interleukin-1 on
antidepressant treatment. Biol. Psychiatry 46, 1181–1191. stress-related neuroendocrine neurons. J. Neurosci. 14, 89–913.
Duman, R.S., Malberg, J., Nakagawa, S., D’Sa, C., 2000. Neuronal plasticity Escriba, P.V., Ozaita, A., Garcia-Sevilla, J.A., 2004. Increased mRNA expres-
and survival in mood disorders. Biol. Psychiatry 48, 732–739. sion of alpha2A-adrenoceptors, serotonin receptors and mu-opioid
Duman, R.S., Nakagawa, S., Malberg, J., 2001. Regulation of adult receptors in the brains of suicide victims. Neuropsychopharmacology
neurogenesis by antidepressant treatment. Neuropsychopharmacology 29, 1512–1521.
25, 836–844. Esel, E., Ozsoy, S., Tutus, A., Sofuoglu, S., Kartalci, S., Bayram, F., Kokbudak,
Duman, C.H., Schlesinger, L., Kodama, M., Russell, D.S., Duman, R.S., 2007. Z., Kula, M., 2005. Effects of antidepressant treatment and of gender on
A role for MAP kinase signaling in behavioral models of depression and serum leptin levels in patients with major depression. Prog. Neuropsycho-
antidepressant treatment. Biol. Psychiatry 61, 661–670. pharmacol. Biol. Psychiatry 29, 565–570.
Dunn, A.J., Wan, J., Ando, T., 1999. Effects of cytokines on cerebral neuro- Esposito, P., Gheorghe, D., Kandere, K., Pang, X., Connolly, R., Jacobson, S.,
transmission: comparison with the effects of stress. Adv. Exp. Med. Biol. Theoharides, T.C., 2001. Acute stress increases permeability of the
461, 117–128. blood–brain-barrier through activation of brain mast cells. Brain Res.
Dunzendorfer, S., Meierhofer, C., Wiedermann, C.J., 1998. Signaling in 888, 117–127.
neuropeptide-induced migration of human eosinophils. J. Leukoc. Biol. Esposito, E., Mazzon, E., Muià, C., Meli, R., Sessa, E., Cuzzocrea, S., 2007.
64, 828–834. Splanchnic ischemia and reperfusion injury is reduced by genetic or
Duvarci, S., Pare, D., 2007. Glucocorticoids enhance the excitability of pharmacological inhibition of TNF-alpha. J. Leukoc. Biol. 81, 1032–1043.
principal basolateral amygdala neurons. J. Neurosci. 27, 4482–4491. Evans, S.J., Choudary, P.V., Neal, C.R., Li, J.Z., Vawter, M.P., Tomita, H.,
Dwivedi, Y., Rizavi, H.S., Roberts, R.C., Conley, R.C., Tamminga, C.A., Lopez, J.F., Thompson, R.C., Meng, F., Stead, J.D., Walsh, D.M., Myers,
Pandey, G.N., 2001. Reduced activation and expression of ERK1/2 MAP R.M., Bunney, W.E., Watson, S.J., Jones, E.G., Akil, H., 2004. Dysregula-
kinase in the post-mortem brain of depressed suicide subjects. J. Neuro- tion of the fibroblast growth factor system in major depression. Proc. Natl.
chem. 77, 916–928. Acad. Sci. U.S.A. 101, 15506–15511.
Dwivedi, Y., Conley, R.R., Roberts, R.C., Tamminga, C.A., Pandey, G.N., 2002. Fadda, F., Argiolas, A., Melis, M.R., Tissari, A.H., Onali, P.L., Gessa, G.L.,
[(3)H]cAMP binding sites and protein kinase a activity in the prefrontal 1978. Stress induced increase in 3,4-dihydroxyphenylacetic acid (DOPAC)
cortex of suicide victims. Am. J. Psychiatry 159, 66–73. levels in the cerebral cortex and in nucleus accumbens: reversal by
Dwivedi, Y., Rizavi, H.S., Conley, R.R., Roberts, R.C., Tamminga, C.A., diazepam. Life Sci. 23, 2219–2224.
Pandey, G.N., 2003. Altered gene expression of brain-derived neurotrophic Faludi, G., Du, L., Palkovits, M., Antal, B., Sotonyi, P., Hrdina, P.D., 2000.
factor and receptor tyrosine kinase B in postmortem brain of suicide Serotonin transporter, serotonin-2A receptor and tryptophan hydroxylase
subjects. Arch. Gen. Psychiatry 60, 804–815. gene polymorphisms in depressed suicide victims. Neurobiology 8,
Dwivedi, Y., Mondal, A.C., Shukla, P.K., Rizavi, H.S., Lyons, J., 2004. Altered 269–271.
protein kinase a in brain of learned helpless rats: effects of acute and Fan, Y., Liu, Z., Weinstein, P.R., Fike, J.R., Liu, J., 2007. Environmental
repeated stress. Biol. Psychiatry 56, 30–40. enrichment enhances neurogenesis and improves functional outcome after
Dwivedi, Y., Mondal, A.C., Payappagoudar, G.V., Rizavi, H.S., 2005. Differ- cranial irradiation. Eur. J. Neurosci. 25, 38–46.
ential regulation of serotonin (5HT)2A receptor mRNA and protein levels Farrar, W.L., Kilian, P.L., Ruff, M.R., Hill, J.M., Pert, C.B., 1987. Visualization
after single and repeated stress in rat brain: role in learned helplessness and characterization of interleukin-1 receptors in brain. J. Immunol. 139,
behavior. Neuropsychopharmacology 48, 204–214. 459–463.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 55

Fava, G.A., Ruini, C., Belaise, C., 2007. The concept of recovery in major Froger, N., Palazzo, E., Boni, C., Hanoun, N., Saurini, F., Joubert, C., Dutriez-
depression. Psychol. Med. 37, 307–317. Casteloot, I., Enache, M., Maccari, S., Barden, N., Cohen-Salmon, C.,
Feenstra, M.G.P., Botterblom, M.H.A., Van Uum, J.F.M., 1995. Novelty- Hamon, M., Lanfumey, L., 2004. Neurochemical and behavioral alterations
induced increase in dopamine release in the rat prefrontal cortex in vivo: in glucocorticoid receptor-impaired transgenic mice after chronic mild
inhibition by diazepam. Neurosci. Lett. 189, 81–84. stress. J. Neurosci. 24, 2787–2796.
Felger, J.C., Alagbe, O., Hu, F., Mook, D., Freeman, A.A., Sanchez, M.M., Fulton, S., Richard, D., Woodside, B., Shizgal, P., 2004. Food restriction and
Kalin, N.H., Ratti, E., Nemeroff, C.B., Miller, A.H., 2007. Effects of leptin impact brain reward circuitry in lean and obese Zucker rats. Behav.
interferon-alpha on rhesus monkeys: a nonhuman primate model of Brain Res. 155, 319–329.
cytokine-induced depression. Biol. Psychiatry 62, 1324–1333 (Epub Fulton, S., Woodside, B., Shizgal, P., 2000. Modulation of brain reward circuitry
2007). by leptin. Science 287, 125–128.
Ferder, L., Inserra, F., Martı́nez-Maldonado, M., 2006. Inflammation and the Fumagalli, F., Molteni, R., Calabrese, F., Frasca, A., Racagni, G., Riva, M.A.,
metabolic syndrome: role of angiotensin II and oxidative stress. Curr. 2005. Chronic fluoxetine administration inhibits extracellular signal-regu-
Hypertens. Rep. 8, 191–198. lated kinase 1/2 phosphorylation in rat brain. J. Neurochem. 93, 1551–1560.
Ferger, B., Leng, A., Mura, A., Hengerer, B., Feldon, J., 2004. Genetic ablation Furukawa, T.A., Streiner, D.L., Young, L.T., 2002. Antidepressant and benzo-
of tumor necrosis factor-alpha (TNF-alpha) and pharmacological inhibition diazepine for major depression. Cochrane Database Syst. Rev. (1),
of TNF-synthesis attenuates MPTP toxicity in mouse striatum. J. Neuro- CD001026.
chem. 89, 822–833. Gandhi, R., Hayley, S., Gibb, J., Merali, Z., Anisman, H., 2007. Influence of
Fernyhough, P., Smith, D.R., Schapansky, J., Van Der Ploeg, R., Gardiner, N.J., poly I:C on sickness behaviors, plasma cytokines, corticosterone and central
Tweed, C.W., Kontos, A., Freeman, L., Purves-Tyson, T.D., Glazner, G.W., monoamine activity: moderation by social stressors. Brain Behav. Immun.
2005. Activation of nuclear factor-kappaB via endogenous tumor necrosis 21, 477–489.
factor alpha regulates survival of axotomized adult sensory neurons. J. Gao, H.M., Hong, J.S., Zhang, W., Liu, B., 2003a. Synergistic dopaminergic
Neurosci. 25, 1682–1690. neurotoxicity of the pesticide rotenone and inflammogen lipopolysacchar-
Ferri, C.C., Yuill, E.A., Ferguson, A.V., 2005. Interleukin-1beta depolarizes ide: relevance to the etiology of Parkinson’s disease. J. Neurosci. 23, 1228–
magnocellular neurons in the paraventricular nucleus of the hypothalamus 1236.
through prostaglandin-mediated activation of a non selective cationic Gao, H.M., Liu, B., Zhang, W., Hong, J.S., 2003b. Critical role of microglial
conductance. Regul. Pept. 129, 63–71. NADPH oxidase-derived free radicals in the in vitro MPTP model of
Feuerstein, G.Z., Wang, X., Barone, F.C., 1997. Inflammatory gene expression Parkinson’s disease. FASEB 17, 1954–1956.
in cerebral ischemia and trauma. Potential new therapeutic targets. Ann. NY Garcia, J., Ahmadi, A., Wonnacott, A., Sutcliffe, W., Nagga, K., Soderkvist, P.,
Acad. Sci. 825, 179–193. Marcusson, J., 2006. Association of insulin-like growth factor-1 receptor
Finlay, J.M., Zigmond, M.J., Abercrombie, E.D., 1995. Increased dopamine and polymorphism in dementia. Dement. Geriatr. Cogn. Disord. 22, 439–444.
norepinephrine release in medial prefrontal cortex induced by acute and Garrido, M.M., Martin, S., Ambrosio, E., Fuentes, J.A., Manzanares, J., 1998.
chronic stress: effects of diazepam. Neuroscience 64, 619–628. Role of corticotropin-releasing hormone in gastrin-releasing peptide-
Finlay, J.M., Jedema, H.P., Rabinovic, A.D., Mana, M.J., Zigmond, M.J., Sved, mediated regulation of corticotropin and corticosterone secretion in male
A.F., 1997. Impact of corticotropin-releasing hormone on extracellular rats. Neuroendocrinology 68, 116–122.
norepinephrine in prefrontal cortex after chronic cold stress. J. Neurochem. Gass, P., Kretz, O., Wolfer, D.P., Berger, S., Tronche, F., Reichardt, H.M.,
69, 144–150. Kellendonk, C., Lipp, H.P., Schmid, W., Schütz, G., 2000. Genetic disrup-
Flores, C., Stewart, J., 2000. Basic fibroblast growth factor as a mediator of the tion of mineralocorticoid receptor leads to impaired neurogenesis and
effects of glutamate in the development of long-lasting sensitization to granule cell degeneration in the hippocampus of adult mice. EMBO
stimulant drugs: studies in the rat. Psychopharmacology (Berl) 151, 152– Rep. 1, 447–451.
165. Gecici, O., Kuloglu, M., Atmaca, M., Tezcan, A.E., Tunckol, H., Emül, H.M.,
Flores, C., Stewart, J., Salmaso, N., Zhang, Y., Boksa, P., 2002. Astrocytic basic Ustundag, B., 2005. High serum leptin levels in depressive disorders with
fibroblast growth factor expression in dopaminergic regions after perinatal atypical features. Psychiat. Clin. Neurosci. 59, 736–738.
anoxia. Biol. Psychiatry 52, 362–370. Gendek-Kubiak, H., 2002. Effect of intramuscular application of selected
Fontenot, M.B., Kaplan, J.R., Manuck, S.B., Arango, V., Mann, J.J., 1995. neuropeptides on morphology of muscle. Cell Mol. Biol. Lett. 7, 1059–
Long-term effects of chronic social stress on serotonergic indices in the 1064.
prefrontal cortex of adult male cynomolgus macaques. Brain Res. 705, Geracioti, T.D., Loosen, P.T., Orth, D.N., 1997. Low cerebrospinal fluid
105–108. corticotropin-releasing hormone concentrations in eucortisolemic depres-
Francis, J., Zhang, Z.H., Weiss, R.M., Felder, R.B., 2004. Neural regulation of sion. Biol. Psychiatry 42, 165–174.
the proinflammatory cytokine response to acute myocardial infarction. Am. Gibbs, J., Fauser, D.J., Rowe, E.A., Rolls, B.J., Rolls, E.T., Maddison, S.P.,
J. Physiol. Heart Circ. Physiol. 287, 791–797. 1979. Bombesin suppresses feeding in rats. Nature 282, 208–210.
Frasure-Smith, N., Lesperance, F., 2005. Reflections on depression as a cardiac Gibb, J.E., Ghandi, R., Hayley, S., Poulter, M.O., Anisman, H., 2005. Synergy
risk factor. Psychosom. Med. 67, S19–S25. between immune activation and psychosocial stressors: Behavioral and
Frasure-Smith, N., Lespérance, F., Irwin, M.R., Sauvé, C., Lespérance, J., neurochemical effects. Abstract presented at the Society for Neuroscience
Théroux, P., 2007. Depression, C-reactive protein and two-year major 25th Annual Conference, Washington, DC.
adverse cardiac events in men after acute coronary syndromes. Biol. Gilbertson, M.W., Shenton, M.E., Ciszewski, A., Kasai, K., Lasko, N.B., Orr,
Psychiatry (Epub ahead of print). S.P., Pitman, R.K., 2002. Smaller hippocampal volume predicts pathologic
Freudenthal, R., Romano, A., Routtenberg, A., 2004. Transcription factor NF- vulnerability to psychological trauma. Nat. Neurosci. 5, 1242–1247.
kappaB activation after in vivo perforant path LTP in mouse hippocampus. Gilmor, M.L., Skelton, K.H., Nemeroff, C.B., Owens, M.J., 2003. The effects of
Hippocampus 14, 677–683. chronic treatment with the mood stabilizers valproic acid and lithium on
Frodl, T., Meisenzahl, E.M., Zetzsche, T., Born, C., Groll, C., Jäger, M., corticotropin-releasing factor neuronal systems. J. Pharmacol. Exp. Ther.
Leinsinger, G., Bottlender, R., Hahn, K., Möller, H.J., 2002. Hippocampal 305, 434–439.
changes in patients with a first episode of major depression. Am. J. Gisslinger, H., Svoboda, T., Clodi, M., Gilly, B., Ludwig, H., Havelec, L.,
Psychiatry 159, 1112–1118. Luger, A., 1993. Interferon-alpha stimulates the hypothalamic–pituitary–
Frodl, T., Meisenzahl, E.M., Zetzsche, T., Höhne, T., Banac, S., Schorr, C., adrenal axis in vivo and in vitro. Neuroendocrinology 57, 489–495.
Jäger, M., Leinsinger, G., Bottlender, R., Reiser, M., Möller, H.J., 2004. Glazer, I., Rivest, S., 2004. Glucocorticoids: protectors of the brain during
Hippocampal and amygdala changes in patients with major depressive innate immune responses. Neuroscientist 10, 538–552.
disorder and healthy controls during a 1-year follow-up. J. Clin. Psychiatry Goehler, L.E., Erisir, A., Gaykema, R.P., 2006. Neural–immune interface in the
65, 492–499. rat area postrema. Neuroscience 140, 1415–1434.
56 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Goekoop, J.G., de Winter, R.P., de Rijk, R., Zwinderman, K.H., Frankhuijzen- Grundy, P.L., Harbuz, M.S., Jessop, D.S., Lightman, S.L., Sharples, P.M., 2001.
Sierevogel, A., Wiegant, V.M., 2006. Depression with above-normal plasma The hypothalamo-pituitary–adrenal axis response to experimental traumatic
vasopressin: validation by relations with family history of depression and brain injury. J. Neurotrauma 18, 1373–1381.
mixed anxiety and retardation. Psychiatry Res. 141, 201–211. Guillin, O., Diaz, J., Carroll, P., Griffon, N., Schwartz, J.C., Sokoloff, P., 2001.
Gold, P.W., Chrousos, G.P., 2002. Organization of the stress system and its BDNF controls dopamine D3 receptor expression and triggers behavioural
dysregulation in melancholic and atypical depression: high vs low CRH/NE sensitization. Nature 411, 86–89.
states. Mol. Psychiatry 7, 254–725. Gullestad, L., Aukrust, P., 2001. The cytokine network in heart failure:
Gold, P.W., Drevets, W.C., Charney, D.S., 2002. New insights into the role of pathogenetic importance and potential therapeutic targets. Heart Fail Monit.
cortisol and the glucocorticoid receptor in severe depression. Biol. Psy- 2, 8–13.
chiatry 52, 381–385. Gur, T.L., Conti, A.C., Holden, J., Bechtholt, A.J., Hill, T.E., Lucki, I., Malberg,
Gonzalez-Rey, E., Delgado, M., 2005. Role of vasoactive intestinal peptide in J.E., Blendy, J.A., 2007. cAMP response element-binding protein deficiency
inflammation and autoimmunity. Curr. Opin. Invest. Drugs 6, 1116–1123. allows for increased neurogenesis and a rapid onset of antidepressant
Gonzalez-Rey, E., Delgado, M., 2007. Vasoactive intestinal peptide and reg- response. J. Neurosci. 27, 7860–7868.
ulatory T-cell induction: a new mechanism and therapeutic potential for Gurevich, I., Englander, M.T., Adlersberg, M., Siegal, N.B., Schmauss, C.,
immune homeostasis. Trends Mol. Med. 13, 241–251. 2002. Modulation of serotonin 2C receptor editing by sustained changes in
Goodnick, P.J., Hernandez, M., 2000. Treatment of depression in comorbid serotonergic neurotransmission. J. Neurosci. 22, 10529–10532.
medical illness. Expert Opin. Pharmacother. 1, 1367–1384. Gutierrez, E.G., Banks, W.A., Kastin, A.J., 1993. Murine tumor necrosis factor
Gordon, J.A., Hen, R., 2004. The serotonergic system and anxiety. Neuromol. alpha is transported from blood to brain in the mouse. J. Neuroimmunol. 47,
Med. 5, 27–40. 169–176.
Goshen, I., Kreisel, T., Ben-Menachem-Zidon, O., Licht, T., Weidenfeld, J., Haack, M., Hinze-Selch, D., Fenzel, T., Kraus, T., Kuhn, M., Schuld, A.,
Ben-Hur, T., Yirmiya, R., 2007. Brain interleukin-1 mediates chronic stress- Pollmacher, T., 1999. Plasma levels of cytokines and soluble cytokine
induced depression in mice via adrenocortical activation and hippocampal receptors in psychiatric patients upon hospital admission: effects of con-
neurogenesis suppression. Mol. Psychiatry, doi:10.1038/sj.mp.4002055. founding factors and diagnosis. J. Psychiatry Res. 33, 407–418.
Gottesman, I.I., Gould, T.D., 2003. The endophenotype concept in psychiatry: Haga, S., Akai, K., Ishii, T., 1989. Demonstration of microglial cells in and
etymology and strategic intentions. Am. J. Psychiatry 160, 636–645. around senile (neuritic) plaques in the Alzheimer brain. An immunohisto-
Govindarajan, A., Rao, B.S., Nair, D., Trinh, M., Mawjee, N., Tonegawa, S., chemical study using a novel monoclonal antibody. Acta Neuropathol. 77,
Chattarji, S., 2006. Transgenic brain-derived neurotrophic factor expression 569–755.
causes both anxiogenic and antidepressant effects. Proc. Natl. Acad. Sci. Hakansson, A., Westberg, L., Nilsson, S., Buervenich, S., Carmine, A., Holm-
U.S.A. 103, 13208–13213. berg, B., Sydow, O., Olson, L., Johnels, B., Eriksson, E., Nissbrandt, H.,
Gravanis, A., Margioris, A.N., 2005. The corticotropin-releasing factor (CRF) 2005. Investigation of genes coding for inflammatory components in
family of neuropeptides in inflammation: potential therapeutic applications. Parkinson’s disease. Mov. Disord. 20, 569–573.
Curr. Med. Chem. 12, 1503–1512. Haleem, D.J., Parveen, T., 1994. Brain regional serotonin synthesis following
Gray, T.S., 1990. The organization and possible function of amygdaloid adaptation to repeated restraint. Neuroreport 5, 1785–1788.
corticotropin-releasing factor pathways. In: De Souza, E.B., Nemeroff, Hammack, S.E., Richey, K.J., Schmid, M.J., LoPresti, M.L., Watkins, L.R.,
C.B. (Eds.), Corticotropin-releasing Factor: Basic and Clinical Studies of a Maier, S.F., 2002. The role of corticotropin-releasing hormone in the dorsal
Neuropeptide. CRC Press, Boca Raton, pp. 53–68. raphe nucleus in mediating the behavioral consequences of uncontrollable
Greenfeder, S.A., Nunes, P., Kwee, L., Labow, M., Chizzonite, R.A., Ju, G., stress. J. Neurosci. 22, 1020–1026.
1995. Molecular cloning and characterization of a second subunit of the Hammack, S.E., Schmid, M.J., LoPresti, M.L., Der-Avakian, A., Pellymounter,
interleukin 1 receptor complex. J. Biol. Chem. 270, 13757–13765. M.A., Foster, A.C., Watkins, L.R., Maier, S.F., 2003. Corticotropin releasing
Greenwood, B.N., Strong, P.V., Foley, T.E., Thompson, R.S., Fleshner, M., hormone type 2 receptors in the dorsal raphe nucleus mediate the behavioral
2007. Learned helplessness is independent of levels of brain-derived consequences of uncontrollable stress. J. Neurosci. 23, 1019–1025.
neurotrophic factor in the hippocampus. Neuroscience 144, 1193–1208. Harden, L.M., du Plessis, I., Poole, S., Laburn, H.P., 2006. Interleukin-6 and
Gresch, P.J., Sved, A.F., Zigmond, M.J., Finlay, J.M., 1994. Stress-induced leptin mediate lipopolysaccharide-induced fever and sickness behavior.
sensitization of dopamine and norepinephrine efflux in medial prefrontal Physiol. Behav. 89, 146–155.
cortex of the rat. J. Neurochem. 63, 575–583. Hart, B.L., 1988. Biological basis of the behavior of sick animals. Neurosci.
Griffin, W.S., Liu, L., Li, Y., Mrak, R.E., Barger, S.W., 2006. Interleukin-1 Biobehav. Rev. 12, 123–137.
mediates Alzheimer and Lewy body pathologies. J. Neuroinflammation 16, Hartai, Z., Klivenyi, P., Janaky, T., Penke, B., Dux, L., Vecsei, L., 2005.
3–5. Kynurenine metabolism in multiple sclerosis. Acta Neurol. Scand. 112,
Griffin, W.S., Sheng, J.G., Roberts, G.W., Mrak, R.E., 1995. Interleukin-1 93–96.
expression in different plaque types in Alzheimer’s disease: significance in Hartman, M.E., O’Connor, J.C., Godbout, J.P., Minor, K.D., Mazzocco, V.R.,
plaque evolution. J. Neuropathol. Exp. Neurol. 54, 276–281. Freund, G.G., 2004. Insulin receptor substrate-2-dependent interleukin-4
Griffiths, J., Ravindran, A.V., Merali, Z., Anisman, H., 2000. Dysthymia: signaling in macrophages is impaired in two models of type 2 diabetes
neurochemical and behavioral perspectives. Mol. Psychiatry 5, 242–261. mellitus. J. Biol. Chem. 279, 28045–28050.
Griswold, D.E., Webb, E.F., Badger, A.M., Gorycki, P.D., Levandoski, P.A., Harvey, B.H., Naciti, C., Brand, L., Stein, D.J., 2003. Endocrine, cognitive
Barnette, M.A., Grous, M., Christensen, S., Torphy, T.J., 1998. SB 207499 and hippocampal/cortical 5HT 1A/2A receptor changes evoked by a
(Ariflo), a second generation phosphodiesterase 4 inhibitor, reduces tumor time-dependent sensitisation (TDS) stress model in rats. Brain Res.
necrosis factor alpha and interleukin-4 production in vivo. J. Pharmacol. 983, 97–107.
Exp. Ther. 287, 705–711. Harvey, J., 2007. Leptin: a diverse regulator of neuronal function. J. Neuro-
Groenink, L., van Bogaert, M.J., van der Gugten, J., Oosting, R.S., Olivier, B., chem. 100, 307–313.
2003. 5-HT1A receptor and 5-HT1B receptor knockout mice in stress and Hasler, G., Drevets, W.C., Manji, H.K., Charney, D.S., 2004. Discovering
anxiety paradigms. Behav. Pharmacol. 14, 369–383. endophenotypes for major depression. Neuropsychopharmacology 29,
Grønli, J., Bramham, C., Murison, R., Kanhema, T., Fiske, E., Bjorvatn, B., 1765–1781.
Ursin, R., Portas, C.M., 2006. Chronic mild stress inhibits BDNF protein Hayley, S., Anisman, H., 2005. Multiple mechanisms of cytokine action in
expression and CREB activation in the dentate gyrus but not in the neurodegenerative and psychiatric states: neurochemical and molecular
hippocampus proper. Pharmacol. Biochem. Behav. 85, 842–849. substrates. Curr. Pharm. Des. 11, 947–962.
