Sunteți pe pagina 1din 18

Chapter 75

Enteric Neurobiology of Stress


Sumei Liu and Jackie D. Wood

75.1  STRESS: CASE EXAMPLES


A routine history revealed that Ms. A, a 44-yr-old attor-
ney, had experienced up to six episodes of cramping lower
abdominal pain associated with watery diarrhea per month
over a period of 15 years. These symptoms were worsened
by stress associated with anticipation of a courtroom trial
or the stress of presentation of the defense of a client dur-
ing the actual trial.
FIGURE 75.1  Effects of cold-restraint stress on fecal pellet out-
Barbara, a 22-year-old second year medical student, put in guinea pig.  (A) The non-stressed control remained caged in its
walked to the front of the lecture auditorium with a ques- normal environment in the vivarium. (B) Guinea pig was restrained in a
tion for the professor at the end of the final lecture in a supine position by placing a tie around each ankle and loosely extending
series on gastrointestinal physiology. The question: Can and tying each of the four extremities to metal hooks at the corners of
a wooden rectangular flat platform and placed in a 4°C cold room for 2
you explain to me why I routinely experienced lower
hours.
abdominal distress and diarrhea as I prepared for and
anticipated the examinations in the medicine course mod-
ules throughout the year?
A male guinea pig was held in a supine position by initiates colitis in the cotton-top tamarin model for ulcera-
placing a tie around each ankle and gently extending and tive colitis and colon cancer.6 Acutely applied forms of
tying each of the four extremities to metal hooks at the stress — among which are handling, water avoidance,
corners of a rectangular flat platform. Thus restrained, the abdominal surgery, acoustic stimulation, wrap restraint at
guinea pig was placed in a 4°C cold room for 2 hours. A room temperature, swimming, and ether anesthesia — all
second male guinea pig of the same weight and age and suppress gastric emptying and simultaneously increase
from the same holding rack in the vivarium was caged at secretion and propulsive motility in the large intestine of
room temperature and allowed to feed and drink H2O ad mice, rats, guinea pigs, and dogs. Anger, fear, pre-surgical
libitum during the 2 hour period. Figure 75.1 shows fecal anxiety, and intense exercise are forms of stress that exac-
output for both guinea pigs during the period of cold- erbate the symptoms of IBS.9,13,14 Cold exposure, painful
restraint stress in the 2 hour period. stimuli, dichotomous listening tests, fear, anxiety, anger, or
The three examples illustrate an expanding body of stressful interviews all stimulate colonic motor activity in
converging clinical and experimental evidence that impli- healthy human volunteers.14,15
cates stress as a factor in disordered gastrointestinal func-
tion in both humans and animals. Stressful life events in 75.2  STRESS AND THE SYMPATHETIC
humans can be associated with the onset, symptom exac-
NERVOUS SYSTEM
erbation, and reactivation of gastrointestinal disorders,
which include irritable bowel syndrome (IBS), ulcerative Earlier physiological explanation for the above exam-
colitis, Crohn’s disease, gastroesophageal reflux disease, ples of stress-associated gastrointestinal symptoms would
and peptic ulcer disease.1–6 Exposing rats or guinea pigs likely have been that effects of stress reflect a brain–gut
to stress inhibits gastric contractions and gastric empty- interaction mediated by activation of the sympathetic
ing while stimulating contractile and secretory activity division of the autonomic nervous system. The sympa-
in the colon.7–12 Stress, in rats, exacerbates colitis3 and thetic nervous hypothesis had credibility because the

Physiology of the Gastrointestinal Tract, Two Volume Set. DOI: 10.1016/B978-0-12-382026-6.00075-0