Gross-Isseroff, R., Biegon, A., Voet, H., Weizman, A., 1998. The suicide brain: Hayley, S., Brebner, K., Lacosta, S., Merali, Z., Anisman, H., 1999a. Sensitiza-
a review of postmortem receptor/transporter binding studies. Neurosci. tion to the effects of tumor necrosis factor-a; neuroendocrine, central
Biobehav. Rev. 22, 653–661. monoamine and behavioral variations. J. Neurosci. 19, 5654–5665.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 57

Hayley, S., Staines, W.A., Merali, Z., Anisman, H., 1999b. Time-dependent Heuser, I., Bissette, G., Dettling, M., Schweiger, U., Gotthardt, U., Schmider, J.,
sensitization effects associated with tumor necrosis factor-a: behavioral and Lammers, C.H., Nemeroff, C.B., Holsboer, F., 1998. Cerebrospinal fluid
neurochemical processes. Neuroimmunomodulation 6, 224. concentrations of corticotropin-releasing hormone, vasopressin, and soma-
Hayley, S., Borowski, T., Merali, Z., Anisman, H., 2001a. Central monoamine tostatin in depressed patients and healthy controls: response to amitriptyline
activity in genetically distinct strains of mice following a psychogenic treatment. Depress. Anxiety 8, 71–79.
stressor: effects of predator exposure. Brain Res. 892, 293–300. Heuser, I., Yassouridis, A., Holsboer, F., 1994. The combined dexamethasone/
Hayley, S., Staines, W.M., Merali, Z., Anisman, H., 2001b. Time-dependent CRH test: a refined laboratory test for psychiatric disorders. J. Psychiatry
sensitization of corticotropin-releasing hormone, arginine vasopressin and Res. 28, 341–356.
c-fos immunoreactivity within the mouse brain in response to tumor Hill, J.M., Cuasay, K., Abebe, D.T., 2007. Vasoactive intestinal peptide
necrosis factor-alpha. Neuroscience 106, 137–148. antagonist treatment during mouse embryogenesis impairs social behavior
Hayley, S., Lacosta, S., Merali, Z., van Rooijen, N., Anisman, H., 2001c. and cognitive function of adult male offspring. Exp. Neurol. 206, 101–113.
Central monoamine and plasma corticosterone changes induced by a Hillhouse, E.W., Kida, S., Iannotti, F., 1998. Middle cerebral artery occlusion in
bacterial endotoxin: sensitization and cross-sensitization effects. Eur. J. the rat causes a biphasic production of immunoreactive interleukin-1beta in
Neurosci. 13, 1155–1165. the cerebral cortex. Neurosci. Lett. 249, 177–179.
Hayley, S., Merali, Z., Anisman, H., 2002. Cytokines and sensitization: Con- Hilsabeck, R.C., Hassanein, T.I., Ziegler, E.A., Carlson, M.D., Perry, W., 2005.
tribution to depression. Acta Neuropsychiat. 14, 322–335. Effect of interferon-alpha on cognitive functioning in patients with chronic
Hayley, S., Kelly, O., Anisman, H., 2002a. Murine tumor necrosis factor a hepatitis C. J. Int. Neuropsychol. Soc. 11, 16–22.
sensitizes plasma corticosoterone and manifestation of shock: modulation Himmerich, H., Binder, E.B., Künzel, H.E., Schuld, A., Lucae, S., Uhr, M.,
by histamine. J. Neuroimmunol. 131, 60–69. Pollmächer, T., Holsboer, F., Ising, M., 2006. Successful antidepressant
Hayley, S., Wall, P., Anisman, H., 2002b. Sensitization to the neuroendocrine. therapy restores the disturbed interplay between TNF-alpha system and
Central monoamine and behavioral effects of murine tumor necrosis factor- HPA axis. Biol. Psychiatry 60, 882–888.
a: peripheral and central mechanisms. Eur. J. Neurosci. 15, 1061–1076. Himmerich, H., Zimmermann, P., Ising, M., Kloiber, S., Lucae, S., Kunzel,
Hayley, S., Crocker, S.J., Smith, P.D., Shree, T., Jackson-Lewis, V., Przedborski, H.E., Binder, E.B., Holsboer, F., Uhr, M., 2007. Changes in the hypotha-
S., Mount, M., Slack, R., Anisman, H., Park, D.S., 2004a. Regulation of lamic–pituitary–adrenal axis and leptin levels during antidepressant treat-
dopaminergic loss by Fas in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyri- ment. Neuropsychobiology 55, 28–35.
dine model of Parkinson’s disease. J. Neurosci. 24, 2045–2053. Ho, W.Z., Kaufman, D., Uvaydova, M., Douglas, S.D., 1996. Substance P
Hayley, S., Kelly, O.P., Anisman, H., 2004b. Corticosterone changes in response augments interleukin-10 and tumor necrosis factor-alpha release by human
to stressors, acute and protracted actions of tumor necrosis factor-a, and cord blood monocytes and macrophages. J. Neuroimmunol. 71, 73–80.
lipopolysaccharide treatments in mice lacking the tumor necrosis factor-a Hollis, J.H., Evans, A.K., Bruce, K.P., Lightman, S.L., Lowry, C.A., 2006.
p55 receptor gene. Neuroimmunomodulation 11, 241–246. Lipopolysaccharide has indomethacin-sensitive actions on Fos expression
Hayley, S., Poulter, M., Merali, Z., Anisman, H., 2005. The pathogenesis of in topographically organized subpopulations of serotonergic neurons. Brain
clinical depression: stressor- and cytokine-induced alterations of neuro- Behav. Immun. 20, 569–577.
plasticity. Neuroscience 135, 659–678. Holmes, A., Murphy, D.L., Crawley, J.N., 2002. Reduced aggression in mice
Hedtjärn, M., Leverin, A.L., Eriksson, K., Blomgren, K., Mallard, C., Hagberg, lacking the serotonin transporter. Psychopharmacology 161, 160–167.
H., 2002. Interleukin-18 involvement in hypoxic-ischemic brain injury. J. Holmes, A., Yang, R.J., Lesch, K.P., Crawley, J.N., Murphy, D.L., 2003a. Mice
Neurosci. 22, 5910–5919. lacking the serotonin transporter Exhibit 5-HT(1A) receptor-mediated
Heinrichs, S.C., Richard, D., 1999. The role of corticotropin-releasing factor abnormalities in tests for anxiety-like behavior. Neuropsychopharmacology
and urocortin in the modulation of ingestive behavior. Neuropeptides 33, 28, 2077–2088.
350–359. Holmes, A., Lit, Q., Murphy, D.L., Gold, E., Crawley, J.N., 2003b. Abnormal
Heinrichs, S.C., Cole, B.J., Pich, E.M., Menzaghi, F., Koob, G.F., Hauger, R.L., anxiety-related behavior in serotonin transporter null mutant mice: the
1992. Endogenous corticotropin-releasing factor modulates feeding influence of genetic background. Genes Brain Behav. 2, 365–380.
induced by neuropeptide Y or a tail-pinch stressor. Peptides 13, 879–884. Holsboer, F., 2000. The corticosteroid receptor hypothesis of depression.
Heinsbroek, R., van Haaren, F., Feenstra, M., van de Poll, N., 1989. Changes in Neuropsychopharmacology 23, 477–501.
dopamine and noradrenaline activity in the frontal cortex produced by Holsboer, F., 2003. Corticotropin-releasing hormone modulators and depres-
controllable and uncontrollable shock. Behav. Pharmacol. 1, 61. sion. Curr. Opin. Invest. Drugs 4, 46–50.
Heldmann, U., Thored, P., Claasen, J.H., Arvidsson, A., Kokaia, Z., Lindvall, Honig, A., Bartlett, J.R., Bouras, N., Bridges, P.K., 1988. Amino acid levels in
O., 2005. TNF-alpha antibody infusion impairs survival of stroke-generated depression: a preliminary investigation. J. Psychiatry Res. 22, 159–164.
neuroblasts in adult rat brain. Exp. Neurol. 196, 204–208. Hopkins, S.J., Rothwell, N.J., 1995. Cytokines and the nervous system. Trends
Hellsten, J., Wennström, M., Mohapel, P., Ekdahl, C.T., Bengzon, J., Tingström, Neurosci. 18, 83–88.
A., 2002. Electroconvulsive seizures increase hippocampal neurogenesis Hori, T., Katafuchi, T., Take, S., Shimizu, N., 1998. Neuroimmunomodulatory
after chronic corticosterone treatment. Eur. J. Neurosci. 16, 283–290. actions of hypothalamic interferon-alpha. Neuroimmunomodulation 5,
Helwig, B.G., Musch, T.I., Craig, R.A., Kenney, M.J., 2007. Increased inter- 172–177.
leukin-6 receptor expression in the paraventricular nucleus of rats with heart Hotta, M., Shibasaki, T., Arai, K., Demura, H., 1999. Corticotropin-releasing
failure. Am. J. Physiol. Regul. Integr. Comp. Physiol. 292, R1165–R1173. factor receptor type 1 mediates emotional stress-induced inhibition of food
Heneka, M.T., O’Banion, M.K., 2007. Inflammatory processes in Alzheimer’s intake and behavioral changes in rats. Brain Res. 823, 221–225.
disease. J. Neuroimmunol. 184, 69–91. Houben, H., Vandenbroucke, A.T., Verheyden, A.M., Denef, C., 1993. Expres-
Henle, F., Fischer, C., Meyer, D.K., Leemhuis, J., 2006. Vasoactive intestinal sion of the genes encoding bombesin-related peptides and their receptors in
peptide and PACAP38 control N-methyl-D-aspartic acid-induced dendrite anterior pituitary tissue. Mol. Cell Endocrinol. 97, 159–164.
motility by modifying the activities of Rho GTPases and phosphatidylino- Hrdina, P.D., Demeter, E., Vu, T.B., Sotonyi, P., Palkovits, M., 1993. 5-HT
sitol 3-kinases. J. Biol. Chem. 281, 24955–24969. uptake sites and 5-HT2 receptors in brain of antidepressant-free suicide
Herman, J.P., Cullinan, W.E., 1997. Neurocircuitry of stress: central control of victims/depressives: increase in 5-HT2 sites in cortex and amygdala. Brain
hypothalamo-pituitary–adrenocortical axis. Trends Neurosci. 20, 78–84. Res. 614, 37–44.
Herman, J.P., Stinus, L., Le Moal, M., 1984. Repeated stress increases loco- Huang, J., Gao, X., Li, S., Cao, Z., 1997. Recruitment of IRAK to the interleukin
motor response to amphetamine. Psychopharmacology 84, 431–435. 1 receptor complex requires interleukin 1 receptor accessory protein. Proc.
Herman, J.P., Figueiredo, H., Mueller, N.K., Ulrich-Lai, Y., Ostrander, M.M., Natl. Acad. Sci. U.S.A. 94, 12829–12832.
Choi, D.C., Cullinan, W.E., 2003. Central mechanisms of stress integration: Huang, T.L., Lee, C.T., Liu, Y.L., 2007. Serum brain-derived neurotrophic
hierarchical circuitry controlling hypothalamo-pituitary–adrenocortical factor levels in patients with major depression: effects of antidepressants. J.
responsiveness. Front. Neuroendocrinol. 24, 151–180. Psychiatry Res., doi:10.1016/j.spsycires.2007.05.007.
58 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Huang, T.Y., Lin, C.H., 2006. Role of amygdala MAPK activation on immo- Ji, J.Z., Elyaman, W., Yip, H.K., Lee, V.W., Yick, L.W., Hugon, J., So, K.F.,
bility behavior of forced swim rats. Behav. Brain Res. 173, 104–111. 2004. CNTF promotes survival of retinal ganglion cells after induction of
Hucks, D., Lowther, S., Crompton, M.R., Katona, C.L., Horton, R.W., 1997. ocular hypertension in rats: the possible involvement of STAT3 pathway.
Corticotropin-releasing factor binding sites in cortex of depressed suicides. Eur. J. Neurosci. 19, 265–272.
Psychopharmacology 174–178. Joca, S.R., Padovan, C.M., Guimaraes, F.S., 2003. Activation of post-synaptic
Hulley, P., Hartikka, J., Abdel’Al, S., Engels, P., Buerki, H.R., Wiederhold, 5-HT(1A) receptors in the dorsal hippocampus prevents learned helpless-
K.H., Müller, T., Kelly, P., Lowe, D., Lübbert, H., Hussain, Mitra, 1995. ness development. Brain Res. 978, 177–184.
Inhibitors of type IV phosphodiesterases reduce the toxicity of MPTP in Johnson, A.B., Bake, S., Lewis, D.K., Sohrabji, F., 2006. Temporal expression
substantia nigra neurons in vivo. Eur. J. Neurosci. 7, 2431–2440. of IL-1beta protein and mRNA in the brain after systemic LPS injection is
Hussain, A.M., Mitra, A.K., 2000. Effect of aging on tryptophan hydroxylase in affected by age and estrogen. J. Neuroimmunol. 174, 82–91.
rat brain: implications on serotonin level. Drug Metab. Dispos. 28, 1038– Jow, G.M., Yang, T.T., Chen, C.L., 2006. Leptin and cholesterol levels are low
1042. in major depressive disorder, but high in schizophrenia. J. Affect. Disord.
Inder, W.J., Donald, R.A., Prickett, T.C., Frampton, C.M., Sullivan, P.F., Mulder, 90, 21–27.
R.T., Joyce, P.R., 1997. Arginine vasopressin is associated with hypercorti- Johnson, J.D., O’Connor, K.A., Deak, T., Stark, M., Watkins, L.R., Maier, S.F.,
solemia and suicide attempts in depression. Biol. Psychiatry 42, 744–747. 2002. Prior stressor exposure sensitizes LPS-induced cytokine production.
Ingram, K.M., Jones, D.A., Fass, R.J., Neidig, J.D., Song, Y.S., 1999. Social Brain Behav. Immun. 16, 461–476.
support and unsupportive social interactions: their association with depres- Johnson, J.D., O’Connor, K.A., Hansen, M.K., Watkins, L.R., Maier, S.F., 2003.
sion among people living with HIV. AIDS Care 11, 313–329. Effects of prior stress on LPS-induced cytokine and sickness responses. Am.
Inoue, W., Poole, S., Bristow, A.F., Luheshi, G.N., 2006. Leptin induces J. Physiol. Regul. Integr. Comp. Physiol. 284, R422–R432.
cyclooxygenase-2 via an interaction with interleukin-1beta in the rat brain. Johnson, J.D., O’Connor, K.A., Watkins, L.R., Maier, S.F., 2004. The role of IL-
Eur. J. Neurosci. 24, 2233–2245. 1beta in stress-induced sensitization of proinflammatory cytokine and
Intiso, D., Zarrelli, M.M., Lagioia, G., Di Rienzo, F., Checchia De Ambrosio, corticosterone responses. Neuroscience 127, 569–577.
C., Simone, P., Tonali, P., Cioffi, P., Dagger, R.P., 2004. Tumor necrosis Johnstone, T., van Reekum, C.M., Urry, H.L., Kalin, N.H., Davidson, R.J., 2007.
factor alpha serum levels and inflammatory response in acute ischemic Failure to regulate: counterproductive recruitment of top-down prefrontal-
stroke patients. Neurol. Sci. 24, 390–396. subcortical circuitry in major depression. J. Neurosci. 27, 8877–8884.
Imbe, H., Murakami, S., Okamoto, K., Iwai-Liao, Y., Senba, E., 2004. The Jones, M.D., Lucki, I., 2005. Sex differences in the regulation of serotonergic
effects of acute and chronic restraint stress on activation of ERK in the transmission and behavior in 5-HT receptor knockout mice. Neuropsycho-
rostral ventromedial medulla and locus coeruleus. Pain 112, 361–371. pharmacology 30, 1039–1047.
Iosifescu, D.V., Nierenberg, A.A., Alpert, J.E., Papakostas, G.I., Perlis, R.H., Jordan, S., Kramer, G.L., Zukas, P.K., Petty, F., 1994. Previous stress increases
Sonawalla, S., Fava, M., 2004. Comorbid medical illness and relapse of in vivo biogenic amine response to swim stress. Neurochem. Res. 19, 1521–
major depressive disorder in the continuation phase of treatment. Psychoso- 1525.
matics 45, 419–425. Judge, S.J., Young, R.L., Gartside, S.E., 2006. GABA(A) receptor modulation
Iredale, P.A., Terwilliger, R., Widnell, K.L., Nestler, E.J., Duman, R.S., 1996. of 5-HT neuronal firing in the median raphe nucleus: implications for the
Differential regulation of corticotropin-releasing factor1 receptor expres- action of anxiolytics. Eur. Neuropsychopharmacol. 16, 612–619.
sion by stress and agonist treatments in brain and cultured cells. Mol. Juengling, F.D., Ebert, D., Gut, O., Engelbrecht, M.A., Rasenack, J., Nitzsche,
Pharmacol. 1103–1110. E.U., Bauer, J., Lieb, K., 2000. Prefrontal cortical hypometabolism during
Irwin, J., Ahluwalia, P., Anisman, H., 1986. Sensitization of norepinephrine low-dose interferon alpha treatment. Psychopharmacology 152, 383–389.
activity following acute and chronic footshock. Brain Res. 379, 98–103. Kalen, P., Rosegren, E., Lindvall, O., Bjorklund, A., 1989. Hippocampal
Irwin, M., 1999. Immune correlates of depression. Adv. Exp. Med. Biol. 461, noradrenaline and serotonin release over 24 h as measured by the dialysis
1–24. technique in freely moving rats: correlation to behavioural activity state,
Itoh, S., Takashima, A., Itoh, T., Morimoto, T., 1994. Open-field behavior of rats effect of handling and tail pinch. Eur. J. Neurosci. 1, 181–188.
following intracerebroventricular administration of neuromedin B, neuro- Kalivas, P.W., Duffy, P., 1995. Selective activation of dopamine transmission in
medin C, and related amphibian peptides. Jpn. J. Physiol. 44, 271–281. the shell of the nucleus accumbens by stress. Brain Res. 675, 325–328.
Itoi, K., Helmreich, D.L., Lopez-Figueroa, M.O., Watson, S.J., 1999. Differ- Kalivas, P.W., Nakamura, M., 1999. Neural systems for behavioral activation
ential regulation of corticotropin-releasing hormone and vasopressin gene and reward. Curr. Opin. Neurobiol. 9, 223–227.
transcription in the hypothalamus by norepinephrine. J. Neurosci. 19, 5464– Kalivas, P.W., Stewart, J., 1991. Dopamine transmission in the initiation and
5472. expression of drug- and stress-induced sensitization of motor activity. Brain
Jacobs, B.L., 2002. Adult brain neurogenesis and depression. Brain Behav. Res. Rev. 16, 223–244.
Immun. 16, 602–609. Kalueff, A.V., Gallagher, P.S., Murphy, D.L., 2006. Are serotonin transporter
Jander, S., Schroeter, M., Stoll, G., 2002. Interleukin-18 expression after focal knockout mice ’depressed’?: hypoactivity but no anhedonia. Neuroreport
ischemia of the rat brain: association with the late-stage inflammatory 17, 1347–1351.
response. J. Cereb. Blood Flow Metab. 22, 62–70. Kamm, K., Vanderkolk, W., Lawrence, C., Jonker, M., Davis, A.T., 2006. The
Jedema, H.P., Sved, A.F., Zigmond, M.J., Finlay, J.M., 1999. Sensitization of effect of traumatic brain injury upon the concentration and expression of
norepinephrine release in medial prefrontal cortex: effect of different interleukin-1beta and interleukin-10 in the rat. J. Trauma 60, 152–157.
chronic stress protocols. Brain Res. 830, 211–217. Kaneko, N., Kudo, K., Mabuchi, T., Takemoto, K., Fujimaki, K., Wati, H.,
Jenner, P., 2001. Parkinson’s disease, pesticides and mitochondrial dysfunction. Iguchi, H., Tezuka, H., Kanba, S., 2006. Suppression of cell proliferation by
Trends Neurosci. 24, 245–247. interferon-alpha through interleukin-1 production in adult rat dentate gyrus.
Jensen, M.B., Finsen, B., Zimmer, J., 1997. Morphological and immunophe- Neuropsychopharmacology 31, 2619–2626.
notypic microglial changes in the denervated fascia dentata of adult rats: Kaneyuki, H., Yokoo, H., Tsuda, A., Yoshida, M., Mizuki, Y., Yamada, M.,
correlation with blood–brain barrier damage and astroglial reactions. Exp. Tanaka, M., 1991. Psychological stress increases dopamine turnover selec-
Neurol. 143, 103–116. tively in mesoprefrontal dopamine neurons of rats: reversal by diazepam.
Jeschke, M.G., Einspanier, R., Klein, D., Jauch, K.W., 2002. Insulin attenu- Brain Res. 557, 154–161.
ates the systemic inflammatory response to thermal trauma. Mol. Med. 8, Karten, Y.J., Nair, S.M., van Essen, L., Sibug, R., Joëls, M., 1999. Long-term
443–450. exposure to high corticosterone levels attenuates serotonin responses in rat
Jha, S., Rajendran, R., Davda, J., Vaidya, V.A., 2006. Selective serotonin hippocampal CA1 neurons. Proc. Natl. Acad. Sci. U.S.A. 96, 13456–13461.
depletion does not regulate hippocampal neurogenesis in the adult rat brain: Kassed, C.A., Herkenham, M., 2004. NF-kappaB p50-deficient mice show
differential effects of p-chlorophenylalanine and 5,7-dihydroxytryptamine. reduced anxiety-like behaviors in tests of exploratory drive and anxiety.
Brain Res. 1075, 48–59. Behav. Brain Res. 154, 577–584.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 59

Kato, M., Iwata, H., Okamoto, M., Ishii, T., Narita, H., 2000. Focal cerebral Kinouchi, K., Brown, G., Pasternak, G., Donner, D.B., 1991. Identification and
ischemia-induced escape deficit in rats is ameliorated by a reversible characterization of receptors for tumor necrosis factor-a in the brain.
inhibitor of monoamine oxidase-a: implications for a novel animal model Biochem. Biophys. Res. Commun. 181, 1532–1538.
of post-stroke depression. Biol. Pharm. Bull. 23, 406–410. Kirby, L.G., Rice, K.C., Valentino, R.J., 2000. Effects of corticotropin-releasing
Kauffman, R.P., Castracane, V.D., White, D.L., Baldock, S.D., Owens, R., 2005. factor on neuronal activity in the serotonergic dorsal raphe nucleus.
Impact of the selective serotonin reuptake inhibitor citalopram on insulin Neuropsychopharmacology 22, 148–162.
sensitivity, leptin and basal cortisol secretion in depressed and non- Kishimoto, T., Radulovic, J., Radulovic, M., Lin, C.R., Schrick, C., Hooshman,
depressed euglycemic women of reproductive age. Gynecol. Endocrinol. F., Hermanson, O., Rosenfeld, M.G., Spiess, J., 2000. Deletion of crhr2
21, 129–137. reveals an anxiolytic role for corticotropin-releasing hormone receptor-2.
Kaur, C., Sivakumar, V., Dheen, S.T., Ling, E.A., 2006. Insulin-like growth Nat. Genet. 24, 415–419.
factor I and II expression and modulation in amoeboid microglial cells by Kita, T., Liu, L., Tanaka, N., Kinoshita, Y., 1997. The expression of tumor
lipopolysaccharide and retinoic acid. Neuroscience 138, 1233–1244. necrosis factor-alpha in the rat brain after fluid percussive injury. Int. J.
Kaura, V., Ingram, C.D., Gartside, S.E., Young, A.H., Judge, S.J., 2007. The Legal Med. 110, 305–311.
progesterone metabolite allopregnanolone potentiates GABA(A) receptor- Kitagami, T., Yamada, K., Miura, H., Hashimoto, R., Nabeshima, T., Ohta, T.,
mediated inhibition of 5-HT neuronal activity. Eur. Neuropsychopharmacol. 2003. Mechanism of systemically injected interferon-alpha impeding
17, 108–115. monoamine biosynthesis in rats: role of nitric oxide as a signal crossing
Keck, M.E., Muller, M.B., 2005. Mutagenesis and knockout models: hypotha- the blood–brain barrier. Brain Res. 978, 104–114.
lamic–pituitary–adrenocortical system. Handb. Exp. Pharmacol. 169, Kling, M.A., Roy, A., Doran, A.R., Calabrese, J.R., Rubinow, D.R., Whitfield
113–141. Jr., H.J., May, C., Post, R.M., Chrousos, G.P., Gold, P.W., 1991. Cere-
Keck, M.E., Welt, T., Wigger, A., Renner, U., Engelmann, M., Holsboer, F., brospinal fluid immunoreactive corticotropin-releasing hormone and adre-
Landgraf, R., 2001. The anxiolytic effect of the CRH1 receptor antagonist nocorticotropin secretion in Cushing’s disease and major depression:
R121919 depends on innate emotionality in rats. Eur. J. Neurosci. 13, 373– potential clinical implications. J. Clin. Endocrinol. Metab. 72, 260–271.
380. Kling, M.A., Geracioti, T.D., Licinio, J., Michelson, D., Oldfield, E.H., Gold,
Keeney, A., Jessop, D.S., Harbuz, M.S., Marsden, C.A., Hog, S., Blackburn- P.W., 1994. Effects of electroconvulsive therapy on the CRH-ACTH-
Munro, R.E., 2006. Differential effects of acute and chronic social defeat cortisol system in melancholic depression: preliminary findings. Psycho-
stress on hypothalamic–pituitary–adrenal axis function and hippocampal pharmacol. Bull. 30, 489–494.
serotonin release in mice. J. Neuroendocrinol. 18, 330–338. Knoblach, S.M., Fan, L., Faden, A.I., 1999. Early neuronal expression of tumor
Kelly, O., Matheson, K., Ravindran, A., Merali, Z., Anisman, H., 2007. necrosis factor-alpha after experimental brain injury contributes to neuro-
Ruminative coping among patients with dysthymia before and after phar- logical impairment. J. Neuroimmunol. 95, 115–125.
macotherapy. Depress. Anxiety 24, 233–243. Kobayashi, T., Ahlenius, H., Thored, P., Kobayashi, R., Kokaia, Z., Lindvall, O.,
Kempermann, G., Gast, D., Kronenberg, G., Yamaguchi, M., Gage, F.H., 2003. 2006. Intracerebral infusion of glial cell line-derived neurotrophic factor
Early determination and long-term persistence of adult-generated new promotes striatal neurogenesis after stroke in adult rats. Stroke 37, 2361–
neurons in the hippocampus of mice. Development 130, 391–399. 2367.
Kendler, K.S., Kuhn, J.W., Vittum, J., Prescott, C.A., Riley, B., 2005. The Konishi, N., Otaka, M., Odashima, M., Jin, M., Wada, I., Komatsu, K., Sato, T.,
interaction of stressful life events and a serotonin transporter polymorphism Kato, S., Matsuhashi, T., Watanabe, S., 2006. Systemic stress increases
in the prediction of episodes of major depression: a replication. Arch. Gen. serum leptin level. J. Gastroenterol. Hepatol. 21, 1099–1102.
Psychiatry 62, 529–635. Konsman, J.P., Dantzer, R., 2001. How the immune and nervous systems
Kent, P., Anisman, H., Merali, Z., 1998. Are bombesin-like peptides involved in interact during disease-associated anorexia. Nutrition 17, 664–668.
the mediation of the stress response. Life Sci. 62, 103–114. Konsman, J.P., Luheshi, G.N., Bluthe, R.M., Dantzer, R., 2000. The vagus nerve
Kent, P., Anisman, H., Merali, Z., 2001a. Central bombesin activates the mediates behavioural depression, but not fever, in response to peripheral
hypothalamic–pituitary–adrenal axis: effects on regional levels and release immune signals; a functional anatomical analysis. Eur. J. Neurosci. 12,
of corticotropin-releasing hormone and arginine vasopressin. Neuroendo- 4434–4446.
crinology 73, 203–214. Konsman, J.P., Parnet, P., Dantzer, R., 2002. Cytokine-induced sickness beha-
Kent, P., Bedard, T., Khan, S., Anisman, H., Merali, Z., 2001b. Bombesin- viour: mechanisms and implications. Trends Neurosci. 25, 154–159.
induced HPA and sympathetic activation requires CRH receptors. Peptides Kooijman, R., Coppens, A., 2004. Insulin-like growth factor-I stimulates IL-10
22, 57–65. production in human T cells. J. Leukoc. Biol. 76, 862–867.
Kent, S., Bluthe, R.-M., Kelley, K.W., Dantzer, R., 1992. Sickness behavior as a Korpi, E.R., Kleinman, J.E., Wyatt, R.J., 1988. GABA concentrations in
new target for drug development. Trends Pharmacol. Sci. 13, 24–28. forebrain areas of suicide victims. Biol. Psychiatry 23, 109–114.
Kent, S., Bret-Dibat, J.L., Kelley, K.W., Dantzer, R., 1996. Mechanisms of Koutsilieri, E., Sopper, S., Scheller, C., ter Meulen, V., Riederer, P., 2002a.
sickness-induced decreases in food-motivated behavior. Neurosci. Biobe- Involvement of dopamine in the progression of AIDS dementia complex. J.
hav. Rev. 20, 171–175. Neural Transm. 109, 399–410.
Kern, W., Sieghart, W., 1994. Polyclonal antibodies directed against an epitope Koutsilieri, E., Sopper, S., Scheller, C., ter Meulen, V., Riederer, P., 2002b.
specific for the alpha 4-subunit of GABAA receptors identify a 67-kDa Parkinsonism in HIV dementia. J. Neural Transm. 109, 767–775.
protein in rat brain membranes. J. Neurochem. 62, 764–769. Kovacs, K.J., Sawchenko, P.E., 1996. Regulation of stress-induced transcrip-
Kessler, R.C., 1997. The effects of stressful life events on depression. Ann. Rev. tional changes in the hypothalamic neurosecretory neurons. J. Mol. Neu-
Psychol. 48, 191–214. rosci. 7, 125–133.
Khan, Z.U., Gutierrez, A., De Blas, A.L., 1996. The alpha 1 and alpha 6 subunits Kozlovsky, N., Matar, M.A., Kaplan, Z., Kotler, M., Zohar, J., Cohen, H., 2007.
can coexist in the same cerebellar GABAA receptor maintaining their Long-term down-regulation of BDNF mRNA in rat hippocampal CA1
individual benzodiazepine-binding specificities. J. Neurochem. 66, 685–691. subregion correlates with PTSD-like behavioural stress response. Int. J.
Kim, Y.S., Joh, T.H., 2006. Microglia, major player in the brain inflammation: Neuropsychopharmacol. 10, 741–758.
their roles in the pathogenesis of Parkinson’s disease. Exp. Mol. Med. 38, Kozorovitskiy, Y., Gould, E., Brustolim, D., Ribeiro-dos-Santos, R., Kast, R.E.,
333–347. Altschuler, E.L., Soares, M.B., 2004. Dominance hierarchy influences adult
Kimball, E.S., Persico, F.J., Vaught, J.L., 1988. Substance P, neurokinin A, and neurogenesis in the dentate gyrus. J. Neurosci. 24, 6755–6759.
neurokinin. J. Immunol. 141, 3564–3569 (B induce generation of IL-1-like Kraus, M.R., Schafer, A., Al-Taie, O., Scheurlen, M., 2005a. Prophylactic SSRI
activity in P388D1 cells. Possible relevance to arthritic disease). during interferon alpha re-therapy in patients with chronic hepatitis C and a
Kimura, M., Tateno, A., Robinson, R.G., 2003. Treatment of poststroke history of interferon-induced depression. J. Viral Hepat. 12, 96–100.
generalized anxiety disorder comorbid with poststroke depression: merged Kraus, M.R., Schafer, A., Csef, H., Scheurlen, M., 2005b. Psychiatric side
analysis of nortriptyline trials. Am. J. Geriatr. Psychiatry 11, 320–327. effects of pegylated interferon alfa-2b as compared to conventional
60 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

interferon alfa-2b in patients with chronic hepatitis C. World J. Gastro- Lazarus, R.S., Folkman, S., 1984. Stress, Appraisal, and Coping. Springer, New
enterol. 11, 1769–1774. York.
Kraus, M.R., Schafer, A., Faller, H., Csef, H., Scheurlen, M., 2005c. Paroxetine Leake, A., Perry, E.K., Perry, R.H., Fairbairn, A.F., Ferrier, I.N., 1990. Cortical
for the treatment of interferon-alpha-induced depression in chronic hepatitis concentrations of corticotropin-releasing hormone and its receptor in
C. Aliment. Pharmacol. Ther. 16, 1091–1099. Alzheimer type dementia and major depression. Biol. Psychiatry 28,
Kraus, M.R., Schafer, A., Wissmann, S., Reimer, P., Scheurlen, M., 2005d. 603–608.
Neurocognitive changes in patients with hepatitis C receiving interferon Leake, A., Fairbairn, A.F., McKeith, I.G., Ferrier, I.N., 1991. Studies on the
alfa-2b and ribavirin. Clin. Pharmacol. Ther. 77, 90–100. serotonin uptake binding site in major depressive disorder and control post-
Kraus, T., Haack, M., Schuld, A., Hinze-Selch, D., Pollmacher, T., 2001. Low mortem brain: neurochemical and clinical correlates. Psychiatry Res. 39,
leptin levels but normal body mass indices in patients with depression or 155–165.
schizophrenia. Neuroendocrinology 73, 243–247. Ledeboer, A., Binnekade, R., Brave, J.J., Bol, J.G., Tilders, F.J., Van Dam,
Kretz, A., Happold, C.J., Marticke, J.K., Isenmann, S., 2005. Erythropoietin A.M., 2002. Site-specific modulation of LPS-induced fever and interleukin-
promotes regeneration of adult CNS neurons via Jak2/Stat3 and PI3K/AKT 1 beta expression in rats by interleukin-10. Am. J. Physiol. Regul. Integr.
pathway activation. Mol. Cell Neurosci. 29, 569–579. Comp. Physiol. 282, R1762–R1772.
Kronfol, Z., Remick, D.G., 2000. Cytokines and the brain: implications for LeDoux, J.E., 2000. Emotion circuits in the brain. Ann. Rev. Neurosci. 23,
clinical psychiatry. Am. J. Psychiatry 157, 683–694. 155–184.
Krystal, J.H., Sanacora, G., Blumberg, H., Anand, A., Charney, D.S., Marek, G., Lee, B.C., Ahn, S.Y., Doo, H.K., Yim, S.V., Lee, H.J., Jin, S.Y., Hong, S.J.,
Epperson, C.N., Goddard, A., Mason, G.F., 2002. Glutamate and GABA Lee, S.H., Kim, S.D., Seo, J.C., Leem, K.H., Chung, J.H., 2004. Suscept-
systems as targets for novel antidepressant and mood-stabilizing treatments. ibility for ischemic stroke in Korean population is associated with
Mol. Psychiatry 7 (Suppl. 1), S71–S80. polymorphisms of the interleukin-1 receptor antagonist and tumor necro-
Kuipers, S.D., Bramham, C.R., 2006. Brain-derived neurotrophic factor sis factor-alpha genes, but not the interleukin-1beta gene. Neurosci. Lett.
mechanisms and function in adult synaptic plasticity: new insights and 357, 33–36.
implications for therapy. Curr. Opin. Drug Discov. Dev. 9, 580–586. Lee, J.H., Park, E.J., Kim, O.S., Kim, H.Y., Joe, E.H., Jou, I., 2005. Double-
Kulkosky, P.J., Gibbs, J., Smith, G.P., 1982. Feeding suppression and groom- stranded RNA-activated protein kinase is required for the LPS-induced
ing repeatedly elicited by intraventricular bombesin. Brain Res. 242, activation of STAT1 inflammatory signaling in rat brain glial cells. Glia 50,
194–196. 66–79.
Kulmatycki, K.M., Jamali, F., 2006. Drug disease interactions: role of inflam- Lee, K.J., Kim, S.J., Kim, S.W., Choi, S.H., Shin, Y.C., Park, S.H., Moon, B.H.,
matory mediators in depression and variability in antidepressant drug Cho, E., Lee, M.S., Choi, S.H., Chun, B.G., Shin, K.H., 2006a. Chronic mild
response. J. Pharm. Sci. 9, 292–306. stress decreases survival, but not proliferation, of new-born cells in adult rat
Kuroda, Y., Watanabe, Y., Albeck, D.S., Hastings, N.B., McEwen, B.S., 1994. hippocampus. Exp. Mol. Med. 38, 44–54.
Effects of adrenalectomy and type I or type II glucocorticoid receptor Lee, S.H., Kim, Y.J., Lee, K.M., Ryu, S., Yoon, B.W., 2007. Ischemic
activation on 5-HT1A and 5-HT2 receptor binding and 5-HT transporter preconditioning enhances neurogenesis in the subventricular zone. Neu-
mRNA expression in rat brain. Brain Res. 648, 157–161. roscience 146, 1020–1031.
Kusnecov, A.W., Liang, R., Shurin, G., 1999. T-lymphocyte activation increases Lee, Y., Davis, M., 1997. Role of the hippocampus, bed nucleus of the stria
hypothalamic and amygdaloid expression of CRH mRNA and emotional terminalis, and the amygdala in the excitatory effect of corticotropin-
reactivity to novelty. J. Neurosci. 19, 4533–4543. releasing hormone on the acoustic startle reflex. J. Neurosci. 17, 6434–6446.
Kwidzinski, E., Bechmann, I., 2007. IDO expression in the brain: a double- Lee, Y.J., Choi, B., Lee, E.H., Choi, K.S., Sohn, S., 2006b. Immobilization
edged sword. J. Mol. Med. 85, 1351–1359. stress induces cell death through production of reactive oxygen species in
LaBar, K.S., Gatenby, J.C., Gore, J.C., LeDoux, J.E., Phelps, E.A., 1998. the mouse cerebral cortex. Neurosci. Lett. 392, 27–31.
Human amygdala activation during conditioned fear acquisition and extinc- Leentjens, A.F., Van den Akker, M., Metsemakers, J.F., Lousberg, R., Verhey,
tion: a mixed-trial fMRI study. Neuron 20, 937–945. F.R., 2003. Higher incidence of depression preceding the onset of Parkin-
Lacosta, S., Merali, Z., Anisman, H., 1998. Influence of interleukin-1 on son’s disease: a register study. Mov. Disord. 18, 414–418.
exploratory behaviors, plasma ACTH and cortisol, and central biogenic Lemonde, S., Turecki, G., Bakish, D., Du, L., Hrdina, P.D., Bown, C.D.,
amines in mice. Psychopharmacology 137, 351–361. Sequeira, A., Kushwaha, N., Morris, S.J., Basak, A., Ou, X.M., Albert,
Ladenheim, E.E., Jensen, R.T., Mantey, S.A., Moran, T.H., 1992. Distinct P.R., 2003. Impaired repression at a 5-hydroxytryptamine 1A receptor gene
distributions of two bombesin receptor subtypes in the rat central nervous polymorphism associated with major depression and suicide. J. Neurosci.
system. Brain Res. 593, 168–178. 23, 8788–8799.
Laflamme, N., Rivest, S., 1999. Effects of systemic immunogenic insults and Lesch, K.P., Zeng, Y., Reif, A., Gutknecht, L., 2003. Anxiety-related traits in
circulating proinflammatory cytokines on the transcription of the inhibitory mice with modified genes of the serotonergic pathway. Eur. J. Pharmacol.
factor kappaB alpha within specific cellular populations of the rat brain. J. 480, 185–204.
Neurochem. 73, 309–321. Lespérance, F., Frasure-Smith, N., Théroux, P., Irwin, M., 2004. The association
Landgraf, R., 2006. The involvement of the vasopressin system in stress-related between major depression and levels of soluble intercellular adhesion
disorders. CNS Neurol. Disord. Drug Targets 5, 167–179. molecule 1, interleukin-6, and C-reactive protein in patients with recent
Lanfumey, L., Mannotry La Cour, C., Froger, N., Hamon, M., 2000. 5-HT-HPA acute coronary syndromes. Am. J. Psychiatry 161, 271–277.
interactions in two models of transgenic mice relevant to major depression. Li, J., Takeda, H., Tsuji, M., Liu, L., Matsumiya, T., 1998. Antagonism of
Neurochem. Res. 25, 1199–1206. central CRF systems mediates stress-induced changes in noradrenaline and
Lapiz, M.D., Bondi, C.O., Morilak, D.A., 2007. Chronic treatment with serotonin turnover in rat brain regions. Methods Find Exp. Clin. Pharmacol.
desipramine improves cognitive performance of rats in an attentional 20, 409–417.
set-shifting test. Neuropsychopharmacology 32, 1000–1010. Li, R., Yang, L., Lindholm, K., Konishi, Y., Yue, X., Hampel, H., Zhang, D.,
Larson, S.J., Dunn, A.J., 2001. Behavioral effects of cytokines. Brain Behav. Shen, Y., 2004a. Tumor necrosis factor death receptor signaling cascade is
Immun. 15, 371–387. required for amyloid-beta protein-induced neuron death. J. Neurosci. 24,
Larson, S.J., Romanoff, R.L., Dunn, A.J., Glowa, J.R., 2002. Effects of 1760–1771.
interleukin-1beta on food-maintained behavior in the mouse. Brain Behav. Li, W.G., Gavrila, D., Liu, X., Wang, L., Gunnlaugsson, S., Stoll, L.L.,
Immun. 16, 398–410. McCormick, M.L., Sigmund, C.D., Tang, C., Weintraub, N.L., 2004b.
Laveille, S., Tassin, J., Thierry, A., Blanc, G., Herve, D., Barthelemy, C., Ghrelin inhibits proinflammatory responses and nuclear factor-kappaB
Glowinski, J., 1978. Blockade by benzodiazepines of the selective high activation in human endothelial cells. Circulation 109, 2221–2226.
increase in dopamine turnover induced by stress in mesocortical dopami- Li, Z.G., Zhang, W., Sima, A.A., 2007. Alzheimer-like changes in rat models of
nergic neurons of the rat. Brain Res. 168, 585–594. spontaneous diabetes. Diabetes 56, 1817–1824.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 61

Liang, X., Wang, Q., Hand, T., Wu, L., Breyer, R.M., Montine, T.J., Andreasson, brain samples from depressed suicides and controls. J. Affect. Disord. 42,
K., 2005. Deletion of the prostaglandin E2 EP2 receptor reduces oxidative 199–207.
damage and amyloid burden in a model of Alzheimer’s disease. J. Neurosci. Lu, X.Y., Kim, C.S., Frazer, A., Zhang, W., 2006. Leptin: a potential novel
25, 10180–10187. antidepressant. Proc. Natl. Acad. Sci. 103, 1593–1598.
Lieb, K., Engelbrecht, M.A., Gut, O., Fiebich, B.L., Bauer, J., Janssen, G., Lu, Z.W., Hayley, S., Ravindran, A.V., Merali, Z., Anisman, H., 1999. Influence
Schaefer, M., 2006. Cognitive impairment in patients with chronic hepatitis of psychosocial, psychogenic and neurogenic stressors on several aspects of
treated with interferon alpha (IFNalpha): results from a prospective study. immune functioning in mice. Stress 3, 55–70.
Eur. Psychiatry 21, 204–210. Lucas, S.M., Rothwell, N.J., Gibson, R.M., 2006. The role of inflammation in
Lightman, S.L., 1994. How does the hypothalamus respond to stress. Sem. CNS injury and disease. Br. J. Pharmacol. 147 (Suppl. 1), S232–S240.
Neurosci. 6, 215–219. Lucas, P.C., McAllister-Lucas, L.M., Nunez, G., 2004. NF-kappaB signaling in
Lieberman, A., 2006. Depression in Parkinson’s disease—a review. Acta lymphocytes: a new cast of characters. J. Cell Sci. 117, 31–39.
Neurol. Scand. 113, 1–8. Lucassen, P.J., Vollmann-Honsdorf, G.K., Gleisberg, M., Czéh, B., De Kloet,
Lightman, S.L., Windle, R.J., Julian, M.D., Harbuz, M.S., Shanks, N., Wood, E.R., Fuchs, E., 2001. Chronic psychosocial stress differentially affects
S.A., Kershaw, Y.M., Ingram, C.D., 2000. Significance of pulsatility in the apoptosis in hippocampal subregions and cortex of the adult tree shrew. Eur.
HPA axis. Novartis Found. Symp. 227, 244–257. J. Neurosci. 14, 161–166.
Linthorst, A.C.E., Flachskamm, C., Muller-Preuss, P., Holsboer, F., Reul, Luheshi, G.N., Gardner, J.D., Rushforth, D.A., Loudon, A.S., Rothwell, N.J.,
J.M.H.M., 1995. Effect of bacterial endotoxin and interleukin-1b on hip- 1999. Leptin actions on food intake and body temperature are mediated by
pocampal serotonergic neurotransmision, behavioral activity, and free IL-1. Proc. Natl. Acad. Sci. 96, 7047–7052.
corticosterone levels: an in vivo microdialysis study. J. Neurosci. 15, Luo, Y., Henricksen, L.A., Giuliano, R.E., Prifti, L., Callahan, L.M., Federoff,
2920–2934. H.J., 2007. VIP is a transcriptional target of Nurr1 in dopaminergic cells.
Linthorst, A.C.E., Flachskamm, C., Hopkins, S.J., Hoadley, M.E., Labeur, M.S., Exp. Neurol. 203, 221–232.
Holsboer, F., Reul, J.M.H.M., 1997. Long-term intracerebroventricular Lupien, S.J., Evans, A., Lord, C., Miles, J., Pruessner, M., Pike, B., Pruessner,
infusion of corticotropin-releasing hormone alters neuroendocrine, neuro- J.C., 2007. Hippocampal volume is as variable in young as in older adults:
chemical, autonomic, behavioral, and cytokine responses to a systemic implications for the notion of hippocampal atrophy in humans. Neuroimage
inflammatory challenge. J. Neurosci. 17, 4448–4460. 34, 479–485.
Linthorst, A.C., Penalva, R.G., Flachskamm, C., Holsboer, F., Reul, J.M., 2002. Lyte, M., Varcoe, J.J., Bailey, M.T., 1998. Anxiogenic effect of subclinical
Forced swim stress activates rat hippocampal serotonergic neurotransmis- bacterial infection in mice in the absence of overt immune activation.
sion involving a corticotropin-releasing hormone receptor-dependent Physiol. Behav. 65, 63–68.
mechanism. Eur. J. Neurosci. 16, 2441–2452. Ma, W., Quirion, R., 2006. Increased calcitonin gene-related peptide in
Lira, A., Zhou, M., Castanon, N., Ansorge, M.S., Gordon, J.A., Francis, J.H., neuroma and invading macrophages is involved in the up-regulation of
Bradley-Moore, M., Lira, J., Underwood, M.D., Arango, V., Kung, H.F., interleukin-6 and thermal hyperalgesia in a rat model of mononeuropathy. J.
Hofer, M.A., Hen, R., Gingrich, J.A., 2003. Altered depression-related Neurochem. 98, 180–192.
behaviors and functional changes in the dorsal raphe nucleus of serotonin Ma, X.M., Lightman, S.L., Aguilera, G., 1999. Vasopressin and corticotropin-
transporter-deficient mice. Biol. Psychiatry 54, 960–971. releasing hormone gene responses to novel stress in rats adapted to repeated
Liu, B., Gao, H.M., Hong, J.S., 2003. Parkinson’s disease and exposure to restraint. Endocrinology 140, 3623–3632.
infectious agents and pesticides and the occurrence of brain injuries: role of MacMaster, F.P., Kusumakar, V., 2004. Hippocampal volume in early onset
neuroinflammation. Environ. Health Perspect. 111, 1065–1073. depression. BMC Med. 2 (Jan 29), 2.
Liu, L., Li, Y., Van Eldik, L.J., Griffin, W.S., Barger, S.W., 2005. S100B- MacQueen, G.M., Campbell, S., McEwen, B.S., Macdonald, K., Amano, S.,
induced microglial and neuronal IL-1 expression is mediated by cell type- Joffe, R.T., Nahmias, C., Young, L.T., 2003. Course of illness, hippocampal
specific transcription factors. J. Neurochem. 92, 546–553. function, and hippocampal volume in major depression. Proc. Natl. Acad.
Loddick, S.A., Wong, M.L., Bongiorno, P.B., Gold, P.W., Licinio, J., Rothwell, Sci. 100, 1387–1392.
N.J., 1997. Endogenous interleukin-1 receptor antagonist is neuroprotec- Maddock, C., Baita, A., Orru, M.G., Sitzia, R., Costa, A., Muntoni, E., Farci,
tive. Biochem. Biophys. Res. Commun. 234, 211–215. M.G., Carpiniello, B., Pariante, C.M., 2004. Psychopharmacological treat-
Loftis, J.M., Hauser, P., 2004. The phenomenology and treatment of interferon- ment of depression, anxiety, irritability and insomnia in patients receiving
induced depression. J. Affect. Disord. 82, 175–190. interferon-alpha: a prospective case series and a discussion of biological
Long, N.C., Morimoto, A., Nakamori, T., Murakami, N., 1992. Systemic mechanisms. J. Psychopharmacol. 18, 41–46.
injection of TNF-alpha attenuates fever due to IL-1 beta and LPS in rats. Maes, M., 1995. Evidence for an immune response in major depression: a review
Am. J. Physiol. 263, R987–R991. and hypothesis. Prog. Neuropsychopharmacol. Biol. Psychiatry 19, 11–38.
Lopez, J.F., Vazquez, D.M., Chalmers, D.T., Watson, S.J., 1997. Regulation of Maes, M., 1999. Major depression and activation of the inflammatory response
5-HT receptors and the hypothalamic–pituitary–adrenal axis. Implications system. Adv. Exp. Med. Biol. 461, 25–45.
for the neurobiology of suicide. Ann. NY Acad. Sci. 836, 106–134. Maes, M., Bonaccorso, S., 2004. Lower activities of serum peptidases predict
Lopez, J.F., Chalmers, D.T., Little, K.Y., Watson, S.J., 1998. Regulation of higher depressive and anxiety levels following interferon-alpha-based
serotonin1A, glucocorticoid, and mineralocorticoid receptor in rat and immunotherapy in patients with hepatitis C. Acta Psychiatr. Scand. 109,
human hippocampus: implications for the neurobiology of depression. Biol. 126–131.
Psychiatry 43, 547–573. Maes, M., Meltzer, H.Y., 1995. The serotonin hypothesis of major depression.
Lopez-Lopez, C., Dietrich, M.O., Metzger, F., Loetscher, H., Torres-Aleman, I., In: Bloom, F.E. (Ed.), Psychopharmacology: The Third Generation of
2007. Disturbed cross talk between insulin-like growth factor I and AMP- Progress. Raven Press, New York, pp. 944–993.
activated protein kinase as a possible cause of vascular dysfunction in the Maes, M., Capuron, L., Ravaud, A., Gualde, N., Bosmans, E., Egyed, B.,
amyloid precursor protein/presenilin 2 mouse model of Alzheimer’s dis- Dantzer, R., Neveu, P.J., 2001. Lowered serum dipeptidyl peptidase IV
ease. J. Neurosci. 27, 824–831. activity is associated with depressive symptoms and cytokine production in
Lotz, M., Vaughan, J.H., Carson, D.A., 1988. Effect of neuropeptides on cancer patients receiving interleukin-2-based immunotherapy. Neuropsy-
production of inflammatory cytokines by human monocytes. Science chopharmacology 24, 130–140.