© 2012 Elsevier Inc. All rights reserved. 2001
2002 SECTION  |  VI  Consequences of Disregulated Physiology

neuroendocrine components in the stress response were 75.4  PSYCHOGENIC STRESS AND ENTERIC
known to include increased secretion of epinephrine and MAST CELLS
norepinephrine by the sympathetic nervous system and
adrenal glands.16 Nevertheless, sympathetic activation Figure 75.2 is a heuristic model for the physiology of
is no longer a plausible explanation for stress-related gut brain–gut interactions in the stressed animal or human.
symptoms. Descending spinal activation of the sympa- A unifying concept emerges from consideration that a
thetic nervous system cannot explain the stress-associ- major impact of stress on the intestinal tract is reflected by
ated symptoms of cramping lower abdominal pain, fecal symptoms reminiscent of the diarrhea-predominant form
urgency, and watery diarrhea in humans or the similar of IBS. Cramping abdominal pain, acute fecal urgency,
effects of stress in animals. and explosive watery diarrhea are hallmarks not only
Advances in what is known about the sympathetic of diarrhea-predominant IBS, but also infectious enteri-
interface with the enteric nervous system (ENS) rule out tis, radiation-induced enteritis, food allergy, and noxious
sympathetic involvement.17,18 The majority of sympathetic mucosal irritation (e.g., senna laxatives). The concept
noradrenergic fibers entering the small and large intestine starts with the evidence that enteric mast cells receive
innervate ganglion cells in the ENS. Sympathetic activa- brain-derived input in the stressed individual. The same
tion and the release of norepinephrine inside the ENS mast cells intercept and become sensitized to microor-
inhibits firing of secretomotor neurons in the submucosal ganisms and other sensitizing antigens that find their way
plexus, thereby suppressing the neurogenic secretion across the intestinal mucosal barrier. Mast cells detect
responsible for generation of loose watery stools.17,19 Once threats of this nature to the integrity of the whole animal
released in the ENS, norepinephrine acts at presynaptic and signal their presence to the ENS, which responds by
alpha2a receptors to suppress transmission at both slow and calling up from its program library a program of special-
fast excitatory synapses and at excitatory neuromuscular ized integrated secretion and motility that removes the
junctions.20,21 Sympathetic inhibition of neurotransmis- threat from the lumen.33–36 Threat removal starts with
sion in the synaptic microcircuitry makes improbable any stimulation of mucosal secretion, which flushes the threat-
suggestion that sympathetic activation is responsible for ening agent out of the mucosa into the lumen and holds it
initiation of the powerful propulsive motility in the colon in suspension. The secretory response then links to pow-
that accounts for cramping lower abdominal pain in dogs erful propulsive motility, which propels the secretions
and humans.22 Noradrenergic inhibitory synapses on together with the offending agent quickly in the anal direc-
secretomotor neurons “shut down” secretion in the face tion. Cramping abdominal pain accompanies the strong
of sympathetically reduced intestinal blood flow making propulsive contractions.22,37 Urgency is experienced when
it unlikely that stress-associated diarrhea involves sympa- arrival of the liquid bolus distends the rectosigmoid region
thetic activation (see Chapter 21). The evidence against and reflexively opens the internal anal sphincter with con-
sympathetic activation directs attention to involvement of tinence protection now provided only by spinal reflexes
other enteric neurophysiological mechanisms that come that contract the puborectalis and external anal sphincter.
into play in stress and will be dealt with in this chapter. Sensory information arriving in the brain from receptors in
the rapidly distending rectosigmoid accounts for the con-
scious sensation of urgency and fear of incontinence might
further exacerbate the individual’s emotional stress. The
75.3  PSYCHOGENIC STRESS AND symptom of acute explosive watery diarrhea becomes self-
FUNCTIONAL GASTROINTESTINAL explanatory in this scenario.
Enteric mast cells are implicated as a key step in the
DISORDERS initiation of stress-evoked symptoms (Figures 75.2 and
The symptoms in the “stressed-out” attorney and medi- 75.3). Enteric mast cells are packed with granules that are
cal student are characteristic of functional gastrointestinal sites of storage for a broad mix of preformed chemical
disorders (FGIDs). FGIDs are those disorders in which no mediators (Figure 75.3). Antigens and neural input stimu-
abnormal metabolic or physical processes, which account late the mast cells to release mediators, which diffuse into
for the symptoms, can be identified.23 IBS is an example the extracellular milieu as paracrine signals to other kinds
of a significant FGID, which compromises the quality of of cells including the neurons and glia of the ENS. Mast
life for 10–20% of humans worldwide.24–27 Predominant cells express high-affinity receptors for IgE antibodies or
symptoms of IBS are abnormal defecation associated with other immunoglobulins on their surfaces. When antibod-
abdominal pain, both of which are exacerbated by psy- ies to a sensitizing antigen occupy the receptors and cross-
chogenic stress.28 Psychological stress and negative life linking occurs by interaction of the sensitizing antigen
experiences are widely recognized as exacerbating psycho- with the bound antibody, a flood of different mediators is
social factors in IBS.24,29–32 released from the mast cells (Figure 75.3).
Chapter  |  75  Enteric Neurobiology of Stress 2003