241, 1218–1221. Maes, M., Kenis, G., Bosmans, E., 2002. The negative immunoregulatory
Lowther, S., De Paermentier, F., Crompton, M.R., Katona, C.L., Horton, R.W., effects of serotonin (5-HT) moduline, an endogenous 5-HT1B receptor
1994. Brain 5-HT2 receptors in suicide victims: violence of death, depres- antagonist with anti-anxiety properties. Cytokine 19, 308–311.
sion and effects of antidepressant treatment. Brain Res. 642, 281–289. Magaki, S., Mueller, C., Dickson, C., Kirsch, W., 2007. Increased production of
Lowther, S., De Paermentier, F., Cheetham, S.C., Crompton, M.R., Katona, inflammatory cytokines in mild cognitive impairment. Exp. Gerontol. 42,
C.L., Horton, R.W., 1997. 5-HT1A receptor binding sites in post-mortem 233–240.
62 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Magarinos, A.M., McEwen, B.S., Flugge, G., Fuchs, E., 1996. Chronic psy- Mann, J.J., Stanley, M., McBride, P.A., McEwen, B.S., 1986. Increased
chosocial stress causes apical dendritic atrophy of hippocampal CA3 serotonin2 and beta-adrenergic receptor binding in the frontal cortices of
pyramidal neurons in subordinate tree shrews. J. Neurosci. 16, 3534–3540. suicide victims. Arch. Gen. Psychiatry 43, 954–959.
Maguire, J., Mody, I., 2007. Neurosteroid synthesis-mediated regulation of Mann, J.J., Huang, Y.Y., Underwood, M.D., Kassir, S.A., Oppenheim, S., Kelly,
GABA(A) receptors: relevance to the ovarian cycle and stress. J. Neurosci. T.M., Dwork, A.J., Arango, V., 2000. A serotonin transporter gene promoter
27, 2155–2162. polymorphism (5-HTTLPR) and prefrontal cortical binding in major
Maher, F.O., Martin, D.S., Lynch, M.A., 2004. Increased IL-1beta in cortex of depression and suicide. Arch. Gen. Psychiatry 57, 729–738.
aged rats is accompanied by downregulation of ERK and PI-3 kinase. Manni, L., Di Fausto, V., Chaldakov, G.N., Aloe, L., 2007. Brain leptin and
Neurobiol. Aging 25, 795–806. nerve growth factor are differently affected by stress in male and female
Maher, F.O., Clarke, R.M., Kelly, A., Nally, R.E., Lynch, M.A., 2006. Inter- mice: Possible neuroendocrine and cardio-metabolic implications. Neu-
action between interferon gamma and insulin-like growth factor-1 in rosci. Lett. 426, 39–44.
hippocampus impacts on the ability of rats to sustain long-term potentiation. Mansbach, R.S., Brooks, E.N., Chen, Y.L., 1997. Antidepressant-like effects of
J. Neurochem. 96, 1560–1571. CP-154,526, a selective CRF1 receptor antagonist. Eur. J. Pharmacol. 323,
Maher, S.G., Romero-Weaver, A.L., Scarzello, A.J., Gamero, A.M., 2007. 21–26.
Interferon: cellular executioner or white knight? Curr. Med. Chem. 14, Mantovani, A., Muzio, M., Ghezzi, P., Colotta, F., Introna, M., 1996. Negative
1279–1289. regulators of the interleukin-1 system: receptor antagonists and a decoy
Mahley, R.W., Weisgraber, K.H., Huang, Y., 2006. Apolipoprotein E4: a receptor. Int. J. Clin. Lab. Res. 26, 7–14.
causative factor and therapeutic target in neuropathology, including Alz- Mapou, R.L., Law, W.A., Wagner, K., Malone, J.L., Skillman, D.R., 1996.
heimer’s disease. Proc. Natl. Acad. Sci. U.S.A. 103, 5644–5651. Neuropsychological effects of interferon Alfa-n3 treatment in asympto-
Maier, S.F., Watkins, L.R., 1998. Cytokines for psychologists: Implications of matic human immunodeficiency virus-1-infected individuals. J. Neuropsy-
bidirectional immune-to-brain communication for understanding behavior, chiat. Clin. Neurosci. 8, 74–81.
mood, and cognition. Psychol. Rev. 105, 83–107. Marowsky, A., Yanagawa, Y., Obata, K., Vogt, K.E., 2005. A specialized
Maier, S.F., Watkins, L.R., 2005. Stressor controllability and learned help- subclass of interneurons mediates dopaminergic facilitation of amygdala
lessness: the roles of the dorsal raphe nucleus, serotonin, and corticotropin- function. Neuron 48, 1025–1037.
releasing factor. Neurosci. Biobehav. Rev. 29, 829–841. Martin, J.B., 2002. The integration of neurology, psychiatry, and neuroscience
Maier, S.F., Nguyen, K.T., Deak, T., Milligan, E.D., Watkins, L.R., 1999. Stress, in the 21st century. Am. J. Psychiatry 159, 695–704.
learned helplessness, and brain interleukin-1 beta. Adv. Exp. Med. Biol. Martin-Villalba, A., Hahne, M., Kleber, S., Vogel, J., Falk, W., Schenkel, J.,
461, 235–249. Krammer, P.H., 2001. Therapeutic neutralization of CD95-ligand and TNF
Maier, S.F., Amat, J., Baratta, M.V., Paul, E., Watkins, L.R., 2006. Behavioral attenuates brain damage in stroke. Cell Death Differ. 8, 679–686.
control, the medial prefrontal cortex, and resilience. Dialogues Clin. Masana, M.I., Heyes, M.P., Mefford, I.N., 1990. Indomethacin prevents
Neurosci. 8, 397–406. increased catecholamine turnover in rat brain following systemic endotoxin
Makino, S., Shibasaki, T., Yamauchi, N., Nishioka, T., Mimoto, T., Wakabaya- challenge. Prog. Neuropsychopharmacol. Biol. Psychiatry 14, 609–621.
shi, I., Gold, P.W., Hashimoto, K., 1999. Psychological stress increased Matheson, K., Anisman, H., 2003. Systems of coping associated with psycho-
corticotropin-releasing hormone mRNA and content in the central nucleus logical distress: a profile perspective. Stress 6, 223–234.
of the amygdala but not in the hypothalamic paraventricular nucleus in the Mattson, M.P., Maudsley, S., Martin, B., 2004. BDNF and 5-HT: a dynamic duo
rat. Brain Res. 850, 136–143. in age-related neuronal plasticity and neurodegenerative disorders. Trends
Malberg, J.E., Duman, R.S., 2003. Cell proliferation in adult hippocampus is Neurosci. 27, 589–594.
decreased by inescapable stress: reversal by fluoxetine treatment. Neurop- McCaffery, J.M., Frasure-Smith, N., Dubé, M.P., Théroux, P., Rouleau, G.A.,
sychopharmacology 28, 1562–1671. Duan, Q., Lespérance, F., 2006. Common genetic vulnerability to depres-
Malendowicz, L.K., Nussdorfer, G.G., Markowska, A., Tortorella, C., Nowak, sive symptoms and coronary artery disease: a review and development of
M., 1995. Investigations on the acute effects of neuropeptides on the candidate genes related to inflammation and serotonin. Psychosom. Med.
pituitary–adrenocortical function in normal and cold-stressed rats. II. 68, 187–200.
Neurotensin and neuromedin N. Exp. Toxicol. Pathol. 47, 41–45. McColl, B.W., Rothwell, N.J., Allan, S.M., 2007. Systemic inflammatory
Malendowicz, L.K., Rucinski, M., Belloni, A.S., Ziolkowska, A., Nussdorfer, stimulus potentiates the acute phase and CXC chemokine responses to
G.G., 2007. Leptin and the regulation of the hypothalamic–pituitary– experimental stroke and exacerbates brain damage via interleukin-1- and
adrenal axis. Int. Rev. Cytol. 263, 63–102. neutrophil-dependent mechanisms. J. Neurosci. 27, 4403–4412.
Malison, R.T., Price, L.H., Berman, R., van Dyck, C.H., Pelton, G.H., Car- McCulley, M.C., Day, I.N., Holmes, C., 2004. Association between inter-
penter, L., Sanacora, G., Owens, M.J., Nemeroff, C.B., Rajeevan, N., leukin 1-beta promoter (-511) polymorphism and depressive symptoms in
Baldwin, R.M., Seibyl, J.P., Innis, R.B., Charney, D.S., 1998. Reduced Alzheimer’s disease. Am. J. Med. Genet. B: Neuropsychiatr. Genet. 124,
brain serotonin transporter availability in major depression as measured by 50–53.
[123I]-2 beta-carbomethoxy-3 beta-(4-iodophenyl)tropane and single McDonald, W.M., Richard, I.H., DeLong, M.R., 2003. Prevalence, etiology,
photon emission computed tomography. Biol. Psychiatry 44, 1090–1098. and treatment of depression in Parkinson’s disease. Biol. Psychiatry 54,
Mangano, E.N., Hayley, S., in press. Inflammatory priming of the substantia 363–375.
nigra influences the impact of later paraquat exposure: Neuroimmune McEvoy, A.N., Bresnihan, B., FitzGerald, O., Murphy, E.P., 2004. Cycloox-
sensitization of neurodegeneration. Neurobiol. Aging, doi:10.1016/j. ygenase 2-derived prostaglandin E2 production by corticotropin-releasing
neurobiolaging.2007.11.020. hormone contributes to the activated cAMP response element binding
Manji, H.K., Chen, G., 2002. PKC, MAP kinases and the bcl-2 family of protein content in rheumatoid arthritis synovial tissue. Arthritis Rheum.
proteins as long-term targets for mood stabilizers. Mol. Psychiatry 7 (Suppl. 50, 1132–1145.
1), S46–S56. McEwen, B.S., 1994. Corticosteroids and hippocampal plasticity. Ann. NY
Manji, H.K., Duman, R.S., 2001. Impairments of neuroplasticity and cellular Acad. Sci. 746, 134–142.
resilience in severe mood disorders: implications for the development of McEwen, B.S., 1999. Permanence of brain sex differences and structural
novel therapeutics. Psychopharmacol. Bull. 35, 5–49. plasticity of the adult brain. Proc. Natl. Acad. Sci. U.S.A. 96, 7128–7130.
Manji, H.K., Drevets, W.C., Charney, D.S., 2001. The cellular neurobiology of McEwen, B.S., 2000. Allostasis and allostatic load: implications for neurop-
depression. Nat. Med. 7, 541–547. sychopharmacology. Neuropsychopharmacology 22, 108–124.
Manji, H.K., Quiroz, J.A., Sporn, J., Payne, J.L., Denicoff, K.A., Gray, N., McEwen, B.S., 2005. Glucocorticoids, depression, and mood disorders: struc-
Zarate Jr., C.A., Charney, D.S., 2003. Enhancing neuronal plasticity and tural remodeling in the brain. Metabolism 54, 20–23.
cellular resilience to develop novel, improved therapeutics for difficult-to- McEwen, B.S., 2006. Protective and damaging effects of stress mediators:
treat depression. Biol. Psychiatry 53, 707–742. central role of the brain. Dialogues Clin. Neurosci. 8, 367–381.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 63

McEwen, B.S., Magarinos, A.M., 2001. Stress and hippocampal plasticity: Merali, Z., Kent, P., Du, L., Hrdina, P., Palkovits, M., Faludi, G., Poulter, M.O.,
implications for the pathophysiology of affective disorders. Hum. Psycho- Bedard, T., Anisman, H., 2006b. Corticotropin-releasing hormone, arginine
pharmacol. 16, S7–S19. vasopressin, gastrin-releasing peptide, and neuromedin B alterations in
McGeer, E.G., McGeer, P.L., 1999. Brain inflammation in Alzheimer disease stress-relevant brain regions of suicides and control subjects. Biol. Psy-
and the therapeutic implications. Curr. Pharm. Des. 5, 821–836. chiatry 59, 594–602.
McGeer, P.L., Yasojima, K., McGeer, E.G., 2002. Association of interleukin-1 Mercier, G., Lennon, A.M., Renouf, B., Dessouroux, A., Ramaugé, M.,
beta polymorphisms with idiopathic Parkinson’s disease. Neurosci. Lett. Courtin, F., Pierre, M., 2004. MAP kinase activation by fluoxetine and
326, 67–69. its relation to gene expression in cultured rat astrocytes. J. Mol. Neurosci.
McGeer, P.L., Schwab, C., Parent, A., Doudet, D., 2003. Presence of 24, 207–216.
reactive microglia in monkey substantia nigra years after 1-methyl-4- Mertens, S., Benke, D., Mohler, H., 1993. GABA(A) receptor populations with
phenyl-1,2,3,6-tetrahydropyridine administration. Ann. Neurol. 54, novel subunit combinations and drug binding profiles identified in brain by
599–604. alpha 5- and delta-subunit-specific immunopurification. J. Biol. Chem. 268,
McGinn, L.K., Asnis, G.M., Rubinson, E., 1996. Biological and clinical 5965–5973.
validation of atypical depression. Psychiatry Res. 60, 191–198. Mervaala, E., Föhr, J., Könönen, M., Valkonen-Korhonen, M., Vainio, P.,
McGrath, P.J., Stewart, J.W., Harrison, W.M., Ocepek-Wilekson, K., Rabkin, Partanen, K., Partanen, J., Tiihonen, J., Viinamäki, H., Karjalainen,
J.G., Nunes, E.N., Wager, S.G., Tricamo, E., Quitkin, F.M., Klein, D.F., A.K., Lehtonen, J., 2000. Quantitative MRI of the hippocampus and
1992. Predictive value of symptoms of atypical depression for differential amygdala in severe depression. Psychol. Med. 30, 117–125.
drug treatment outcome. J. Clin. Psychopharmacol. 12, 197–202. Meyer, J.H., Kapur, S., Houle, S., DaSilva, J., Owczarek, B., Brown, G.M.,
Meaney, M.J., 2001. Maternal care, gene expression, and the transmission of Wilson, A.A., Kennedy, S.H., 1999. Prefrontal cortex 5-HT2 receptors in
individual differences in stress reactivity across generations. Ann. Rev. depression: an [18F]setoperone PET imaging study. Am. J. Psychiatry 156,
Neurosci. 24, 1161–1192. 1029–1034.
Melnikova, T., Savonenko, A., Wang, Q., Liang, X., Hand, T., Wu, L., Meyer, J.H., Kapur, S., Eisfeld, B., Brown, G.M., Houle, S., DaSilva, J., Wilson,
Kaufmann, W.E., Vehmas, A., Andreasson, K.I., 2006. Cycloxygenase-2 A.A., Rafi-Tari, S., Mayberg, H.S., Kennedy, S.H., 2001a. The effect of
activity promotes cognitive deficits but not increased amyloid burden in a paroxetine on 5-HT(2A) receptors in depression: an [(18)F]setoperone PET
model of Alzheimer’s disease in a sex-dimorphic pattern. Neuroscience 141, imaging study. Am. J. Psychiatry 158, 78–85.
1149–1162. Meyer, J.H., Wilson, A.A., Ginovart, N., Goulding, V., Hussey, D., Hood, K.,
Meltzer, C.C., Price, J.C., Mathis, C.A., Greer, P.J., Cantwell, M.N., Houck, Houle, S., 2001b. Occupancy of serotonin transporters by paroxetine and
P.R., Mulsant, B.H., Ben-Eliezer, D., Lopresti, B., DeKosky, S.T., Reynolds, citalopram during treatment of depression: a [(11)C]DASB PET imaging
C.F., 1999. PET imaging of serotonin type 2A receptors in late-life study. Am. J. Psychiatry 158, 1843–1849.
neuropsychiatric disorders. Am. J. Psychiatry 156, 1871–1878. Meyer, J.H., McMain, S., Kennedy, S.H., Korman, L., Brown, G.M., DaSilva,
Menkes, D.B., MacDonald, J.A., 2000. Interferons, serotonin and neurotoxicity. J.N., Wilson, A.A., Blak, T., Eynan-Harvey, R., Goulding, V.S., Houle, S.,
Psychol. Med. 30, 259–268. Links, P., 2003. Dysfunctional attitudes and 5-HT2 receptors during depres-
Menzies, R., Phelps, C., Wiranowska, M., Oliver, J., Chen, L., Horvath, E., Hall, sion and self-harm. Am. J. Psychiatry 160, 90–99.
N., 1996. The effect of interferon-alpha on the pituitary–adrenal axis. J. Meyer, J.H., Houle, S., Sagrati, S., Carella, A., Hussey, D.F., Ginovart, N.,
Interferon Cytokine Res. 16, 619–629. Goulding, V., Kennedy, J., Wilson, A.A., 2004. Brain serotonin transporter
Merali, Z., Lacosta, S., Anisman, H., 1997. Effects of interleukin-1b and mild binding potential measured with carbon 11-labeled DASB positron emis-
stress on alterations of central monoamines: a regional microdialysis study. sion tomography: effects of major depressive episodes and severity of
Brain Res. 761, 225–235. dysfunctional attitudes. Arch. Gen. Psychiatry 61, 1271–1279.
Merali, Z., McIntosh, J., Kent, P., Michaud, D., Anisman, H., 1998. Aversive as Meyers, C.A., Valentine, A.D., 1995. Neurological and psychiatric adverse
well as appetitive events evoke the release of corticotropin releasing effects of immunological therapy. CNS Drugs 3, 56–68.
hormone and bombesin-like peptides at the central nucleus of the amygdala. Meyers, C.A., 1999. Mood and cognitive disorders in cancer patients receiving
J. Neurosci. 18, 4758–4766. cytokine therapy. Adv. Exp. Med. Biol. 461, 75–81.
Merali, Z., McIntosh, J., Anisman, H., 1999. Role of bombesin-related peptides Meynen, G., Unmehopa, U.A., van Heerikhuize, J.J., Hofman, M.A., Swaab,
in the control of food intake: a review. Neuropeptides 33, 376–386. D.F., Hoogendijk, W.J., 2006. Increased arginine vasopressin mRNA
Merali, Z., Kent, P., Michaud, D., McIntyre, D., Anisman, H., 2001. Differential expression in the human hypothalamus in depression: a preliminary report.
effects of psychogenic and neurogenic stressors on central corticotropin- Biol. Psychiatry 60, 892–895.
releasing hormone and bombesin-like peptides in the Fast and Slow seizing Meynen, G., Unmehopa, U.A., Hofman, M.A., Swaab, D.F., Hoogendijk, W.J.,
rat. Brain Res. 906, 60–73. 2007. Relation between corticotropin-releasing hormone neuron number in
Merali, Z., Brennan, K., Brau, P., Anisman, H., 2002a. Dissociating anorexia the hypothalamic paraventricular nucleus and depressive state in Alzhei-
and anhedonia elicited by interleukin-1b: antidepressant and gender effects mer’s disease. Neuroendocrinology 85, 37–44.
on responding for ‘‘Free Chow’’ and ‘‘Earned’’ sucrose intake. Psycho- Millan, M.J., 2005. Serotonin 5-HT2C receptors as a target for the treatment of
pharmacology 165, 413–418. depressive and anxious states: focus on novel therapeutic strategies. Ther-
Merali, Z., Du, L., Hrdina, P., Palkovits, M., Faludi, G., Poulter, M.O., Anisman, apie 60, 441–460.
H., 2004b. Dysregulation in the suicide brain: mRNA expression of Millan, M.J., 2006. Multi-target strategies for the improved treatment of
corticotropin-releasing hormone receptors and GABA(A) receptor subunits depressive states: conceptual foundations and neuronal substrates, drug
in frontal cortical brain region. J. Neurosci. 24, 1478–1485. discovery and therapeutic application. Pharmacol. Ther. 110, 135–370.
Merali, Z., Kent, P., Anisman, H., 2002b. Role of bombesin-related peptides in Miller, A.H., Raison, C.L., 2006. Cytokines, p38 MAP kinase and the patho-
the mediation or integration of the stress response. Cell Mol. Life Sci. 59, physiology of depression. Neuropsychopharmacology 31, 2089–2090.
272–287. Miller, G.E., Stetler, C.A., Carney, R.M., Freedland, K.E., Banks, W.A., 2002.
Merali, Z., Khan, S., Michaud, D.S., Shippy, S.A., Anisman, H., 2004a. Does Clinical depression and inflammatory risk markers for coronary heart
amygdaloid corticotropin-releasing hormone (CRH) mediate anxiety-like disease. Am. J. Cardiol. 90, 1279–1283.
behaviors? Dissociation of anxiogenic effects and CRH release. Eur. J. Miller, G.E., Freedland, K.E., Duntley, S., Carney, R.M., 2005. Relation of
Neurosci. 20, 229–239. depressive symptoms to C-reactive protein and pathogen burden (cytome-
Merali, Z., Bédard, T., Andrews, N., Davis, B., McKnight, A.T., Isabel galovirus, herpes simplex virus, Epstein-Barr virus) in patients with earlier
Gonzalez, M., Pritchard, M., Kent, P., Anisman, H., 2006a. Bombesin acute coronary syndromes. Am J. Cardiol. 95, 317–321.
receptors as novel anti-anxiety therapeutic target; non-peptide antagonist Minami, M., Kuraishi, Y., Yabuuchi, K., Yamazaki, A., Satoh, M., 1992.
PD 176252 reduces anxiety and 5-HT release through BB1 receptor. J. Induction of interleukin-1 beta mRNA in rat brain after transient forebrain
Neurosci. 26, 10387–10396. ischemia. J. Neurochem. 58, 390–392.
64 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Minami, M., Katayama, T., Satoh, M., 2006. Brain cytokines and chemokines: Moody, T.W., Merali, Z., 2004. Bombesin-like peptides and associated recep-
roles in ischemic injury and pain. J. Pharmacol. Sci. 100, 461–470. tors within the brain: distribution and behavioral implications. Peptides 25,
Minami, M., Kuraishi, Y., Yamaguchi, T., Nakai, S., Hirai, Y., Satoh, M., 1991. 511–520.
Immobilization stress induces interleukin-1b mRNA in rat hypothalamus. Moody, T.W., O’Donohue, T.L., Jacobowitz, D.M., 1981. Biochemical locali-
Neurosci. Lett. 123, 254–256. zation and characterization of bombesin-like peptides in discrete regions of
Minamino, N., Kangawa, K., Matsuo, H., 1984. Neuromedin B is a major rat brain. Peptides 2, 75–80.
bombesin-like peptide in rat brain: regional distribution of neuromedin B Morganti-Kossmann, M.C., Kossmann, T., Wahl, S.M., 1992. Cytokines and
and neuromedin C in rat brain, pituitary and spinal cord. Biochem. Biophys. neuropathology. Trends Pharmacol. Sci. 13, 286–291.
Res. Commun. 124, 925–932. Morikawa, O., Sakai, N., Obara, H., Saito, N., 1998. Effects of interferon-alpha,
Minogue, A.M., Schmid, A.W., Fogarty, M.P., Moore, A.C., Campbell, V.A., interferon-gamma and cAMP on the transcriptional regulation of the
Herron, C.E., Lynch, M.A., 2003. Activation of the c-Jun N-terminal kinase serotonin transporter. Eur. J. Pharmacol. 349, 317–324.
signaling cascade mediates the effect of amyloid-beta on long term poten- Morrow, B.A., Redmond, A.J., Roth, R.H., Elsworth, J.D., 2000. The predator
tiation and cell death in hippocampus: a role for interleukin-1beta? J. Biol. odor, TMT, displays a unique, stress-like pattern of dopaminergic and
Chem. 278, 27971–27980. endocrinological activation in the rat. Brain Res. 864, 146–151.
Mintun, M.A., Sheline, Y.I., Moerlein, S.M., Vlassenko, A.G., Huang, Y., Mount, M., Lira, A., Alyeason, H., Grimes, D., Smith, P., Slack, R., Anisman,
Snyder, A.Z., 2004. Decreased hippocampal 5-HT2A receptor binding in H., Hayley, S., Park, D.S., 2007. Involvement of interferon-gamma in
major depressive disorder: in vivo measurement with [18F]altanserin microglial mediated loss of dopaminergic neurons. J. Neurosci. 27,
positron emission tomography. Biol. Psychiatry 55, 217–224. 3328–3337.
Mitchell, A.J., 1998. The role of corticotropin releasing factor in depressive Mountney, C., Sillberg, V., Kent, P., Anisman, H., Merali, Z., 2006. The role of
illness: a critical review. Neurosci. Biobehav. Rev. 22, 635–651. gastrin-releasing peptide on conditioned fear: differential cortical and
Mitchell, A., O’Keane, V., 1998. Steroids and depression. BMJ 316, amygdaloid responses in the rat. Psychopharmacology 189, 287–296.
244–245. Mrak, R.E., Sheng, J.G., Griffin, W.S., 1995. Glial cytokines in Alzheimer’s
Miyahara, S., Komori, T., Fujiwara, R., Shizuya, K., Yamamoto, M., Ohmori, disease: review and pathogenic implications. Hum. Pathol. 26, 816–823.
M., Okazaki, Y., 2000. Effects of repeated stress on expression of inter- Mullberg, J., Oberthur, W., Lottspeich, F., Mehl, E., Dittrich, E., Graeve, L.,
leukin-6 (IL-6) and IL-6 receptor mRNAs in rat hypothalamus and mid- Heinrich, P.C., Rose-John, S., 1994. The soluble human IL-6 receptor.
brain. Life Sci. 66, PL93–PL98. Mutational characterization of the proteolytic cleavage site. J. Immunol.
Miyaoka, H., Otsubo, T., Kamijima, K., Ishii, M., Onuki, M., Mitamura, K., 152, 4958–4968.
1999. Depression from interferon therapy in patients with hepatitis C. Am. Muller, M.B., Zimmermann, S., Sillaber, I., Hagemeyer, T.P., Deussing, J.M.,
J. Psychiatry 156, 1120. Timpl, P., Kormann, M.S., Droste, S.K., Kuhn, R., Reul, J.M., Holsboer, F.,
Mogi, M., Harada, M., Riederer, P., Narabayashi, H., Fujita, K., Nagatsu, T., Wurst, W., 2003. Limbic corticotropin-releasing hormone receptor 1 med-
1994. Tumor necrosis factor-alpha (TNF-alpha) increases both in the brain iates anxiety-related behavior and hormonal adaptation to stress. Nat.
and in the cerebrospinal fluid from parkinsonian patients. Neurosci. Lett. Neurosci. 6, 1100–1107.
165, 208–210. Muller, N., Schwarz, M.J., Dehning, S., Douhe, A., Cerovecki, A., Goldstein-
Mogi, M., Harada, M., Kondo, T., Riederer, P., Nagatsu, T., 1995. Brain beta 2- Muller, B., Spellmann, I., Hetzel, G., Maino, K., Kleindienst, N., Moller,
microglobulin levels are elevated in the striatum in Parkinson’s disease. J. H.J., Arolt, V., Riedel, M., 2006. The cyclooxygenase-2 inhibitor celecoxib
Neural. Transm. Park. Dis. Dement. 9, 87–92. has therapeutic effects in major depression: results of a double-blind,
Mogi, M., Harada, M., Narabayashi, H., Inagaki, H., Minami, M., Nagatsu, T., randomized, placebo controlled, add-on pilot study to reboxetine. Mol.
1996. Interleukin (IL)-1 beta, IL-2, IL-4, IL-6 and transforming growth Psychiatry 11, 680–684.
factor-alpha levels are elevated in ventricular cerebrospinal fluid in Murakami, S., Imbe, H., Morikawa, Y., Kubo, C., Senba, E., Nadjar, et al.,
juvenile parkinsonism and Parkinson’s disease. Neurosci. Lett. 211, 2005. Chronic stress, as well as acute stress, reduces BDNF mRNA
13–16. expression in the rat hippocampus but less robustly. Neurosci. Res. 53,
Mohankumar, S.M., MohanKumar, P.S., 2005. Systemic interleukin-1beta 129–139.
stimulates the simultaneous release of norepinephrine in the paraventricular Murphy, B.E., 1997. Antiglucocorticoid therapies in major depression: a review.
nucleus and the median eminence. Brain Res. Bull. 65, 451–456. Psychoneuroendocrinology 22 (Suppl. 1), S125–S132.
Mohankumar, P.S., Quadri, S.K., 1993. Systemic administration of interleukin- Murphy, E.K., Sved, A.F., Finlay, J.M., 2003. Corticotropin-releasing hormone
1 stimulates norepinephrine release in the paraventricular nucleus. Life Sci. receptor blockade fails to alter stress-evoked catecholamine release in
52, 1961–1967. prefrontal cortex of control or chronically stressed rats. Neuroscience
Mohr, D.C., 2007. Stress and multiple sclerosis. J. Neurol. 254 (Suppl. 2), II65– 116, 1081–1087.
II68. Murray, P.J., 2007. The JAK-STAT signaling pathway: input and output
Moldofsky, H., 1995. Sleep and the immune system. Int. J. Immunopharmacol. integration. J. Immunol. 178, 2623–2629.
17, 649–665. Musselman, D.L., Lawson, D.H., Gumnick, J.F., Manatunga, A., Penna, S.,
Momose, K., Inui, A., Asakawa, A., Ueno, N., Nakajima, M., Fujimiya, M., Goodkin, R., Greiner, K., Nemeroff, C., Miller, A.H., 2001. Paroxetine for
Kasuga, M., 1999. Intracerebroventricularly administered corticotropin- the prevention of the depression and neurotoxicity induced by high dose
releasing factor inhibits food intake and produces anxiety-like behaviour interferon alpha. N. Engl. J. Med. 344, 961–966.
at very low doses in mice. Diabetes. Obes. Metab. 1, 281–284. Nadeau, S., Rivest, S., 1999. Effects of circulating tumor necrosis factor on the
Monje, M.L., Toda, H., Palmer, T.D., 2003. Inflammatory blockade restores neuronal activity and expression of the genes encoding the tumor necrosis
adult hippocampal neurogenesis. Science 302, 1760–1765. factor receptors (p55 and p75) in the rat brain: a view from the blood–brain
Monroe, S.M., Depue, R.A., 1991. Life stress and depression. In: Becker, J., barrier. Neuroscience 93, 1449–1464.
Kleinman, A. (Eds.), Psychosocial Aspects of Depression. Erlbaum, Hills- Nadjar, A., Combe, C., Busquet, P., Dantzer, R., Parnet, P., 2005. Signaling
dale, NJ, pp. 101–130. pathways of interleukin-1 actions in the brain: anatomical distribution of
Montaron, M.F., Drapeau, E., Dupret, D., Kitchener, P., Aurousseau, C., Le phospho-ERK1/2 in the brain of rat treated systemically with interleukin-
Moal, M., Piazza, P.V., Abrous, D.N., 2006. Lifelong corticosterone level 1beta. Neuroscience 134, 921–932.
determines age-related decline in neurogenesis and memory. Neurobiol. Naef, L., Woodside, B., 2007. Prolactin/leptin interactions in the control of food
Aging 27, 645–654. intake in rats. Endocrinology 148, 5977–5983.
Monteggia, L.M., Luikart, B., Barrot, M., Theobold, D., Malkovska, I., Nef, S., Nahon, E., Israelson, A., Abu-Hamad, S., Varda, S.B., 2005. Fluoxetine
Parada, L.F., Nestler, E.J., 2007. Brain-derived neurotrophic factor condi- (Prozac) interaction with the mitochondrial voltage-dependent anion chan-
tional knockouts show gender differences in depression-related behaviors. nel and protection against apoptotic cell death. FEBS Lett. 579, 105.R–
Biol. Psychiatry 61, 187–197. 110.R.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 65

Nankai, M., Yamada, S., Muneoka, K., Toru, M., 1995. Increased 5HT2 Nishimoto, M., Furuta, A., Aoki, S., Kudo, Y., Miyakawa, H., Wada, K., 2007.
receptor-mediated behavior 11 days after shock in learned helplessness PACAP/PAC1 autocrine system promotes proliferation and astrogenesis in
rats. Eur. J. Pharmacol. 281, 123–130. neural progenitor cells. Glia 55, 317–327.
Nankova, B., Devlin, D., Kvetnansky, R., Kopin, I.J., Sabban, E.L., 1993. Nolen-Hoeksema, S., 1998. Ruminative coping with depression. In: Heckhau-
Repeated immobilization stress increases the binding of c-fos-like proteins sen, J., Dweck, C.S. (Eds.), Motivation and Self-regulation across the Life
to a rat dopamine-b-hydroxylase promoter enhancer sequence. J. Neuro- Span. Cambridge University Press, Cambridge, pp. 237–256.
chem. 61, 776–779. Nutt, D.J., 2002. The neuropharmacology of serotonin and noradrenaline in
Nelson, O., Tu, H., Lei, T., Bentahir, M., de Strooper, B., Bezprozvanny, I., depression. Int. Clin. Psychopharmacol. 17 (Suppl. 1), S1–S12.
Neta, et al., 2007. Familial Alzheimer disease-linked mutations specifically Nutt, D.J., Stein, D.J., 2006. Understanding the neurobiology of comorbidity in
disrupt Ca2+ leak function of presenilin 1. J. Clin. Invest. 117, 1230–1239. anxiety disorders. CNS Spectr. 11, 13–20.
Nemeroff, C.B., 1996. The corticotropin-releasing factor (CRF) hypothesis of Nuyen, J., Schellevis, F.G., Satariano, W.A., Spreeuwenberg, P.M., Birkner,
depression: New findings and new directions. Mol. Psychiatry 1, 336–342. M.D., van den Bos, G.A., Groenewegen, P.P., 2006. Comorbidity was
Nemeroff, C.B., Vale, W., 2005. The neurobiology of depression: inroads to associated with neurologic and psychiatric diseases: a general practice-
treatment and new drug discovery. J. Clin. Psychiatry 66 (Suppl. 7), 5–13. based controlled study. J. Clin. Epidemiol. 59, 1274–1284.
Nemeroff, C.B., Widerlov, E., Bissette, G., Walleus, H., Karlsson, I., Eklund, Oberholzer, A., Souza, S.M., Tschoeke, S.K., Oberholzer, C., Abouhamze, A.,
K., Kilts, C.D., Loosen, P.T., Vale, W., 1984. Elevated concentrations of Pribble, J.P., Moldawer, L.L., 2005. Plasma cytokine measurements aug-
CSF corticotropin-releasing factor-like immunoreactivity in depressed ment prognostic scores as indicators of outcome in patients with severe
patients. Science 226, 1342–1344. sepsis. Shock 23, 488–493.
Nemeroff, C.B., Owens, M.J., Bissette, G., Andorn, A.C., Stanley, M., 1988. Obuchowicz, E., Kowalski, J., Labuzek, K., Krysiak, R., Pendzich, J., Herman,
Reduced corticotropin releasing factor binding sites in the frontal cortex of Z.S., 2006. Amitriptyline and nortriptyline inhibit interleukin-1 release by
suicide victims. Arch. Gen. Psychiatry 45, 577–579. rat mixed glial and microglial cell cultures. Int. J. Neuropsychopharmacol.
Nemeroff, C.B., Bissette, G., Akil, H., Fink, M., 1991. Neuropeptide concen- 9, 27–35.
trations in the cerebrospinal fluid of depressed patients treated with elec- Odagaki, Y., Garcı́a-Sevilla, J.A., Huguelet, P., La Harpe, R., Koyama, T.,
troconvulsive therapy. Corticotrophin-releasing factor, beta-endorphin and Guimón, J., 2001. Cyclic AMP-mediated signaling components are upre-
somatostatin. Br. J. Psychiatry 158, 59–63. gulated in the prefrontal cortex of depressed suicide victims. Brain Res. 898,
Nestler, E.J., Barrot, M., DiLeone, R.J., Eisch, A.J., Gold, S.J., Monteggia, 224–231.
L.M., 2002. Neurobiology of depression. Neuron 34, 13–25. Offen, D., Sherki, Y., Melamed, E., Fridkin, M., Brenneman, D.E., Gozes, I.,
Neta, R., Sayers, T.J., Oppenheim, J.J., 1992. Relationship of TNF to inter- 2000. Vasoactive intestinal peptide (VIP) prevents neurotoxicity in neuronal
leukins. Immunol. Ser. 56, 499–566. cultures: relevance to neuroprotection in Parkinson’s disease. Brain Res.
Neumeister, A., Konstantinidis, A., Stastny, J., Schwarz, M.J., Vitouch, O., 854, 257–262.
Willeit, M., Praschak-Rieder, N., Zach, J., de Zwaan, M., Bondy, B., Ohata, H., Arai, K., Shibasaki, T., 2002. Effect of chronic administration of a
Ackenheil, M., Kasper, S., 2002. Association between serotonin transporter CRF(1) receptor antagonist, CRA1000, on locomotor activity and endocrine
gene promoter polymorphism (5HTTLPR) and behavioral responses to responses to stress. Eur. J. Pharmacol. 457, 201–206.
tryptophan depletion in healthy women with and without family history of Olsen, L., Knigge, U., Warberg, J., 1992. Gastrin-releasing peptide stimulation
depression. Arch. Gen. Psychiatry 59, 613–620. of corticotropin secretion in male rats. Endocrinology 130, 2710–2716.
Neumeister, A., Nugent, A.C., Waldeck, T., Geraci, M., Schwarz, M., Bonne, Orellana, D.I., Quintanilla, R.A., Maccioni, R.B., 2007. Neuroprotective effect
O., Bain, E.E., Luckenbaugh, D.A., Herscovitch, P., Charney, D.S., Drevets, of TNFalpha against the beta-amyloid neurotoxicity mediated by CDK5
W.C., 2004. Neural and behavioral responses to tryptophan depletion in kinase. Biochim. Biophys. Acta 1773, 254–263.
unmedicated patients with remitted major depressive disorder and controls. Oshima, A., Flachskamm, C., Reul, J.M., Holsboer, F., Linthorst, A.C., 2003.
Arch. Gen. Psychiatry 61, 765–773. Altered serotonergic neurotransmission but normal hypothalamic–pitui-
Neumaier, J.F., Petty, F., Kramer, G.L., Szot, P., Hamblin, M.W., 1997. Learned tary–adrenocortical axis activity in mice chronically treated with the
helplessness increases 5-hydroxytryptamine1B receptor mRNA levels in the corticotropin-releasing hormone receptor Type 1 Antagonist NBI 30775.
rat dorsal raphe nucleus. Biol. Psychiatry 41, 668–674. Neuropsychopharmacology 28, 2148–2159.
Newton, S.S., Duman, R.S., 2004. Regulation of neurogenesis and angiogenesis Ossowska, G., Nowa, G., Kata, R., Klenk-Majewska, B., Danilczuk, Z.,
in depression. Curr. Neurovasc. Res. 1, 261–267. Zebrowska-Lupina, I., 2001. Brain monoamine receptors in a chronic
Nguyen, K.T., Deak, T., Owens, S.M., Kohno, T., Fleshner, M., Watkins, L.R., unpredictable stress model in rats. J. Neural Transm. 108, 311–319.
Maier, S.F., 1998. Exposure to acute stress induces brain interleukin-1b Otsuka, R., Yatsuya, H., Tamakoshi, K., Matsushita, K., Wada, K., Toyoshima,
protein in the rat. J. Neurosci. 19, 2799–2805. H., 2006. Perceived psychological stress and serum leptin concentrations in
Nguyen, P.V., Woo, N.H., 2003. Regulation of hippocampal synaptic plasticity Japanese men. Obesity 14, 1832–1838.
by cyclic AMP-dependent protein kinases. Prog. Neurobiol. 71, 401–437. Owen, F., Cross, A.J., Crow, T.J., Deakin, J.F., Ferrier, I.N., Lofthouse, R.,
Niemand, C., Nimmesgern, A., Haan, S., Fischer, P., Schaper, F., Rossaint, R., Poulter, M., 1983. Brain 5-HT-2 receptors and suicide. Lancet 2, 1256.
Heinrich, P.C., Muller-Newen, G., 2003. Activation of STAT3 by IL-6 and Owen, F., Chambers, D.R., Cooper, S.J., Crow, T.J., Johnson, J.A., Lofthouse,
IL-10 in primary human macrophages is differentially modulated by R., Poulter, M., 1986. Serotonergic mechanisms in brains of suicide victims.
suppressor of cytokine signaling 3. J. Immunol. 170, 3263–3272. Brain Res. 362, 185–188.
Niimi, M., Sato, M., Wada, Y., Takahara, J., Kawanishi, K., 1997. Effect of Owens, M.J., Edwards, E., Nemeroff, C.B., 1990. Effects of 5-HT1A receptor
continuous intravenous injection of interleukin-6 and pretreatment with agonists on hypothalamo–pituitary–adrenal axis activity and corticotropin-
cyclooxygenase inhibitor on brain c-fos expression in the rat. Neuroendo- releasing factor containing neurons in the rat brain. Eur. J. Pharmacol. 190,
crinology 66, 47–53. 113–122.
Niissalo, S., Hukkanen, M., Imai, S., Törnwall, J., Konttinen, Y.T., 2002. Owens, T., 2002. Identification of new therapeutic targets for prevention of CNS
Neuropeptides in experimental and degenerative arthritis. Ann. NY Acad. inflammation. Expert Opin. Ther. Targets 6, 203–215.
Sci. 966, 384–399. Ownby, R.L., Crocco, E., Acevedo, A., John, V., Loewenstein, D., 2006.
Nisenbaum, L.K., Abercrombie, E.D., 1992. Enhanced tyrosine hydroxylation Depression and risk for Alzheimer disease: systematic review, meta-ana-
in hippocampus of chronically stressed rats upon exposure to a novel lysis, and metaregression analysis. Arch. Gen. Psychiatry 63, 530–538.
stressor. J. Neurochem. 58, 276–281. Pacak, K., McCarty, R., Palkovits, M., Kopin, I.J., Goldstein, D.S., 1995a.
Nisenbaum, L.K., Zigmond, M.J., Sved, A.F., Abercrombie, E.D., 1991. Prior Effects of immobilization on in vivo release of norepinephrine in the bed
exposure to chronic stress results in enhanced synthesis and release of nucleus of the stria terminalis in conscious rats. Brain Res. 688, 242–246.
hippocampal norepinephrine in response to a novel stressor. J. Neurosci. 11, Pacak, K., Palkovits, M., Kopin, I.J., Goldstein, D.S., 1995b. Stress-induced
1478–1484. norepinephrine release in the hupothalamic paraventricular nucleus and
66 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

pituitary–adrenal and sympathoadrenal activity: in vivo microdialysis stu- Perera, T.D., Coplan, J.D., Lisanby, S.H., Lipira, C.M., Arif, M., Carpio, C.,
dies. Front. Neuroedocrinol. 16, 89–150. Spitzer, G., Santarelli, L., Scharf, B., Hen, R., Rosoklija, G., Sackeim, H.A.,
Pacchioni, A.M., Gioino, G., Assis, A., Cancela, L.M., 2002. A single exposure Dwork, A.J., 2007. Antidepressant-induced neurogenesis in the hippocam-
to restraint stress induces behavioral and neurochemical sensitization to pus of adult nonhuman primates. J. Neurosci. 27, 4894–4901.
stimulating effects of amphetamine: involvement of NMDA receptors. Ann. Perry, V.H., Newman, T.A., Cunningham, C., 2003. The impact of systemic
NY Acad. Sci. 965, 233–246. infection on the progression of neurodegenerative disease. Nat. Rev.
Pahan, K., Sheikh, F.G., Liu, X., Hilger, S., McKinney, M., Petro, T.M., 2001. Neurosci. 4, 103–112.
Induction of nitric-oxide synthase and activation of NF-kappaB by inter- Petersen, A.M., Pedersen, B.K., 2005. The anti-inflammatory effect of exercise.
leukin-12 p40 in microglial cells. J. Biol. Chem. 276, 7899–7905. J. Appl. Physiol. 98, 1154–1162.
Palmer, K., Berger, A.K., Monastero, R., Winblad, B., Bäckman, L., Fratiglioni, Petty, F., Kramer, G., Wilson, L., 1992. Prevention of learned helplessness: in
L., 2007. Predictors of progression from mild cognitive impairment to vivo correlation with cortical serotonin. Pharmacol. Biochem. Behav. 43,
Alzheimer disease. Neurology 68, 1596–1602. 361–367.
Pandey, G.N., Dwivedi, Y., Rizavi, H.S., Ren, X., Pandey, S.C., Pesold, C., Petty, F., Kramer, G., Wilson, L., Jordan, S., 1994. In vivo serotonin release and
Roberts, R.C., Conley, R.R., Tamminga, C.A., 2002. Higher expression of learned helplessness. Psychiatry Res. 52, 285–293.
serotonin 5-HT(2A) receptors in the postmortem brains of teenage suicide Pezzone, M.A., Lee, W.-S., Hoffman, G.E., Rabin, B.S., 1992. Induction of c-
victims. Am. J. Psychiatry 159, 419–429. Fos immunoreactivity in the rat forebrain by conditioned and unconditioned
Pandey, G.N., Dwivedi, Y., Ren, X., Rizavi, H.S., Roberts, R.C., Conley, R.R., aversive stimuli. Brain Res. 597, 41–50.
2006. Cyclic AMP response element-binding protein in post-mortem brain Pezzone, M.A., Lee, W.-S., Hoffman, G.E., Pezzone, K.M., Rabin, B.S., 1993.
of teenage suicide victims: specific decrease in the prefrontal cortex but not Activation of brainstem catecholaminergic neurons by conditioned and
the hippocampus. Int. J. Neuropsychopharmacol. 18, 1–9. unconditioned aversive stimuli as revealed by c-Fos immunoreactivity.
Parissis, J.T., Fountoulaki, K., Filippatos, G., Adamopoulos, S., Paraskevaidis, Brain Res. 608, 310–318.
I., Kremastinos, D., 2007. Depression in coronary artery disease: novel Piao, C.S., Yu, Y.M., Han, P.L., Lee, J.K., 2003. Dynamic expression of p38beta
pathophysiologic mechanisms and therapeutic implications. Int. J. Cardiol. MAPK in neurons and astrocytes after transient focal ischemia. Brain Res.
116, 153–160. 976, 120–124.
Parsadaniantz, S.M., Lebeau, A., Duval, P., Grimaldi, B., Terlain, B., Kerdelhué, Piazza, P.V., Le Moal, M., 1997. Glucocorticoids as a biological substrate of
B., 2000. Effects of the inhibition of cyclo-oxygenase 1 or 2 or 5-lipox- reward: physiological and pathophysiological implications. Brain Res.
ygenase on the activation of the hypothalamic–pituitary–adrenal axis induced Brain Res. Rev. 25, 359–372.
by interleukin-1beta in the male rat. J. Neuroendocrinol. 12, 766–773. Piccinni, A., Marazziti, D., Catena, M., Domenici, L., Del Debbio, A., Bianchi,
Parsey, R.V., Oquendo, M.A., Ogden, R.T., Olvet, D.M., Simpson, N., Huang, C., Mannari, C., Martini, C., Da Pozzo, E., Schiavi, E., Mariotti, A.,
Y.Y., Van Heertum, R.L., Arango, V., Mann, J.J., 2006b. Altered serotonin Roncaglia, I., Palla, A., Consoli, G., Giovannini, L., Massimetti, G.,
1A binding in major depression: a [carbonyl-C-11]WAY100635 positron Dell’osso, L., 2007. Plasma and serum brain-derived neurotrophic factor
emission tomography study. Biol. Psychiatry 59, 106–113. (BDNF) in depressed patients during 1 year of antidepressant treatments. J.
Parsey, R.V., Olvet, D.M., Oquendo, M.A., Huang, Y.Y., Ogden, R.T., Mann, Affect. Disord. 105, 279–283.
J.J., 2006a. Higher 5-HT1A receptor binding potential during a major Pich, E.M., Lorang, M., Yeganeh, M., de Fonseca, F.R., Raber, J., Koob, G.F.,
depressive episode predicts poor treatment response: preliminary data from Weiss, F., 1995. Increase of extracellular corticotropin-releasing factor-like
a naturalistic study. Neuropsychopharmacology 31, 1745–1749. immunoreactivity levels in the amygdala of awake rats during restraint
Parsey, R.V., Hastings, R.S., Oquendo, M.A., Huang, Y.Y., Simpson, N., stress and ethanol withdrawal as measured by microdialysis. J. Neurosci. 15,
Arcement, J., Huang, Y., Ogden, R.T., Van Heertum, R.L., Arango, V., 5439–5447.
Mann, J.J., 2006c. Lower serotonin transporter binding potential in the Pineyro, G., Blier, P., 1999. Autoregulation of serotonin neurons: role in
human brain during major depressive episodes. Am. J. Psychiatry 163, 52– antidepressant drug action. Pharmacol. Rev. 51, 533–591.
58. Pinnock, R.D., Woodruff, G.N., 1991. Bombesin excites a subpopulation of 5-
Pasco, J.A., Jacka, F.N., Williams, L.J., Henry, M.J., Nicholson, G.C., Koto- hydroxytryptamine-sensitive neurones in the rat dorsal raphe nucleus in
wicz, M.A., Berk, M., 2007. Leptin in depressed women: cross-sectional vitro. J. Physiol. 440, 55–65.
and longitudinal data from an epidemiologic study. J. Affect. Disord., Pinnock, R.D., Reynolds, T., Woodruff, G.N., 1994. Different types of bombe-
doi:10.1016/j.jad.2007.07.024. sin receptors on neurons in the dorsal raphe nucleus and the rostral
Pasic, J., Levy, W.C., Sullivan, M.D., 2003. Cytokines in depression and heart hypothalamus in rat brain slices in vitro. Brain Res. 653, 119–124.
failure. Psychosom. Med. 65, 181–193. Pinteaux, E., Inoue, W., Schmidt, L., Molina-Holgado, F., Rothwell, N.J.,
Pavol, M.A., Meyers, C.A., Rexer, J.L., Valentine, A.D., Mattis, P.J., Talpaz, M., Luheshi, G.N., 2007. Leptin induces interleukin-1beta release from rat
1995. Pattern of neurobehavioral deficits associated with interferon alfa microglial cells through a caspase 1 independent mechanism. J. Neurochem.
therapy for leukemia. Neurology 45, 947–950. 102, 826–833.
Paykel, E.S., 2003. Life events and affective disorders. Acta Psychiatr. Scand. Pitman, R.K., Orr, S.P., Shalev, A.Y., Metzger, L.J., Mellman, T.A., 1999.
Suppl. 418, 61–66. Alterations in post-traumatic stress disorder. Psychophysiological Semin.
Penalva, R.G., Flachskamm, C., Zimmermann, S., Wurst, W., Holsboer, F., Clin. Neuropsychiatry 4, 234–241.
Reul, J.M., Linthorst, A.C., 2002. Corticotropin-releasing hormone receptor Pitts, A.F., Samuelson, S.D., Meller, W.H., Bissette, G., Nemeroff, C.B., Kathol,
type 1-deficiency enhances hippocampal serotonergic neurotransmission: R.G., 1995. Cerebrospinal fluid corticotropin-releasing hormone, vasopres-
an in vivo microdialysis study in mutant mice. Neuroscience 109, 253–266. sin, and oxytocin concentrations in treated patients with major depression
Pecoraro, N., Reyes, F., Gomez, F., Bhargava, A., Dallman, M.F., 2004. Chronic and controls. Biol. Psychiatry 38, 330–335.
stress promotes palatable feeding, which reduces signs of stress: feedfor- Plamondon, H., Merali, Z., 1994. Push-pull perfusion reveals meal-dependent
ward and feedback effects of chronic stress. Endocrinology 145, 3754– changes in the release of bombesin-like peptides in the rat paraventricular
3762. nucleus. Brain Res. 668, 54–61.