Brain STRESS

ENTERIC EFFECTOR
NERVOUS SYSTEM SYSTEMS

Interneurons Muscle
Information
Brain to Mast Cell Connection

Sensory Motor
Processing Glands
Neurons Neurons
Reflexes
Program Blood Vessels
Library

INTESTINAL
BEHAVIOR
Mast Cell
Mediators (Hyper-Secretion)

Inflammatory
Surveillance
Antibodies (Power Propulsion)

Antigen
SYMPTOMS
MAST CELL
(Diarrhea)
(Pain)

FIGURE 75.2  Heuristic model for the physiology of brain–gut interactions in stressed animals or humans.  The ENS is a minibrain with
sensory neurons, interneurons, and motor neurons. Enteric mast cells are in position to detect foreign antigens and signal their presence to the ENS.
Stimulated mast cells release multiple paracrine mediators (e.g., histamine and proteases) simultaneously. Some of the mediators signal the ENS while
others are attractant factors for polymorphonuclear leukocytes responsible for acute inflammatory responses. The ENS responds to the mast cell signal
by initiating a program of coordinated secretion and propulsive motility that expels the source of antigenic stimulation from the bowel. Symptoms of
cramping abdominal pain and acute urgency and diarrhea result from operation of the neural program. Neural inputs to mast cells from the brain stimu-
lates simultaneous release of chemoattractant factors for inflammatory cells, which establish inflammatory surveillance at a dangerous interface and
chemical signals to the ENS with effects that mimic the symptoms of antigenic stimulation. Stress acts through the brain–mast cell connection.

that manage access across the mucosal barrier.38,39 Results


from nematode-infected animals and food-allergic models
have expanded our understanding of mast cell involvement
in enteric immunoneural communication. This work shows
that a second exposure to antigens isolated from the infec-
tious agent (e.g., intestinal nematodes in rats, guinea pigs,
or dogs) or food products (e.g., β-lactoglobulin in milk-
sensitized guinea pigs) results in predictable integration of
intestinal motility and secretory behavior.35,37,39,40–42
Recognition of sensitizing antigens by antibodies
FIGURE 75.3  Multiple mast cell mediators, which become signals bound to Fc receptors on the sensitized mast cells triggers
to other systems in the gut, are stored in granules inside enteric mast degranulation and release of multiple mediators. Once in
cells or are synthesized from membrane lipids. Close spatial asso-
the interstitial compartment, the mediators become para-
ciation of inflammatory/immune cells with the mast cell reflects release
of chemoattractant factors from the mast cell in the inflamed mucosa/ crine messages to the ENS, which responds by suspending
submucosa. operation of other programs in its library and “running”
a defense program that integrates secretion and special-
ized propulsive motility to eliminate the antigen from the
Experimental infections with nematode intestinal para- lumen. Copious neurogenic secretion and elevated intesti-
sites (e.g., Trichinella spiralis) in animals stimulate prolif- nal blood flow followed by orthograde power propulsion
eration of enteric mast cells and are experimental models (see Chapter 21) of the luminal contents are the behavioral
for mast cell functions in detection and signaling of the components of the program.22,37 Mast cells are equipped
presence of sensitizing antigens and infectious invaders and strategically placed to recognize foreign agents that
2004 SECTION  |  VI  Consequences of Disregulated Physiology