Peng, C.H., Chiou, S.H., Chen, S.J., Chou, Y.C., Ku, H.H., Cheng, C.K., Yen, Plamondon, H., Merali, Z., 1998. Anorectic action of bombesin requires
C.J., Tsai, T.H., Chang, Y.L., Kao, C.L., 2007. Neuroprotection by Imi- receptor for corticotropin-releasing factor but not oxytocin. Eur. J. Phar-
pramine against lipopolysaccharide-induced apoptosis in hippocampus- macol. 340, 99–109.
derived neural stem cells mediated by activation of BDNF and the MAPK Plamondon, H., Lambert, C., Merali, Z., 1998. Sustained bombesin exposure
pathway. Eur. Neuropsychopharmacol. 18, 128–140. results in receptor down-regulation and tolerance to the chronic but not
Peng, J., Mao, X.O., Stevenson, F.F., Hsu, M., Andersen, J.K., 2004. The acute effects of bombesin on ingestion. Brain Res. 782, 202–211.
herbicide paraquat induces dopaminergic nigral apoptosis through sustained Plotsky, P.M., 1991. Pathways to the secretion of adrencorticotropin: a view
activation of the JNK pathway. J. Biol. Chem. 279, 32626–32632. from the portal. J. Neuroendocinol. 3, 1–9.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 67

Pokrovskaja, K., Panaretakis, T., Grandér, D., 2005. Alternative signaling in the hypothalamic paraventricular nucleus of depressed patients. Neu-
pathways regulating type I interferon-induced apoptosis. J. Interferon roendocrinology 60, 436–444.
Cytokine Res. 25, 799–810. Raadsheer, F.C., van Heerikhuize, J.J., Lucassen, P.J., Hoogendijk, W.J.,
Post, R.M., 1992. Transduction of psychosocial stress into the neurobiology of Tilders, F.J., Swaab, D.F., 1995. Corticotropin-releasing hormone mRNA
recurrent affective disorder. Am. J. Psychiatry 149, 999–1010. levels in the paraventricular nucleus of patients with Alzheimer’s disease
Pournajafi Nazarloo, H., Takao, T., Taguchi, T., Ito, H., Hashimoto, K., 2003. and depression. Am. J. Psychiatry 152, 1372–1376.
Modulation of type I IL-1 receptor and IL-1 beta mRNA expression Raber, J., Koob, G.F., Bloom, F.E., 1997. Interferon-alpha and transforming
followed by endotoxin treatment in the corticotropin-releasing hormone- growth factor-beta 1 regulate corticotropin-releasing factor release from the
deficient mouse. J. Neuroimmunol. 140, 102–108. amygdala: comparison with the hypothalamic response. Neurochem. Int.
Poynter, M.E., Cloots, R., van Woerkom, T., Butnor, K.J., Vacek, P., Taatjes, 30, 455–463.
D.J., Irvin, C.G., Janssen-Heininger, Y.M., 2004. NF-kappa B activation in Rabin, B.S., Pezzone, M.A., Kusnecov, A., Hoffman, G.E., 1995. Identification
airways modulates allergic inflammation but not hyperresponsiveness. J. of stressor-activated areas in the central nervous system. Methods Neurosci.
Immunol. 173, 7003–7009. 24, 185–193.
Pozzoli, G., Bilezikjian, L.M., Perrin, M.H., Blount, A.L., Vale, W.W., 1996. Raison, C.L., Demetrashvili, M., Capuron, L., Miller, A.H., 2005a. Neurop-
Corticotropin-releasing factor (CRF) and glucocorticoids modulate the sychiatric adverse effects of interferon-alpha: recognition and management.
expression of type 1 CRF receptor messenger ribonucleic acid in rat anterior CNS Drugs 19, 105–123.
pituitary cell cultures. Endocrinology 137, 65–71. Raison, C.L., Schafer, A., Faller, H., Csef, H., Scheurlen, A.H., 2005b.
Pradillo, J.M., Romera, C., Hurtado, O., Cardenas, A., Moro, M.A., Leza, J.C., Neuropsychiatric adverse effects of interferon-alpha: recognition and man-
Davalos, A., Castillo, J., Lorenzo, P., Lizasoain, I., 2005. TNFR1 upregula- agement. CNS Drugs 19, 105–123.
tion mediates tolerance after brain ischemic preconditioning. J. Cereb. Raison, C.L., Capuron, L., Miller, A.H., 2006. Cytokines sing the blues:
Blood Flow Metab. 25, 193–203. inflammation and the pathogenesis of depression. Trends Immunol. 27,
Pratt, L.A., Ford, D.E., Crum, R.M., Armenian, H.K., Gallo, J.J., Eaton, W.W., 24–31.
1996. Depression, psychotropic medication, and risk of myocardial infarc- Rajkowska, G., 2000a. Histopathology of the prefrontal cortex in major
tion. Prospective data from the Baltimore ECA follow-up. Circulation 94, depression: what does it tell us about dysfunctional monoaminergic
3123–3129. circuits? Prog. Brain Res. 126, 397–412.
Preil, J., Müller, M.B., Gesing, A., Reul, J.M., Sillaber, I., van Gaalen, M.M., Rajkowska, G., 2000b. Postmortem studies in mood disorders indicate altered
Landgrebe, J., Holsboer, F., Stenzel-Poore, M., Wurst, W., 2001. Regulation numbers of neurons and glial cells. Biol. Psychiatry 48, 766–777.
of the hypothalamic–pituitary–adrenocortical system in mice deficient for Rajora, N., Boccoli, G., Burns, D., Sharma, S., Catania, A.P., Lipton, J.M.,
CRH receptors 1 and 2. Endocrinology 142, 4946–4955. 1997. a-MSH modulates local and circulating tumor necrosis factor-a in
Price, M.L., Lucki, I., 2001. Regulation of serotonin release in the lateral experimental brain inflammation. J. Neurosci. 15, 2181–2186.
septum and striatum by corticotropin-releasing factor. J. Neurosci. 21, Ramasubbu, S., 2000. Psychological model of post-stroke major depression. Br.
2833–2841. J. Psychiatry 176, 294–296.
Prinz, M., Hanisch, U.K., 1999. Murine microglial cells produce and respond to Ransjo, M., Marklund, M., Persson, M., Lerner, U.H., 1998. Synergistic
interleukin-18. J. Neurochem. 72, 2215–2218. interactions of bradykinin, thrombin, interleukin 1 and tumor necrosis
Puglisi-Allegra, S., Imperato, A., Angelucci, L., Cabib, S., 1991. Acute stress factor on prostanoid biosynthesis in human periodontal-ligament cells.
induces time-dependent responses in dopamine mesolimbic system. Brain Arch. Oral Biol. 43, 253–260.
Res. 554, 217–222. Rasmusson, A.M., Shi, L., Duman, R., 2002. Downregulation of BDNF mRNA
Purba, J.S., Hoogendijk, W.J., Hofman, M.A., Swaab, D.F., 1996. Increased in the hippocampal dentate gyrus after re-exposure to cues previously
number of vasopressin- and oxytocin-expressing neurons in the paraven- associated with footshock. Neuropsychopharmacology 27, 133–142.
tricular nucleus of the hypothalamus in depression. Arch. Gen. Psychiatry Ravindran, A.V., Anisman, H., Merali, Z., Charbonneau, Y., Telner, J., Bialik,
53, 137–143. R.J., Wiens, A., Ellis, J., Griffiths, J., 1999. Treatment of primary dysthymia
Purselle, D.C., Nemeroff, C.B., 2003. Serotonin transporter: a potential with cognitive therapy and pharmacotherapy: clinical symptoms and func-
substrate in the biology of suicide. Neuropsychopharmacology 28, tional impairments. Am. J. Psychiatry 156, 1608–1617.
613–619. Reagan, L.P., Hendry, R.M., Reznikov, L.R., Piroli, G.G., Wood, G.E., McE-
Qi, X., Lin, W., Li, J., Pan, Y., Wang, W., 2006. The depressive-like behaviors wen, B.S., Grillo, C.A., 2007. Tianeptine increases brain-derived neuro-
are correlated with decreased phosphorylation of mitogen-activated protein trophic factor expression in the rat amygdala. Eur. J. Pharmacol. 565, 68–75.
kinases in rat brain following chronic forced swim stress. Behav. Brain Res. Reddy, D.S., 2003. Is there a physiological role for the neurosteroid THDOC in
175, 233–240. stress-sensitive conditions? Trends Pharmacol. Sci. 24, 103–106.
Qian, L., Block, M.L., Wei, S.J., Lin, C.F., Reece, J., Pang, H., Wilson, B., Reichenberg, A., Yirmiya, R., Schuld, A., Kraus, T., Haack, M., Morag, A.,
Hong, J.S., Flood, P.M., 2006. Interleukin-10 protects lipopolysaccharide- Pollmacher, T., 2001. Cytokine-associated emotional and cognitive distur-
induced neurotoxicity in primary midbrain cultures by inhibiting the bances in humans. Arch. Gen. Psychiatry 58, 445–452.
function of NADPH oxidase. J. Pharmacol. Exp. Ther. 319, 44–52. Ren-Patterson, R.F., Cochrane, L.W., Holmes, A., Lesch, K.P., Lu, B., Murphy,
Quan, N., Herkenham, M., 2002. Connecting cytokines and brain: a review of D.L., 2006. Gender-dependent modulation of brain monoamines and anxi-
current issues. Histol. Histopathol. 17, 273–288. ety-like behaviors in mice with genetic serotonin transporter and BDNF
Quan, N., Avitsur, R., Stark, J.L., He, L., Lai, W., Dhabhar, F., Sheridan, J.F., deficiencies. Cell Mol. Neurobiol. 26, 755–780.
2003. Molecular mechanisms of glucocorticoid resistance in splenocytes of Reul, M.H.M., Holsboer, F., 2002. Corticotropin-releasing factor receptors 1
socially stressed male mice. J. Neuroimmunol. 137, 51–58. and 2 in anxiety and depression. Curr. Opin. Pharmacol. 2, 23–33.
Quitkin, F.M., Stewart, J.W., McGrath, P.J., Liebowitz, M.R., Harrison, W.M., Reynolds, J.L., Ignatowski, T.A., Sud, R., Spengler, R.N., 2004. Brain-derived
Tricamo, E., Klein, D.F., Rabkin, J.J., Markowitz, J.S., Wager, S.G., 1988. tumor necrosis factor-alpha and its involvement in noradrenergic neuron
Phenelzine vs. imipramine in the treatment of probable atypical depression: functioning involved in the mechanism of action of an antidepressant. J.
defining syndrome boundaries of selective MAOI responders. Am. J. Pharmacol. Exp. Ther. 310, 1216–1225.
Psychiatry 145, 306–311. Reynolds, J.L., Ignatowski, T.A., Sud, R., Spengler, R.N., 2005. An antide-
Quitkin, F.M., Harrison, W.M., Stewart, J.W., McGrath, P.J., Tricamo, E., pressant mechanism of desipramine is to decrease tumor necrosis factor-
Ocepek-Welikson, K., Rabkin, J.J., Wager, S.G., Nunes, E., Klein, D.F., alpha production culminating in increases in noradrenergic neurotransmis-
1991. Response to phenelzine and imipramine in placebo responders with sion. Neuroscience 133, 519–531.
atypical depression. Arch. Gen. Psychiatry 48, 319–323. Rigby, W.F., 2007. Drug insight: different mechanisms of action of tumor
Raadsheer, F.C., Hoogendijk, W.J., Stam, F.C., Tilders, F.J., Swaab, D.F., 1994. necrosis factor antagonists-passive-aggressive behavior? Nat. Clin. Pract.
Increased numbers of corticotropin-releasing hormone expressing neurons Rheumatol. 3, 227–233.
68 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Riley, J.K., Takeda, K., Akira, S., Schreiber, R.D., 1999. Interleukin-10 receptor inflammatory response during ischemic neuroprotection in mice. Stroke
signaling through the JAK-STAT pathway. Requirement for two distinct 35, 2576–2581.
receptor-derived signals for anti-inflammatory action. J. Biol. Chem. 274, Rosell, D.R., Nacher, J., Akama, K.T., McEwen, B.S., 2003. Spatiotemporal
16513–16521. distribution of gp130 cytokines and their receptors after status epilepticus:
Ringheim, G.E., Conant, K., 2004. Neurodegenerative disease and the neu- comparison with neuronal degeneration and microglial activation. Neu-
roimmune axis (Alzheimer’s and Parkinson’s disease, and viral infections). roscience 122, 29–48.
J. Neuroimmunol. 147, 43–49. Rossi, C., Angelucci, A., Costantin, L., Braschi, C., Mazzantini, M., Babbini, F.,
Ringheim, G.E., Burgher, K.L., Heroux, J.A., 1995. Interleukin-6 mRNA Fabbri, M.E., Tessarollo, L., Maffei, L., Berardi, N., Caleo, M., 2006. Brain-
expression by cortical neurons in culture: evidence for neuronal sources derived neurotrophic factor (BDNF) is required for the enhancement of
of interleukin-6 production in the brain. J. Neuroimmunol. 63, 113–123. hippocampal neurogenesis following environmental enrichment. Eur. J.
Ritter, S., Pelzer, N.L., 1978. Magnitude of stress induced norepinephrine Neurosci. 24, 1850–1856.
depletion varies with age. Brain Res. 152, 1701–1705. Roth, R.H., Tam, S.-Y., Ida, Y., Yang, J.-X., Deutch, Y., 1988. Stress and
Rivest, S., 2001. How circulating cytokines trigger the neural circuits that mesocorticolimbic dopamine system. Ann. NY Acad. Sci. 537, 138–147.
control the hypothalamic–pituitary–adrenal axis. Psychoneuroendocrinol- Rothe, M., Wong, S.C., Henzel, W.J., Goeddel, D.V., 1994. A novel family of
ogy 26, 761–788. putative signal transducers associated with the cytoplasmic domain of the
Rivest, S., Lacroix, S., Vallieres, L., Nadeau, S., Zhang, J., Laflamme, N., 2000. 75 kDa tumor necrosis factor receptor. Cell 78, 681–692.
How the blood talks to the brain parenchyma and the paraventricular Rothwell, N.J., 1999. Cytokines—killers in the brain? J. Physiol. 514, 3–17.
nucleus of the hypothalamus during systemic inflammatory and infectious Rothwell, N.J., Hopkins, S.J., 1995. Cytokines and the nervous system. II.
stimuli. Proc. Soc. Exp. Biol. Med. 223, 22–38. Actions and mechanisms of action. Trends Neurosci. 18, 130–136.
Rivier, C., Vale, W., 1985. Effects of corticotropin-releasing factor, neurohy- Roy, A., Pickar, D., Paul, S., Doran, A., Chrousos, G.P., Gold, P.W., 1987. CSF
pophyseal peptides and catecholamines on pituitary function. Fed. Proc. 44, corticotropin-releasing hormone in depressed patients and normal control
189–195. subjects. Am. J. Psychiatry 144, 641–645.
Roach, S.K., Lee, S.B., Schorey, J.S., 2005. Differential activation of the Ruggiero, D.A., Underwood, M.D., Rice, P.M., Mann, J.J., Arango, V., 1991.
transcription factor cyclic AMP response element binding protein (CREB) Corticotropic-releasing hormone and serotonin interact in the human brain-
in macrophages following infection with pathogenic and nonpathogenic stem: behavioral implications. Neuroscience 1343–1354.
mycobacteria and role for CREB in tumor necrosis factor alpha production. Saarelainen, T., Hendolin, P., Lucas, G., Koponen, E., Sairanen, M., MacDo-
Infect. Immun. 73, 14–22. nald, E., Agerman, K., Haapasalo, A., Nawa, H., Aloyz, R., Ernfors, P.,
Robinson, T.E., 1988. Stimulant drugs and stress: factors influencing individual Castren, E., 2003. Activation of the TrkB neurotrophin receptor is induced
differences in the susceptibility to sensitization. In: Kalivas, P.W., Barnes, by antidepressant drugs and is required for antidepressant-induced beha-
C.D. (Eds.), Sensitization in the Nervous System. Telford Press, Caldwell, vioral effects. J. Neurosci. 23, 349–357.
NJ, pp. 145–173. Saija, A., Princi, P., Lanza, M., Scalese, M., Aramnejad, E., De Sarro, A., 1995.
Robinson, T.E., Angus, A.L., Becker, J.B., 1987. Sensitization to stress: The Systemic cytokine administration can affect blood–brain barrier perme-
enduring effects of prior stress on amphetamine-induced rotational beha- ability in the rat. Life Sci. 56, 775–784.
vior. Neuropharamacology 26, 679–691. Sairanen, T.R., Lindsberg, P.J., Brenner, M., Carpen, O., Siren, A., 2001.
Roe, S.Y., McGowan, E.M., Rothwell, N.J., 1998. Evidence for the involvement Differential cellular expression of tumor necrosis factor-alpha and Type
of corticotropin-releasing hormone in the pathogenesis of traumatic brain I tumor necrosis factor receptor after transient global forebrain ischemia. J.
injury. Eur. J. Neurosci. 10, 553–559. Neurol. Sci. 186, 87–99.
Roesler, R., Henriques, J.A., Schwartsmann, G., 2006. Gastrin-releasing pep- Sairanen, M., O’Leary, O.F., Knuuttila, J.E., Castrén, E., 2007. Chronic
tide receptor as a molecular target for psychiatric and neurological dis- antidepressant treatment selectively increases expression of plasticity-
orders. CNS Neurol. Disord. Drug Targets 5, 197–204. related proteins in the hippocampus and medial prefrontal cortex of the
Rogers, R.J., Monnier, J.M., Nick, H.S., 2001. Tumor necrosis factor-alpha rat. Neuroscience 144, 368–374.
selectively induces MnSOD expression via mitochondria-to-nucleus signal- Sammut, S., Goodall, G., Muscat, R., 2001. Acute interferon-alpha adminis-
ing, whereas interleukin-1beta utilizes an alternative pathway. J. Biol. tration modulates sucrose consumption in the rat. Psychoneuroendocrinol-
Chem. 276, 20419–20427. ogy 26, 261–272.
Rogóz, Z., Skuza, G., Legutko, B., 2007. Repeated co-treatment with imipra- Sammut, S., Bethus, I., Goodall, G., Muscat, R., 2002. Antidepressant reversal
mine and amantadine induces hippocampal brain-derived neurotrophic of interferon-alpha-induced anhedonia. Physiol. Behav. 75, 765–772.
factor gene expression in rats. J. Physiol. Pharmacol. J. 58, 219–234. Santarelli, L., Saxe, M., Gross, C., Surget, A., Battaglia, F., Dulawa, S.,
Rohatiner, A.Z., Balkwill, F., Lister, T.A., 1983. Interferon in acute myelo- Weisstaub, N., Lee, J., Duman, R., Arancio, O., Belzung, C., Hen, R.,
genous leukaemia: a preliminary report. Haematol. Blood Transfus. 28, 2003. Requirement of hippocampal neurogenesis for the behavioral effects
56–58. of antidepressants. Science 301, 805–809.
Rosel, P., Arranz, B., Vallejo, J., Oros, M., Menchon, J.M., Alvarez, P., Navarro, Saphier, D., 1995. Neuroendocrine effects of interferon-alpha in the rat. Adv.
M.A., 1997. High affinity [3H]imipramine and [3H]paroxetine binding sites Exp. Med. Biol. 373, 209–218.
in suicide brains. J. Neural Transm. 104, 921–929. Sapolsky, R.M., 2000. The possibility of neurotoxicity in the hippocampus in
Rosel, P., Arranz, B., Vallejo, J., Oros, M., Menchon, J.M., Alvarez, P., Navarro, major depression: a primer on neuron death. Biol. Psychiatry 48, 755–765.
M.A., 1998. Variations in [3H]imipramine and 5-HT2A but not [3H]par- Sapolsky, R.M., Krey, L.C., McEwen, B.S., 1985. Prolonged glucocorticoid
oxetine binding sites in suicide brains. Psychiatry Res. 82, 161–170. exposure reduces hippocampal neuron number: implications for aging. J.
Rosel, P., Arranz, B., San, L., Vallejo, J., Crespo, J.M., Urretavizcaya, M., Neurosci. 5, 1222–1227.
Navarro, M.A., 2000. Altered 5-HT(2A) binding sites and second messen- Sapolsky, R.M., Uno, H., Rebert, C.S., Finch, C.E., 1990. Hippocampal damage
ger inositol trisphosphate (IP(3)) levels in hippocampus but not in frontal associated with prolonged glucocorticoid exposure in primates. J. Neurosci.
cortex from depressed suicide victims. Psychiatry Res. 99, 173–181. 10, 2897–2902.
Rosel, P., Arranz, B., Urretavizcaya, M., Oros, M., San, L., Navarro, M.A., Sapolsky, R.M., Romero, L.M., Munck, A.U., 2000. How do glucocorticoids
2004. Altered 5-HT2A and 5-HT4 postsynaptic receptors and their intra- influence stress responses? Integrating permissive, suppressive, stimulatory,
cellular signalling systems IP3 and cAMP in brains from depressed violent and preparative actions. Endocr. Rev. 21, 55–89.
suicide victims. Neuropsychobiology 49, 189–195. Saporito, M.S., Hudkins, R.L., Maroney, A.C., 2002. Discovery of CEP-1347/
Rosenthal, M.H., 2003. The challenge of comorbid disorders in patients with KT-7515, an inhibitor of the JNK/SAPK pathway for the treatment of
depression. J. Am. Osteopath. Assoc. 103 (8 Suppl 4), S10–S15. neurodegenerative diseases. Prog. Med. Chem. 40, 23–62.
Rosenzweig, H.L., Lessov, N.S., Henshall, D.C., Minami, M., Simon, R.P., Sargent, P.A., Kjaer, K.H., Bench, C.J., Rabiner, E.A., Messa, C., Meyer, J.,
Stenzel-Poore, M.P., 2004. Endotoxin preconditioning prevents cellular Gunn, R.N., Grasby, P.M., Cowen, P.J., 2000. Brain serotonin1A receptor
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 69

binding measured by positron emission tomography with [11C]WAY- proliferation in rat hippocampus after transient middle cerebral artery
100635: effects of depression and antidepressant treatment. Arch. Gen. occlusion. Neurosci. Lett. 418, 248–252.
Psychiatry 57, 174–180. Sequeira, A., Turecki, G., 2006. Genome wide gene expression studies in mood
Sato, S., Reiner, S.L., Jensen, M.A., Roos, R.P., 1997. Central nervous system disorders. OMICS 10, 444–454.
mRNA expression following Theiler’s murine encephalomyelitis virus Serretti, A., Benedetti, F., Zanardi, R., Smeraldi, E., 2005. The influence of
infection. J. Neuroimmunol. 76, 213–223. Serotonin Transporter Promoter Polymorphism (SERTPR) and other poly-
Saud, K., Herrera-Molina, R., Von Bernhardi, R., 2005. Pro- and anti-inflam- morphisms of the serotonin pathway on the efficacy of antidepressant
matory cytokines regulate the ERK pathway: implication of the timing for treatments. Prog. Neuropsychopharmacol. Biol. Psychiatry 29, 1074–1084.
the activation of microglial cells. Neurotox. Res. 8, 277–287. Shanks, N., Griffiths, J., Anisman, H., 1994. Norepinephrine and serotonin
Sawada, M., Imamura, K., Nagatsu, T., 2006. Role of cytokines in inflammatory alterations following chronic stressor exposure: mouse strain differences.
process in Parkinson’s disease. J. Neural Transm. (Suppl. 70), 373–381. Pharmacol. Biochem. Behav. 49, 57–65.
Sawchenko, P.E., 1987. Adrenalectomy-induced enhancement of CRF and Shanks, N., Griffiths, J., Zalcman, S., Zacharko, R.M., Anisman, H., 1991.
vasopressin immunoreactivity in parvocellular neurosecretory neurons: Alterations of central norepinephrine, dopamine and serotonin in several
anatomic, peptide, and steroid specificity. J. Neurosci. 7, 1093–1106. strains of mice following acute stressor exposure. Pharmacol. Biochem.
Sawchenko, P.E., Brown, E.R., Chan, R.K.W., Ericsson, A., Li, H.-Y., Roland, Behav. 38, 69–75.
B.L., Kovacs, K.J., 1996. The paraventricular nucleus of the hypothalamus Shanks, N., Windle, R.J., Perks, P.A., Harbuz, M.S., Jessop, D.S., Ingram, C.D.,
and the functional neuroanatomy of visceromotor responses to stress. Prog. Lightman, S.L., 2000. Early-life exposure to endotoxin alters hypothala-
Brain Res. 107, 201–222. mic–pituitary–adrenal function and predisposition to inflammation. Proc.
Scalori, A., Pozzi, M., Bellia, V., Apale, P., Santamaria, G., Bordoni, T., Natl. Acad. Sci. U.S.A. 97, 5645–5650.
Redaelli, A., Avolio, A., Parravicini, P., Piotelli, P., Roffi, L., 2005. Sheline, Y.I., Gado, M.H., Kraemer, H.C., 2003. Untreated depression and
Interferon-induced depression: prevalence and management. Dig. Liver hippocampal volume loss. Am. J. Psychiatry 160, 1516–1518.
Dis. 37, 102–107. Sheline, Y.I., Mintun, M.A., Barch, D.M., Wilkins, C., Snyder, A.Z., Moerlein,
Schabitz, W.R., Kollmar, R., Schwaninger, M., Juettler, E., Bardutzky, J., S.M., 2004. Decreased hippocampal 5-HT(2A) receptor binding in older
Scholzke, N., Sommer, C., Schwab, S., 2003. Neuroprotective effect of depressed patients using [18F]altanserin positron emission tomography.
granulocyte colony-stimulating factor after focal cerebral ischemia. Stroke Neuropsychopharmacology 29, 2235–2241.
34, 745–751. Shen, C.P., Tsimberg, Y., Salvadore, C., Meller, E., 2004. Activation of Erk and
Schaefer, M., Schwaiger, M., Pich, M., Lieb, K., Heinz, A., 2003. Neurotrans- JNK MAPK pathways by acute swim stress in rat brain regions. BMC
mitter changes by interferon-alpha and therapeutic implications. Pharma- Neurosci. 5, 36.
copsychiatry 36, S203–S206. Sheng, J.G., Mrak, R.E., Griffin, W.S., 1997. Glial-neuronal interactions in
Schaefer, M., Schwaiger, M., Garkisch, A.S., Pich, M., Hinzpeter, A., Uebel- Alzheimer disease: progressive association of IL-1alpha+ microglia and
hack, R., 2005. Prevention of interferon-alpha associated depression in S100beta+ astrocytes with neurofibrillary tangle stages. J. Neuropathol.
psychiatric risk patients with chronic hepatitis C. J. Hepatol. 42, 793–798. Exp. Neurol. 56, 285–290.
Sceibel, R.S., Valentine, A.D., O’Brien, S., Meyers, C.A., 2004. Cognitive Shepard, J.D., Liu, Y., Sassone-Corsi, P., Aguilera, G., 2005. Role of gluco-
dysfunction and depression during treatment with interferon-alpha and corticoids and cAMP-mediated repression in limiting corticotropin-releas-
chemotherapy. J. Neuropsychiatr. Clin. Neurosci. 16, 185–191. ing hormone transcription during stress. J. Neurosci. 25, 4073–4081.
Schmauss, C., 2003. Serotonin 2C receptors: suicide, serotonin, and runaway Sherif, F., Marcusson, J., Oreland, L., 1991. Brain gamma-aminobutyrate
RNA editing. Neuroscientist 9, 237–242. transaminase and monoamine oxidase activities in suicide victims. Eur.
Schmidt, E.D., Janszen, A.W.J.W., Wouterlood, F.G., Tilders, F.J.H., 1995. Arch. Psychiatry Clin. Neurosci. 241, 139–144.
Interleukin-1 induced long-lasting changes in hypothalamic corticotropin- Shiah, I.S., Yatham, L.N., 1998. GABA function in mood disorders: an update
releasing hormone (CRH) neurons and hyperresponsiveness of the hypotha- and critical review. Life Sci. 63, 1289–1303.
lamic–pituitary–adrenal axis. J. Neurosci. 15, 7417–7426. Shimizu, E., Hashimoto, K., Okamura, N., Koike, K., Komatsu, N., Kumakiri,
Schmidt, E.D., Binnekade, R., Janszen, A.W., Tlders, F.J., 1996. Short stressor C., Nakazato, M., Watanabe, H., Shinoda, N., Okada, S., Iyo, M., 2003.
induced long-lasting increases of vasopressin stores in hypothalamic corti- Alterations of serum levels of brain-derived neurotrophic factor (BDNF)
cotropin-releasing hormone (CRH) neurons in adult rats. J. Neuroendocri- in depressed patients with or without antidepressants. Biol. Psychiatry 54,
nol. 8, 703–712. 70–75.
Schmieder, R.E., Hilgers, K.F., Schlaich, M.P., Schmidt, B.M., 2007. Renin– Shimizu, H., Ohtani, K., Sato, N., Nagamine, T., Mori, M., 1995. Increase in
angiotensin system and cardiovascular risk. Lancet 369, 1208–1219. serum interleukin-6, plasma ACTH and serum cortisol levels after systemic
Schmitt, A., Benninghoff, J., Moessner, R., Rizzi, M., Paizanis, E., Doenitz, C., interferon-alpha administration. Endocrinol. J. 42, 551–556.
Gross, S., Hermann, M., Gritti, A., Lanfumey, L., Fritzen, S., Reif, A., Shimizu, N., Nakane, H., Hori, T., Hayashi, Y., 1994. CRF receptor antagonist
Hamon, M., Murphy, D.L., Vescovi, A., Lesch, K.P., 2007. Adult neurogen- attenuates stress-induced noradrenaline release in the medial prefrontal
esis in serotonin transporter deficient mice. J. Neural. Transm. 4, 1107–1119. cortex of rats. Brain Res. 654, 145–148.
Schneider, A., Kuhn, H.G., Schäbitz, W.R., 2005. A role for G-CSF (granu- Shintani, F., Kanba, S., Nakaki, T., Nibuya, M., Kinoshita, N., Suzuki, E., Yagi,
locyte-colony stimulating factor) in the central nervous system. Cell Cycle G., Kato, R., Asai, M., 1993. Interleukin-1b augments release of norepi-
4, 1753–1757. nephrine, dopamine and serotonin in the rat anterior hypothalamus. J.
Schobitz, B., De Kloet, E.R., Holsboer, F., 1994. Gene expression and function Neurosci. 13, 3574–3581.
of interleukin 1, interleukin 6, and tumor necrosis factor in the brain. Prog. Shintani, F., Nakaki, T., Kanba, S., Sato, K., Yagi, G., Shiozawa, M., Aiso, S.,
Neurobiol. 44, 397–432. Kato, R., Asai, M., 1995. Involvement of interleukin-1 in immobilization
Schramm, T.M., Lawford, B.R., Macdonald, G.A., Cooksley, W.G., 2000. stress-induced increase in plasma adrenocorticotropic hormone and in
Sertraline treatment of interferon-alfa-induced depressive disorder. Med. release of hypothalamic monoamines in the rat. J. Neurosci. 15,
J. Aust. 173, 359–361. 1961–1970.
Schulkin, J., 2006. Angst and the amygdala. Dialogues Clin. Neurosci. 8, 407– Shirayama, Y., Chen, A.C., Nakagawa, S., Russell, D.S., Duman, R.S., 2002.
416. Brain-derived neurotrophic factor produces antidepressant effects in beha-
Schulkin, J., Gold, P.W., McEwen, B.S., 1998. Induction of corticotropin- vioral models of depression. J. Neurosci. 22, 3251–3261.
releasing hormone gene expression by glucocorticoids: implication for Shumyatsky, G.P., Tsvetkov, E., Malleret, G., Vronskaya, S., Hatton, M.,
understanding the states of fear and anxiety and allostatic load. Psycho- Hampton, L., Battey, J.F., Dulac, C., Kandel, E.R., Bolshakov, V.Y.,
neuroendocrinology 23, 219–243. 2002. Identification of a signaling network in lateral nucleus of amygdala
Sehara, Y., Hayashi, T., Deguchi, K., Zhang, H., Tsuchiya, A., Yamashita, T., important for inhibiting memory specifically related to learned fear. Cell
Lukic, V., Nagai, M., Kamiya, T., Abe, K., 2007. G-CSF enhances stem cell 111, 905–918.
70 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Shuto, H., Kataoka, Y., Horikawa, T., Fujihara, N., Oishi, R., 1997. Repeated Spalletta, G., Bossù, P., Ciaramella, A., Bria, P., Caltagirone, C., Robinson,
interferon-alpha administration inhibits dopaminergic neural activity in the R.G., 2006. The etiology of poststroke depression: a review of the literature
mouse brain. Brain Res. 747, 348–351. and a new hypothesis involving inflammatory cytokines. Mol. Psychiatry
Sibille, E., Pavlides, C., Benke, D., Toth, M., 2000. Genetic inactivation of the 11, 984–991.
serotonin(1A) receptor in mice results in downregulation of major Spencer, S.J., Heida, J.G., Pittman, Q.J., 2005. Early life immune challenge-
GABA(A) receptor alpha subunits, reduction of GABA(A) receptor bind- effects on behavioural indices of adult rat fear and anxiety. Behav. Brain
ing, and benzodiazepine-resistant anxiety. J. Neurosci. 20, 2758–2765. Res. 164, 231–238.
Sibille, E., Arango, V., Galfalvy, H.C., Pavlidis, P., Erraji-Benchekroun, L., Spencer, S.J., Auer, R.N., Pittman, Q.J., 2006. Rat neonatal immune challenge
Ellis, S.P., Mann, J., 2004. Gene expression profiling of depression alters adult responses to cerebral ischaemia. J. Cereb. Blood Flow Metab.
and suicide in human prefrontal cortex. Neuropsychopharmacology 29, 26, 456–467.
351–361. Sriram, K., Matheson, J.M., Benkovic, S.A., Miller, D.B., Luster, M.I., O’Call-
Silverman, E.S., Breault, D.T., Vallone, J., Subramanian, S., Yilmaz, A.D., aghan, J.P., 2002. Mice deficient in TNF receptors are protected against
Mathew, S., Subramaniam, V., Tantisira, K., Pacák, K., Weiss, S.T., Maj- dopaminergic neurotoxicity: implications for Parkinson’s disease. FASEB
zoub, J.A., 2004. Corticotropin-releasing hormone deficiency increases 16, 1474–1476.
allergen-induced airway inflammation in a mouse model of asthma. J. Sriram, K., Matheson, J.M., Benkovic, S.A., Miller, D.B., Luster, M.I., O’Call-
Allergy Clin. Immunol. 114, 747–754. aghan, J.P., 2006. Deficiency of TNF receptors suppresses microglial
Simard, A.R., Soulet, D., Gowing, G., Julien, J.P., Rivest, S., 2006. Bone activation and alters the susceptibility of brain regions to MPTP-induced
marrow-derived microglia play a critical role in restricting senile plaque neurotoxicity: role of TNF-a. FASEB J. 20, 670–682.
formation in Alzheimer’s disease. Neuron 49, 489–502. Stamatovic, S.M., Dimitrijevic, O.B., Keep, R.F., Andjelkovic, A.V., 2006.
Singh, L.K., Pang, X., Alexacos, N., Letourneau, R., Theoharides, T.C., 1999. Inflammation and brain edema: new insights into the role of chemokines and
Acute immobilization stress triggers skin mast cell degranulation via their receptors. Acta Neurochir. Suppl. 96, 444–450.
corticotropin releasing hormone, neurotensin, and substance P: a link to Stanford, S.C., 1995. Central noradrenergic neurones and stress. Pharmacol.
neurogenic skin disorders. Brain Behav. Immun. 13, 225–239. Ther. 68, 242–297.
Skelton, K.H., Nemeroff, C.B., Knight, D.L., Owens, M.J., 2000. Chronic Stanley, M., Mann, J.J., 1983. Increased serotonin-2 binding sites in frontal
administration of the triazolobenzodiazepine alprazolam produces opposite cortex of suicide victims. Lancet 1, 214–216.
effects on corticotropin-releasing factor and urocortin neuronal systems. J. Stanton, A.L., Danoff-Burg, S., Cameron, C.L., Ellis, A.P., 1994. Coping
Neurosci. 20, 1240–1248. through emotional approach: problems of conceptualization and confound-
Sluzewska, A., Rybakowski, J.K., Laciak, M., Mackiewicz, A., Sobieska, M., ing. J. Pers. Soc. Psychol. 66, 350–362.
Wiktorowicz, K., 1995. Interleukin-6 serum levels in depressed patients Stark, G.R., Kerr, I.M., Williams, B.R., Silverman, R.H., Schreiber, R.D., 1998.
before and after treatment with fluoxetine. Ann. NY Acad. Sci. 762, 474– How cells respond to interferons. Annu. Rev. Biochem. 67, 227–264.
476. Starkman, M.N., Giordani, B., Gebarski, S.S., Schteingart, D.E., 2007.
Smedley, H., Katrak, M., Sikora, K., Wheeler, T., 1983. Neurological effects of Improvement in mood and ideation associated with increase in right caudate
recombinant human interferon. Br. Med. J. 286, 262–264. volume. J. Affect. Disord. 101, 139–147.
Smith, A., Tyrrell, D., Coyle, K., Higgins, P., 1988. Effects of interferon alpha Starkstein, S.E., Mizrahi, R., 2006. Depression in Alzheimer’s disease. Expert
on performance in man: a preliminary report. Psychopharmacology 96, Rev. Neurother. 6, 887–895.
414–416. Starkstein, S.E., Jorge, R., Petracca, G., Robinson, R.G., 2007. The construct of
Smith, A.D., Zigmond, M.J., 2003. Can the brain be protected through generalized anxiety disorder in Alzheimer disease. Am. J. Geriatr. Psy-
exercise? Lessons from an animal model of parkinsonism. Exp. Neurol. chiatry 15, 42–49.
184, 31–39. Stein-Behrens, B., Mattson, M.P., Chang, I., Yeh, M., Sapolsky, R., 1994. Stress
Smith, A.D., Castro, S.L., Zigmond, M.J., 2002. Stress-induced Parkinson’s exacerbates neuron loss and cytoskeletal pathology in the hippocampus. J.
disease: a working hypothesis. Physiol. Behav. 77, 527–531. Neurosci. 14, 5373–5380.
Smith, G.W., Aubry, J.M., Dellu, F., Contarino, A., Bilezikjian, L.M., Gold, Steptoe, A., Kunz-Ebrecht, S.R., Owen, N., 2003. Lack of association between
L.H., Chen, R., Marchuk, Y., Hauser, C., Bentley, C.A., Sawchenko, P.E., depressive symptoms and markers of immune and vascular inflammation in
Koob, G.F., Vale, W., Lee, K.F., 1998. Corticotropin releasing factor middle-aged men and women. Psychol. Med. 33, 667–674.
receptor 1-deficient mice display decreased anxiety, impaired stress Stevenson, C.W., Sullivan, R.M., Gratton, A., 2003. Effects of basolateral
response, and aberrant neuroendocrine development. Neuron 20, 1093– amygdala dopamine depletion on the nucleus accumbens and medial
1102. prefrontal cortical dopamine responses to stress. Neuroscience 116, 285–
Smith, M.A., Banerjee, S., Gold, P.W., Glowa, J., 1992. Induction of c-fos 293.
mRNA in rat brain by conditioned and unconditioned stressors. Brain Res. Stewart, J.W., McGrath, P.J., Rabkin, J.G., Quikin, F.M., 1993. Atypical
578, 135–141. depression: a valid clinical entity? Psychiatr. Clin. N. Am. 16, 479–495.
Smith, M.A., Makino, S., Kvetnansky, R., Post, R.M., 1995. Stress and Stockmeier, C.A., 2003. Involvement of serotonin in depression: evidence from
glucocorticoids affect the expression of brain-derived neurotrophic factor postmortem and imaging studies of serotonin receptors and the serotonin
and neurotrophin-3 mRNAs in the hippocampus. J. Neurosci. 15, 1768– transporter. J. Psychiatry Res. 37, 357–373.
1777. Stockmeier, C.A., Dilley, G.E., Shapiro, L.A., Overholser, J.C., Thompson,
Smith, W.J., Stewart, J., Pfaus, J.G., 1997. Tail pinch induces Fos immunor- P.A., Meltzer, H.Y., 1997. Serotonin receptors in suicide victims with major
eactivity within several brain regions of the male rat brain: effects of age. depression. Neuropsychopharmacology 16, 162–173.
Physiol. Behav. 61, 717–723. Stockmeier, C.A., Shapiro, L.A., Dilley, G.E., Kolli, T.N., Friedman, L.,
Sobel, R.M., Lotkowski, S., Mandel, S., 2005. Update on depression in Rajkowska, G., 1998. Increase in serotonin-1A autoreceptors in the mid-
neurologic illness: stroke, epilepsy, and multiple sclerosis. Curr. Psychiatry brain of suicide victims with major depression-postmortem evidence for
Rep. 7, 396–403. decreased serotonin activity. J. Neurosci. 18, 7394–7401.
Solaroglu, I., Jadhav, V., Zhang, J.H., 2007. Neuroprotective effect of granu- Stoll, G., Jander, S., Schroeter, M., 2000. Cytokines in CNS disorders:
locyte-colony stimulating factor. Front. Biosci. 12, 712–724. neurotoxicity versus neuroprotection. J. Neural Transm. 59, 81–89.
Song, C., Merali, Z., Anisman, H., 1999. Variations of nucleus accumbens Stone, E.A., 1987. Central cyclic-AMP-linked noradrenergic receptors: New
dopamine and serotonin following systemic interleukin-1, interleukin-2 or findings on properties as related to the actions of stress. Neurosci. Biobehav.
interleukin-6 treatment. Neuroscience 88, 823–836. Rev. 11, 391–398.
Song, L., Che, W., Min-Wei, W., Murakami, Y., Matsumoto, K., 2006. Stout, S.C., Owens, M.J., Lindsey, K.P., Knight, D.L., Nemeroff, C.B., 2001.
Impairment of the spatial learning and memory induced by learned help- Effects of sodium valproate on corticotropin-releasing factor systems in rat
lessness and chronic mild stress. Pharmacol. Biochem. Behav. 83, 186–193. brain. Neuropsychopharmacology 24, 624–631.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 71

Stutzmann, G.E., McEwen, B.S., LeDoux, J.E., 1998. Serotonin modulation of Thakker, D.R., Natt, F., Husken, D., van der Putten, H., Maier, R., Hoyer, D.,
sensory inputs to the lateral amygdala: dependency on corticosterone. J. Cryan, J.F., 2005. siRNA-mediated knockdown of the serotonin transporter
Neurosci. 18, 529–538. in the adult mouse brain. Mol. Psychiatry 10, 782–789.
Sullivan, R.M., Gratton, A., 1998. Relationships between stress-induced Thase, M.E., Carpenter, L., Kupfer, D.J., Frank, E., 1991. Clinical significance
increases in medial prefrontal cortical dopamine band plasma corticoster- of reversed neurovegetative subtypes of recurrent major depression. Psy-
one levels in rats. Role of cerebral laterality. Neuroscience 83, 81–91. chopharmacol. Bull. 27, 17–22.
Sullivan, P.G., Bruce-Keller, A.J., Rabchevsky, A.G., Christakos, S., Clair, Theoharides, T.C., Konstantinidou, A.D., 2007. Corticotropin-releasing hor-
D.K., Mattson, M.P., Scheff, S.W., 1999. Exacerbation of damage and mone and the blood–brain-barrier. Front. Biosci. 12, 1615–1628.
altered NF-kappaB activation in mice lacking tumor necrosis factor recep- Thiruchelvam, M., Richfield, E.K., Baggs, R.B., Tank, A.W., Cory-Slechta,
tors after traumatic brain injury. J. Neurosci. 19, 6248–6256. D.A., 2000. The nigrostriatal dopaminergic system as a preferential target of
Suls, J., Fletcher, B., 1985. The relative efficacy of avoidant and nonavoidant repeated exposures to combined paraquat and maneb: implications for
coping strategies: a meta-analysis. Health Psychol. 4, 249–288. Parkinson’s disease. J. Neurosci. 20, 9207–9214.
Sunami, M., Nishikawa, T., Yorogi, A., Shimoda, M., 2000. Intravenous Tidey, J.W., Miczek, K.A., 1996. Social defeat stress selectively alters meso-
administration of levodopa ameliorated a refractory akathisia case induced corticolimbic dopamine release: an in vivo microdialysis study. Brain Res.
by interferon-alpha. Clin. Neuropharmacol. 23, 59–61. 721, 140–149.
Sundman, I., Allard, P., Eriksson, A., Marcusson, J., 1997. GABA uptake sites Tilders, F.J.H., Schmidt, E.D., 1998. Interleukin-1-induced plasticity of
in frontal cortex from suicide victims and in aging. Neuropsychobiology 35, hypothalamic CRH neurons and long-term stress hyperresponsiveness.
11–15. Ann. NY Acad. Sci. 840, 65–73.
Sundman-Eriksson, I., Allard, P., 2002. [(3)H]Tiagabine binding to Tilders, F.J.H., Schmidt, E.D., 1999. Cross-sensitization between immune and
GABA transporter-1 (GAT-1) in suicidal depression. J. Affect. Disord. non-immune stressors. A role in the etiology of depression? Adv. Exp. Med.
71, 29–33. Biol. 461, 179–197.
Svenningsson, P., Greengard, P., 2006. p11 (S100A10)—an inducible adaptor Tilders, F.J.H., Schmidt, E.D., De Goeij, D.C.E., 1993. Phenotypic plasticity of
protein that modulates neuronal functions. Curr. Opin. Pharmacol. 7, CRF neurons during stress. Ann. NY Acad. Sci. 697, 39–52.
27–32. Timpl, P., Spanagel, R., Sillaber, I., Kresse, A., Reul, J.M., Stalla, G.K.,
Svenningsson, P., Chergui, K., Rachleff, I., Flajolet, M., Zhang, X., El Yacoubi, Blanquet, V., Steckler, T., Holsboer, F., Wurst, W., 1998. Impaired stress
M., et al., 2006. Alterations in 5-HT1B receptor function by p11 in response and reduced anxiety in mice lacking a functional corticotropin-
depression-like states. Science 311, 77–80. releasing hormone receptor 1. Nat. Genet. 19, 162–166.
Swaab, D.F., 2004. Neuropeptides in hypothalamic neuronal disorders. Int. Rev. Tong, L., Balazs, R., Soiampornkul, R., Thangnipon, W., Cotman, C.W., 2007.
Cytol. 240, 305–375. Interleukin-1beta impairs brain derived neurotrophic factor-induced signal
Swaab, D.F., Bao, A.M., Lucassen, P.J., 2005. The stress system in the human transduction. Neurobiol. Aging, doi:10.1016/j.neurobiolaging.2007.02.027.
brain in depression and neurodegeneration. Ageing Res. Rev. 4, 141–194. Toth, M., 2003. 5-HT1A receptor knockout mouse as a genetic model of
Taga, T., 1996. Gp130, a shared signal transducing receptor component for anxiety. Eur. J. Pharmacol. 463, 177–184.
hematopoietic and neuropoietic cytokines. J. Neurochem. 67, 1–10. Tsai, S.J., 2007. Glatiramer acetate could be a potential antidepressant through
Takahashi, L.K., 2002. CRF(2) Receptors: an emerging role in anxiety. Drug its neuroprotective and anti-inflammatory effects. Med. Hypotheses 69,
News Perspect. 15, 97–101. 145–148.
Takahashi, M., Yamada, T., 1999. Viral etiology for Parkinson’s disease—a Tsatsanis, C., Dermitzaki, E., Venihaki, M., Chatzaki, E., Minas, V., Gravanis,
possible role of influenza A virus infection. Jpn. J. Infect. Dis. 52, 89–98. A., Margioris, A.N., 2007. The corticotropin-releasing factor (CRF) family
Takatsuna, H., Morita, S., Nagatsu, T., Sawada, M., Umezawa, K., 2005. of peptides as local modulators of adrenal function. Cell Mol. Life Sci. 64,
Inhibition of inflammatory cytokine secretion from mouse microglia cells 1638–1655.
by DHMEQ, an NF-kappaB inhibitor. Biomed. Pharmacother. 59, 318–322. Tsui, J.K., Calne, D.B., Wang, Y., Schulzer, M., Marion, S.A., 1999. Occupa-
Takeuchi, H., Jin, S., Wang, J., Zhang, G., Kawanokuchi, J., Kuno, R., Sonobe, tional risk factors in Parkinson’s disease. Can. J. Public Health 90, 334–337.
Y., Mizuno, T., Suzumura, A., 2006. Tumor necrosis factor-alpha induces Tuglu, C., Kara, S.H., Caliyurt, O., Vardar, E., Abay, E., 2003. Increased serum
neurotoxicity via glutamate release from hemichannels of activated micro- tumor necrosis factor-alpha levels and treatment response in major depres-
glia in an autocrine manner. J. Biol. Chem. 281, 21362–21368. sive disorder. Psychopharmacology 170, 429–433.
Tamashiro, K.L., Nguyen, M.M., Sakai, R.R., 2005. Social stress: from rodents Tunçel, N., Sener, E., Cerit, C., Karasu, U., Gürer, F., Sahintürk, V., Bayçu, C.,
to primates. Front. Neuroendocrinol. 26, 27–40. Ak, D., Filiz, Z., 2005. Brain mast cells and therapeutic potential of
Tan, H., Zhong, P., Yan, Z., 2004. Corticotropin-releasing factor and acute stress vasoactive intestinal peptide in a Parkinson’s disease model in rats: brain
prolongs serotonergic regulation of GABA transmission in prefrontal microdialysis, behavior, and microscopy. Peptides 26, 827–836.
cortical pyramidal neurons. J. Neurosci. 24, 5000–5008. Tunnicliff, G., Malatynska, E., 2003. Central GABAergic systems and depres-
Tancredi, V., 1992. Tumor necrosis factor alters synaptic transmission in rat sive illness. Neurochem. Res. 28, 965–976.
hippocampal slices. Neurosci. Lett. 146, 176–178. Turecki, G., Briere, R., Dewar, K., Antonetti, T., Lesage, A.D., Seguin, M.,
Tanji, J., Hoshi, E., 2001. Behavioral planning in the prefrontal cortex. Curr. et al., 1999. Prediction of level of serotonin 2A receptor binding by
Opin. Neurobiol. 11, 164–170. serotonin receptor 2A genetic variation in postmortem brain samples from
Tannenbaum, B., Anisman, H., 2003. Impact of chronic intermittent challenges subjects who did or did not commit suicide. Am. J. Psychiatry 156, 1456–
in stressor-susceptible and resilient strains of mice. Biol. Psychiatry 53, 1458.