threaten whole-body integrity and to signal the ENS to mast cell proteases and histamine into the lumen of the
program a defensive response to eliminate the threat. A small intestine takes place as a response to stress of cold-
similar form of ENS-programmed defense takes place in induced pain and mimics responses to sensitizing antigens
the upper half of the jejunum, duodenum, and stomach in in food allergies (Figure 75.4). Santos et al.46 instructed
the form of emesis. subjects to dip one hand into a container of ice-cold H2O
Mast cell function in immunoneural communication in for 1 minute at 15 second intervals repeatedly for 15
the gut is an immune counterpart of sensory detection and minutes. Continuous aspiration and analysis of the con-
information coding in sensory neurophysiology. In sensory tents from subjects’ small intestine show appearance of
physiology, sensory neurons are genetically programmed mast cell proteases and histamine statistically correlated
to express detection mechanisms for specific stimuli (e.g., with the cold-H2O stressor (Figure 75.4). The speed of
receptors for touch, temperature, or pH) that remain fixed the stress response most likely reflects neurotransmission
throughout the life of the individual. Mast cells, on the over a neural pathway to the enteric mast cells and makes
other hand, acquire specific detection capabilities through it unlikely that the responses resulted from any form of
the flexibility of recognition functions inherent in synthe- endocrine signaling from the brain.
sis by the immune system of new specific antibodies that A neural highway for central nervous signaling to
become attached to Fc receptors (e.g., FceRI receptors in enteric mast cells is suggested by close histological prox-
allergies) on the mast cells.43 Detection specificity for for- imity between vagal and spinal sensory afferent nerves
eign antigens is acquired and reinforced throughout life and enteric mast cells.47 Elevated expression of histamine
due to formation of new antibodies that bind to and remain in intestinal mast cells occurs in response to vagal nerve
bound to the mast cell immunoglobulin receptors. Output stimulation and also suggests existence of a brain–mast
signals from mast cells, which are triggered by cross-link- cell connection.48 Stimulation of neurons in the brainstem
ing of antigens with the attached antibodies, are chemical by intracisternal injection of thyrotropin-releasing hor-
in nature and analogous to chemical output signals (i.e., mone (TRH) evokes degranulation of enteric mast cells
neurotransmitters) from sensory neurons to second-order and release of mast cell protease II into the lumen of rat
neurons in the brain and spinal cord. Both mast cells and small intestine and adds to the evidence for a brain–mast
sensory neurons ultimately code information on the sensed cell connection.49 Vagotomy or administration of the mus-
parameter by releasing a chemical message that is decoded carinic receptor antagonist, atropine, suppresses the action
by information processing circuits in the nervous system. of intracisternal TRH on release of mast cell mediators,
including release of mast cell protease II and prostaglandin
75.4.1  Mast Cells and the Brain–Gut D2 in the small intestine. The evidence for a brain–mast
cell connection takes on significance for understanding
Connection in Stress stress-associated symptoms of gut origin, because the
Enteric mast cells, in addition to their sensing functions, symptoms associated with release of mast cell mediators
are relay points for transmission of information on emo- are the same irrespective of whether mast cells are activated
tional status from the brain to the ENS (see Figure 75.2). by antigen-antibody cross-linking in allergies and infec-
This is a brain–gut connection that links central psycho- tions or by input down the brain–gut axis during stress.
logical status (e.g., psychogenic or physical stress) to a
specific state of the digestive tract via mast cell signal-
ing. Release of mast cell mediators, evoked in this man-
75.5  NEUROBIOLOGY OF STRESS
ner, activates an “alarm program” in the ENS program Symptomatic manifestations of stress in upper and lower
library, which when “running,” produces the same symp- regions of the digestive tract are different. Cold-restraint
toms of diarrhea and abdominal distress as antigen-evoked stress in rats evokes inflammation and erosions in the
degranulation in food allergies and infectious enteritis. stomach. Intracerebral injection of TRH mimics the gas-
The brain-to-enteric mast cell connection can be demon- tric responses to cold-restraint stress. In the large intestine,
strated experimentally as a release of mast cell proteases restraint stress alters motility, induces a diarrheal state,
in a Pavlovian-like conditioned response. Assays for the and exacerbates nociceptive pain responses to distension
release of mast cell proteases into the systemic circulation in association with increased release of histamine from
or into the intestinal lumen are useful tools for measure- enteric mast cells.50 Like the effects of centrally acting
ment and timing of degranulation of enteric mast cells.44 TRH on gastric mucosal pathology, intracerebroventricular
Release of mast cell proteases into the systemic circula- injection of corticotropin-releasing factor (CRF) mimics
tion can be evoked as a Pavlovian-conditioned response large intestinal responses to stress.8,51 CRF receptor antag-
in laboratory rats by light or auditory stimuli after several onists or pretreatment with mast cell-stabilizing drugs sup-
trials of pairing these stimuli with injection of a sensitiz- presses the stress-evoked responses in the large bowel.9,10
ing antigen, such as ovalbumin.45 In humans, release of Neuropharmacology suggests that CRF is working as a
Chapter  |  75  Enteric Neurobiology of Stress 2005