292–303. Turner, C.A., Akil, H., Watson, S.J., Evans, S.J., 2006. The fibroblast growth
Taub, D.D., 2007. Novel connections between the neuroendocrine and immune factor system and mood disorders. Biol. Psychiatry 59, 1128–1135.
systems: the ghrelin immunoregulatory network. Vitam. Horm. 77, 325– Turrin, N.P., Gayle, D., Ilyin, S.E., Flynn, M.C., Langhans, W., Schwartz,
346. G.J., Plata-Salaman, C.R., 2001. Pro-inflammatory and anti-inflammatory
Teare, K.A., Pearson, R.G., Shakesheff, K.M., Raisman, G., Haycock, J.W., cytokine mRNA induction in the periphery and brain following intraper-
2003. Alpha-MSH inhibits inflammatory signalling in olfactory ensheathing itoneal administration of bacterial lipopolysaccharide. Brain Res. Bull.
cells. Neuroreport 14, 2171–2175. 54, 443–453.
Teare, K.A., Pearson, R.G., Shakesheff, K.M., Haycock, J.W., 2004. Alpha- Ueda, S., Sakakibara, S., Yoshimoto, K., 2005. Effect of long-lasting serotonin
MSH inhibits inflammatory signalling in Schwann cells. Neuroreport 15, depletion on environmental enrichment-induced neurogenesis in adult rat
493–498. hippocampus and spatial learning. Neuroscience 135, 395–402.
Tennen, H., Affleck, G., Armeli, S., Carney, M.A., 2000. A daily process Underwood, P.W., 2000. Social support. The promise and reality. In: Rice, B.H.
approach to coping. Linking theory, research, and practice. Am. Psychol. (Ed.), Handbook of Stress, Coping and Health. Sage, Los Angeles, CA, pp.
55, 626–636. 367–391.
72 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Vakili, K., Pillay, S.S., Lafer, B., Fava, M., Renshaw, P.F., Bonello-Cintron, Vitkovic, L., Konsman, J.P., Bockaert, J., Dantzer, R., Homburger, V., Jacque,
C.M., Yurgelun-Todd, D.A., 2000. Hippocampal volume in primary uni- C., 2000. Cytokine signals propagate through the brain. Mol. Psychiatry 5,
polar major depression: a magnetic resonance imaging study. Biol. Psy- 604–615.
chiatry 47, 1087–1090. von Boyen, G.B., Steinkamp, M., Geerling, I., Reinshagen, M., Schäfer, K.H.,
Valentine, A.D., Meyers, C.A., 2005. Neurobehavioral effects of interferon Adler, G., Kirsch, J., 2006. Proinflammatory cytokines induce neurotrophic
therapy. Curr. Psychiatry Rep. 7, 391–395. factor expression in enteric glia: a key to the regulation of epithelial
Valentine, A.D., Meyers, C.A., Talpaz, M., 1995. Treatment of neurotoxic apoptosis in Crohn’s disease. Inflamm. Bowel Dis. 12, 346–354.
side effects of interferon-alpha with naltrexone. Cancer Invest. 13, von Gunten, A., Bouras, C., Kövari, E., Giannakopoulos, P., Hof, P.R., 2006.
561–566. Neural substrates of cognitive and behavioral deficits in atypical Alzhei-
Valentino, R.J., Liouterman, L., Van Bockstaele, E.J., 2001. Evidence for mer’s disease. Brain Res. Rev. 51, 176–211.
regional heterogeneity in corticotropin-releasing factor interactions in the Vulliémoz, N.R., Xiao, E., Xia-Zhang, L., Ferin, M., Wardlaw, S.L., 2006.
dorsal raphe nucleus. J. Comp. Neurol. 435, 450–463. Melanocortin modulation of inflammatory cytokine and neuroendocrine
Vallieres, L., Campbell, I.L., Gage, F.H., Sawchenko, P.E., 2002. Reduced responses to endotoxin in the monkey. Endocrinology 147, 1878–1883.
hippocampal neurogenesis in adult transgenic mice with chronic astrocytic Vythilingam, M., Heim, C., Newport, J., Miller, A.H., Anderson, E., Bronen, R.,
production of interleukin-6. J. Neurosci. 22, 486–492. Brummer, M., Staib, L., Vermetten, E., Charney, D.S., Nemeroff, C.B.,
Vamvakopoulos, N.C., Chrousos, G.P., 1993. Evidence of direct Bremner, J.D., 2002. Childhood trauma associated with smaller hippocam-
estrogenic regulation of human corticotropin-releasing hormone gene pal volume in women with major depression. Am. J. Psychiatry 159, 2072–
expression. Potential implications for the sexual dimophism of the stress 2080.
response and immune/inflammatory reaction. J. Clin. Invest. 92, 1896– Vythilingam, M., Vermetten, E., Anderson, G.M., Luckenbaugh, D., Anderson,
1902. E.R., Snow, J., Staib, L.H., Charney, D.S., Bremner, J.D., 2004. Hippo-
Van der Lely, A.J., Tschop, M., Heiman, M.L., Ghigo, E., 2004. Biological, campal volume, memory, and cortisol status in major depressive disorder:
physiological, pathophysiological, and pharmacological aspects of ghrelin. effects of treatment. Biol. Psychiatry 56, 101–112.
Endocr. Rev. 25, 426–457. Wada, E., Way, J., Lebacq-Verheyden, A.M., Battey, J.F., 1990. Neuromedin B
van der Meer, M.J., Lebeau, A., Duval, P., Grimaldi, B., Terlain, B., Kerdelhue, and gastrin-releasing peptide mRNAs are differentially distributed in the rat
B.A.R., 1995. Synergism between IL-1 beta and TNF-alpha on the activity nervous system. J. Neurosci. 10, 2917–2930.
of the pituitary-adrenal axis and on food intake of rats. Am. J. Physiol. 268, Wallace, T.L., Stellitano, K.E., Neve, R.L., Duman, R.S., 2004. Effects of cyclic
E551–E557. adenosine monophosphate response element binding protein overexpression
van der Meer, M.J., Sweep, C.G., Pesman, G.J., Tilders, F.J., Hermus, A.R., in the basolateral amygdala on behavioral models of depression and anxiety.
1996. Chronic stimulation of the hypothalamus–pituitary–adrenal axis in Biol. Psychiatry 56, 151–160.
rats by interleukin 1beta: central and peripheral mechanisms. Cytokine 8, Wang, C.X., Shuaib, A., 2002. Involvement of inflammatory cytokines in
910–919. central nervous system injury. Prog. Neurobiol. 67, 161–172.
van Gaalen, M.M., Reul, J.M., Gesing, A., Stenzel-Poore, M.P., Holsboer, F., Wang, W., Ji, P., Dow, K.E., 2003. Corticotropin-releasing hormone induces
Steckler, T., 2002a. Mice overexpressing CRH show reduced responsive- proliferation and TNF-alpha release in cultured rat microglia via MAP
ness in plasma corticosterone after a5-HT1A receptor challenge. Genes kinase signalling pathways. J. Neurochem. 84, 189–195.
Brain Behav. 1, 174–177. Wang, W., Solc, M., Ji, P., Dow, K.E., 2004. Corticotropin-releasing hormone
van Gaalen, M.M., Stenzel-Poore, M.P., Holsboer, F., Steckler, T., 2002b. potentiates neural injury induced by oxygen–glucose deprivation: a possible
Effects of transgenic overproduction of CRH on anxiety-like behaviour. involvement of microglia. Neurosci. Lett. 371, 133–137.
Eur. J. Neurosci. 15, 2007–2015. Wang, Y., Su, B., Xia, Z., 2006. Brain-derived neurotrophic factor activates
van Londen, L., Goekoop, J.G., van Kempen, G.M., Frankhuijzen-Sierevogel, ERK5 in cortical neurons via a Rap1-MEKK2 signaling cascade. J. Biol.
A.C., Wiegant, V.M., van der Velde, E.A., De Wied, D., 1997. Plasma levels Chem. 281, 35965–35974.
of arginine vasopressin elevated in patients with major depression. Neu- Warner-Schmidt, J.L., Duman, R.S., 2006. Hippocampal neurogenesis: oppos-
ropsychopharmacology 17, 284–292. ing effects of stress and antidepressant treatment. Hippocampus 16, 239–
van Londen, L., Goekoop, J.G., Zwinderman, A.H., Lanser, J.B., Wiegant, 249.
V.M., De Wied, D., 1998a. Neuropsychological performance and plasma Waselus, M., Valentino, R.J., Van Bockstaele, E.J., 2005. Ultrastructural
cortisol, arginine vasopressin and oxytocin in patients with major depres- evidence for a role of gamma-aminobutyric acid in mediating the effects
sion. Psychol. Med. 28, 275–284. of corticotropin-releasing factor on the rat dorsal raphe serotonin system. J.
van Londen, L., Kerkhof, G.A., van den Berg, F., Goekoop, J.G., Zwinderman, Comp. Neurol. 482, 155–165.
K.H., Frankhuijzen-Sierevogel, A.C., Wiegant, V.M., de Wied, D., 1998b. Wedzony, K., Mackowiak, M., Fijal, K., Golembiowska, K., 1996. Evidence
Plasma arginine vasopressin and motor activity in major depression. Biol. that conditioned stress enhances outflow of dopamine in rat prefrontal
Psychiatry 43, 196–204. cortex: a search for the influence of diazepam and 5-HT1A agonists.
van West, D., Del-Favero, J., Aulchenko, Y., Oswald, P., Souery, D., Forsgren, Synapse 24, 240–247.
T., Sluijs, S., Bel-Kacem, S., Adolfsson, R., Mendlewicz, J., Van Duijn, C., Weiss, J.M., Goodman, P.A., Losito, B.G., Corrigan, S., Charry, J.M., Bailey,
Deboutte, D., Van Broeckhoven, C., Claes, S., 2004. A major SNP haplo- W.H., 1981. Behavioral depression produced by an uncontrollable stressor:
type of the arginine vasopressin 1B receptor protects against recurrent major Relationship to norepinephrine, dopamine and serotonin levels in various
depression. Mol. Psychiatry 9, 287–292. regions of rat brain. Brain Res. Rev. 3, 167–205.
Venters, H.D., Tang, Q., Liu, Q., VanHoy, R.W., Dantzer, R., Kelley, K.W., Weisstaub, N.V., Zhou, M., Lira, A., Lambe, E., Gonzalez-Maeso, J., Hornung,
1999. A new mechanism of neurodegeneration: a proinflammatory cytokine J.P., Sibille, E., Underwood, M., Itohara, S., Dauer, W.T., Ansorge, M.S.,
inhibits receptor signaling by a survival peptide. Proc. Natl. Acad. Sci. Morelli, E., Mann, J.J., Toth, M., Aghajanian, G., Sealfon, S.C., Hen, R.,
U.S.A. 96, 9879–9884. Gingrich, J.A., 2006. Cortical 5-HT2A receptor signaling modulates anxi-
Videbech, P., Ravnkilde, B., 2004. Hippocampal volume and depression: a ety-like behaviors in mice. Science 313, 536–540.
meta-analysis of MRI studies. Am. J. Psychiatry 161, 1957–1966. Wellman, C.L., Izquierdo, A., Garrett, J.E., Martin, K.P., Carroll, J., Millstein,
Vila, N., Castillo, J., Dávalos, A., Chamorro, A., 2000. Proinflammatory R., Lesch, K.P., Murphy, D.L., Holmes, A., 2007. Impaired stress-coping
cytokines and early neurological worsening in ischemic stroke. Stroke and fear extinction and abnormal corticolimbic morphology in serotonin
31, 2325–2329. transporter knock-out mice. J. Neurosci. 27, 684–691.
Vitarbo, E.A., Chatzipanteli, K., Kinoshita, K., Truettner, J.S., Alonso, O.F., Weng, Z., Signore, A.P., Gao, Y., Wang, S., Zhang, F., Hastings, T., Yin, X.M.,
Dietrich, W.D., 2004. Tumor necrosis factor alpha expression and protein Chen, J., 2007. Leptin protects against 6-hydroxydopamine-induced dopa-
levels after fluid percussion injury in rats: the effect of injury severity and minergic cell death via mitogen-activated protein kinase signaling. J. Biol.
brain temperature. Neurosurgery 55, 416–424. Chem. 282, 34479–34491.
H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74 73

Westling, S., Ahrén, B., Träskman-Bendz, L., Westrin, A., 2004. Low CSF Yamamoto, M., Kiyota, T., Horiba, M., Buescher, J.L., Walsh, S.M., Gendel-
leptin in female suicide attempters with major depression. J. Affect. Disord. man, H.E., Ikezu, T., 2007. Interferon-gamma and tumor necrosis
81, 41–48. factor-alpha regulate amyloid-beta plaque deposition and beta-secretase
Wheeler, R.D., Owens, T., 2005. The changing face of cytokines in the brain: expression in Swedish mutant APP transgenic mice. Am. J. Pathol. 170,
perspectives from EAE. Curr. Pharm. Des. 11, 1031–1037. 680–692.
Wichers, M., Maes, M., 2002. The psychoneuroimmuno-pathophysiology of Yamano, M., Ogura, H., Okuyama, S., Ohki-Hamazaki, H., 2002. Modulation of
cytokine-induced depression in humans. Int. J. Neuropsychopharmacol. 5, 5-HT system in mice with a targeted disruption of neuromedin B receptor. J.
375–388. Neurosci. Res. 68, 59–64.
Wichers, M.C., Maes, M., 2004. The role of indoleamine 2,3-dioxygenase Yamasaki, Y., Matsuura, N., Shozuhara, H., Onodera, H., Itoyama, Y., Kogure,
(IDO) in the pathophysiology of interferon-alpha-induced depression. J. K., 1995. Interleukin-1 as a pathogenetic mediator of ischemic brain
Psychiatry Neurosci. 29, 11–17. damage in rats. Stroke 26, 676–680.
Wichers, M.C., Koek, G.H., Robaeys, G., Praamstra, A.J., Maes, M., 2005. Yan, X.X., Baram, T.Z., Gerth, A., Schultz, L., Ribak, C.E., 1998. Co-localiza-
Early increase in vegetative symptoms predicts IFN-alpha-induced cogni- tion of corticotropin-releasing hormone with glutamate decarboxylase and
tive–depressive changes. Psychol. Med. 35, 433–441. calcium-binding proteins in infant rat neocortical interneurons. Exp. Brain
Wichers, M.C., Kenis, G., Leue, C., Koek, G., Robaeys, G., Maes, M., 2006. Res. 123, 334–340.
Baseline immune activation as a risk factor for the onset of depression Yang, K., Xie, G., Zhang, Z., Wang, C., Li, W., Zhou, W., Tang, Y., 2007.
during interferon-alpha treatment. Biol. Psychiatry 60, 77–79. Levels of serum interleukin (IL)-6, IL-1beta, tumour necrosis factor-alpha
Wieczorek, M., Swiergiel, A.H., Pournajafi-Nazarloo, H., Dunn, A.J., 2005. and leptin and their correlation in depression. Aust. N. Z. J. Psychiatry 41,
Physiological and behavioral responses to interleukin-1beta and LPS in 266–273.
vagotomized mice. Physiol. Behav. 85, 500–511. Yang, W., Wang, Q., Kanes, S.J., Murray, J.M., Nishikura, K., 2004. Altered
Windle, R.J., Wood, S.A., Lightman, S.L., Ingram, C.D., 1998. The pulsatile RNA editing of serotonin 5-HT2C receptor induced by interferon: implica-
characteristics of hypothalamo-pituitary–adrenal activity in female Lewis tions for depression associated with cytokine therapy. Brain Res. Mol. Brain
and Fischer 344 rats and its relationship to differential stress responses. Res. 124, 70–78.
Endocrinology 139, 4044–4052. Yaraee, R., Ebtekar, M., Ahmadiani, A., Sabahi, F., 2003. Neuropeptides (SP
Wong, M.L., Kling, M.A., Munson, P.J., Listwak, S., Licinio, J., Prolo, P., Karp, and CGRP) augment pro-inflammatory cytokine production in HSV-
B., McCutcheon, I.E., Geracioti Jr., T.D., DeBellis, M.D., Rice, K.C., infected macrophages. Int. Immunopharmacol. 3, 1883–1887.
Goldstein, D.S., Veldhuis, J.D., Chrousos, G.P., Oldfield, E.H., McCann, Yatham, L.N., Liddle, P.F., Shiah, I.S., Scarrow, G., Lam, R.W., Adam, M.J.,
S.M., Gold, P.W., 2000. Pronounced and sustained central hypernoradre- Zis, A.P., Ruth, T.J., 2000. Brain serotonin2 receptors in major depression:
nergic function in major depression with melancholic features: relation to a positron emission tomography study. Arch. Gen. Psychiatry 57,
hypercortisolism and corticotropin-releasing hormone. Proc. Natl. Acad. 850–858.
Sci. U.S.A. 97, 325–330. Yatsiv, I., Morganti-Kossmann, M.C., Perez, D., Dinarello, C.A., Novick, D.,
Woo, C.H., Eom, Y.W., Yoo, M.H., You, H.J., Han, H.J., Song, W.K., Yoo, Y.J., Rubinstein, M., Otto, V.I., Rancan, M., Kossmann, T., Redaelli, C.A.,
Chun, J.S., Kim, J.H., 2000. Tumor necrosis factor-alpha generates reactive Trentz, O., Shohami, E., Stahel, P.F., 2002. Elevated intracranial IL-18
oxygen species via a cytosolic phospholipase A2-linked cascade. J. Biol. in humans and mice after traumatic brain injury and evidence of neuro-
Chem. 275, 32357–32362. protective effects of IL-18-binding protein after experimental closed head
Wright, C.E., Strike, P.C., Brydon, L., Steptoe, A., 2005. Acute inflammation injury. J. Cereb. Blood Flow Metab. 22, 971–978.
and negative mood: mediation by cytokine activation. Brain Behav. Immun. Yee, B.K., Zhu, S.W., Mohammed, A.H., Feldon, J., 2007. Levels of neuro-
19, 345–350. trophic factors in the hippocampus and amygdala correlate with anxiety-
Wrynn, A.S., Sebens, J.B., Koch, T., Leonard, B.E., Korf, J., 2000. Prolonged c- and fear-related behaviour in C57BL6 mice. J. Neural Transm. 114, 431–
Jun expression in the basolateral amygdala following bulbectomy: possible 444.
implications for antidepressant activity and time of onset. Brain Res. Mol. Ying, S.W., Futter, M., Rosenblum, K., Webber, M.J., Hunt, S.P., Bliss, T.V.,
Brain Res. 76, 7–17. Bramham, C.R., 2002. Brain-derived neurotrophic factor induces long-term
Wu, C.Y., Hsieh, H.L., Jou, M.J., Yang, C.M., 2004. Involvement of p42/p44 potentiation in intact adult hippocampus: requirement for ERK activation
MAPK, p38 MAPK, JNK and nuclear factor-kappa B in interleukin-1beta- coupled to CREB and upregulation of Arc synthesis. J. Neurosci. 22, 1532–
induced matrix metalloproteinase-9 expression in rat brain astrocytes. J. 1540.
Neurochem. 90, 1477–1488. Yirmiya, R., Weidenfeld, J., Pollak, Y., Morag, M., Morag, A., Avitsur, R.,
Wu, R., Dong, W., Cui, X., Zhou, M., Simms, H.H., Ravikumar, T.S., Wang, P., Barak, O., Reichenberg, A., Cohen, E., Shavit, Y., Ovadia, H., 1999.
2007. Ghrelin down-regulates proinflammatory cytokines in sepsis through Cytokines, depression due to a general medical condition, and antidepres-
activation of the vagus nerve. Ann. Surg. 245, 480–486. sant drugs. Adv. Exp. Med. Biol. 461, 283–316.
Wu, Z.L., Ciallella, J.R., Flood, D.G., O’Kane, T.M., Bozyczko-Coyne, D., Yirmiya, R., Pollak, Y., Barak, O., Avitsur, R., Ovadia, H., Bette, M., Weihe, E.,
Savage, M.J., 2006. Comparative analysis of cortical gene expression in Weidenfeld, J., 2001. Effects of antidepressant drugs on the behavioral and
mouse models of Alzheimer’s disease. Neurobiol. Aging 27, 377–386. physiological responses to lipopolysaccharide (LPS) in rodents. Neurop-
Xu, H., Chen, Z., He, J., Haimanot, S., Li, X., Dyck, L., Li, X.M., 2006. sychopharmacology 24, 531–544.
Synergetic effects of quetiapine and venlafaxine in preventing the chronic Yokoyama, M., Suzuki, E., Sato, T., Maruta, S., Inada, K., Watanabe, S.,
restraint stress-induced decrease in cell proliferation and BDNF expression Miyaoka, H., 2005. Effects of intraperitoneal administration of IFN-alpha
in rat hippocampus. Hippocampus 16, 551–559. for one, four, and fourteen days on amino acid levels in various rat brain
Yamada, K., Santo-Yamada, Y., Wada, E., Wada, K., 2002. Role of bombesin regions. J. Interferon Cytokine Res. 25, 187–191.
(BN)-like peptides/receptors in emotional behavior by comparison of Yoshioka, M., Matsumoto, M., Togashi, H., Saito, H., 1995. Effects of condi-
three strains of BN-like peptide receptor knockout mice. Mol. Psychiatry tioned fear stresds on 5-HT release in the rat prefriontal cortex. Pharmacol.
7, 113–117. Biochem. Behav. 51, 515–519.
Yamada, K., Watanabe, A., Iwayama-Shigeno, Y., Yoshikawa, T., 2003a. Young, S.N., Leyton, M., 2002. The role of serotonin in human mood and social
Evidence of association between gamma-aminobutyric acid type A receptor interaction. Insight from altered tryptophan levels. Pharmacol. Biochem.
genes located on 5q34 and female patients with mood disorders. Neurosci. Behav. 71, 857–865.
Lett. 349, 9–12. Zacharko, R.M., Anisman, H., 1991. Stressor-induced anhedonia in the meso-
Yamada, S., Yamamoto, M., Ozawa, H., Riederer, P., Saito, T., 2003b. Reduced corticolimbic system. Neurosci. Biobehav. Rev. 15, 391–405.
phosphorylation of cyclic AMP-responsive element binding protein in the Zaharia, M.D., Ravindran, A.V., Griffiths, J., Merali, Z., Anisman, H., 2000.
postmortem orbitofrontal cortex of patients with major depressive disorder. Lymphocyte proliferation among major depressive and dysthymic patients
J. Neural Transm. 110, 671–680. with typical or atypical features. J. Affect. Disord. 58, 1–10.
74 H. Anisman et al. / Progress in Neurobiology 85 (2008) 1–74

Zanardi, R., Artigas, F., Moresco, R., Colombo, C., Messa, C., Gobbo, C., Zhong, P., Yan, Z., 2004. Chronic antidepressant treatment alters serotonergic
Smeraldi, E., Fazio, F., 2001. Increased 5-hydroxytryptamine-2 receptor regulation of GABA transmission in prefrontal cortical pyramidal neurons.
binding in the frontal cortex of depressed patients responding to paroxetine Neuroscience 129, 65–73.
treatment: a positron emission tomography scan study. J. Clin. Psycho- Zhou, D., Shanks, N., Riechman, S.E., Liang, R., Kusnecov, A.W., Rabin, B.S.,
pharmacol. 21, 53–58. 1996. Interleukin 6 modulates interleukin-1-and stress-induced activation of
Zaremba, J., Losy, J., 2001. Early TNF-alpha levels correlate with ischaemic the hypothalamic–pituitary–adrenal axis in male rats. Neuroendocrinology
stroke severity. Acta Neurol. Scand. 104, 288–295. 63, 227–236.
Zellweger, M.J., Osterwalder, R.H., Langewitz, W., Pfisterer, M.E., 2004. Zhou, J.N., Riemersma, R.F., Unmehopa, U.A., Hoogendijk, W.J., van Heer-
Coronary artery disease and depression. Eur. Heart J. 25, 3–9. ikhuize, J.J., Hofman, M.A., Swaab, D.F., 2001. Alterations in arginine
Zhang, J., Terreni, L., De Simoni, M.G., Dunn, A.J., 2001. Peripheral inter- vasopressin neurons in the suprachiasmatic nucleus in depression. Arch.
leukin-6 administration increases extracellular concentrations of serotonin Gen. Psychiatry 58, 655–662.
and the evoked release of serotonin in the rat striatum. Neurochem. Int. 38, Zhu, J., Mix, E., Winblad, B., 2001. The antidepressant and antiinflammatory
303–308. effects of rolipram in the central nervous system. CNS Drug Rev. 7, 387–
Zhang, W., Qin, L., Wang, T., Wei, S.J., Gao, H.M., Liu, J., Wilson, B., Liu, B., 398.
Zhang, W., Kim, H.C., Hong, J.S., 2005. 3-Hydroxymorphinan is neuro- Zhu, Y., Saito, K., Murakami, Y., Asano, M., Iwakura, Y., Seishima, M., 2006.
trophic to dopaminergic neurons and is also neuroprotective against LPS- Early increase in mRNA levels of pro-inflammatory cytokines and their
induced neurotoxicity. FASEB J. 19, 395–397. interactions in the mouse hippocampus after transient global ischemia.
Zhang, X.M., Duan, R.S., Chen, Z., Quezada, H.C., Mix, E., Winblad, B., Zhu, Neurosci. Lett. 393, 122–126.
J., 2007. IL-18 deficiency aggravates kainic acid-induced hippocampal Zigmond, M.J., Castro, S.L., Keefe, K.A., Abercrombie, E.D., Sved, A.F., 1998.
neurodegeneration in C57BL/6 mice due to an overcompensation by IL- Role of excitatory amino acids in the regulation of dopamine synthesis and
12. Exp. Neurol. 205, 64–73. release in the neostriatum. Amino Acids 14, 57–62.

S-ar putea să vă placă și