(A) Aspiration (B) Aspiration


Cold Cannula
Antigen Cannula
0.6 Stress Balloons 0.5

Mast Cell Tryptase Release


Mast Cell Tryptase Release
Challenge Balloons
(Egg Albumin)

(Units/15 min)
(Units/15 min)
0.4 0.3

0.2 0.2

0 0
15 0 15 30 45 60 15 0 15 30 45 60 75 90
Time (Minutes) Time (Minutes)
FIGURE 75.4  Cold-pain stress or antigen challenge degranulates enteric mast cells in humans.  A group of patients with confirmed allergy to
egg albumin and a group of normal subjects were asked to immerse one hand into ice-cold H2O for 1 minute at 15 second intervals in 2 successive 15
minute trials. (A) Degranulation of mast cells measured as amounts of tryptase in aspirates from the small intestine. Cold-pain stress evoked release of
mast cell tryptase in both food-allergic patients (O) and normal subjects (•). (B) Egg albumin, infused into the small intestinal lumen, evoked mast cell
degranulation manifested as elevated mast cell tryptase in aspirates from food-allergic patients, but not in normal subjects. (Adapted from 46).

neurotransmitter in the neural networks in the brain and


“Puff” Pipette
ENS to generate stress-induced large intestinal symptoms (CRF, Urocortin, etc.)
of abdominal pain, fecal urgency, and diarrhea. Microelectrode

75.5.1  CRF CRF or CRF Receptor


ENTERIC
NEURON
CRF in the “big brain” is expressed predominantly in the
hypothalamic paraventricular nucleus. Nevertheless, it is
distributed in neural elements throughout the brain.52 The
majority of the endocrine, neurochemical, electrophysi- Fluorescent
Tags
ological, and behavioral effects, which are reported to
Primary
occur after injection of CRF into the brain, are essentially Antibody
the same as those associated with experimentally induced Secondary
stress. Virtually all of the effects of intracerebral applica- Antibodies
tions of CRF can be reduced or abolished by administra- FIGURE 75.5  Indirect immunofluorescence and intraneuronal elec-
tion of CRF receptor antagonists, and this reinforces belief trophysiological recording with “sharp” microelectrodes are used to
in a predominant central role for CRF neurotransmission study the neurobiology of CRF and its receptors in the ENS. CRF or
in stress.53–56ab Chronic stress is known to exert an inhibi- other members of the CRF family of peptides are applied to the neurons
by nitrogen pressure ejection (“puffs”) from fine-tipped micropipettes. In
tory effect on hair growth in animals and humans. CRF immunohistochemistry allied with the electrophysiology, primary anti-
and adrenocorticotropic hormone (ACTH), aside from bodies recognize and bind to CRF or its receptor types and fluorescently
being key mediators in the endocrine and neuroimmune tagged second antibodies bind to and mark the location of bound primary
responses to stress, inhibit the growth cycles of cutane- antibodies.
ous hair follicles.56b Intraperitoneal administration of the
non-selective CRF1/CRF2 receptor antagonist, astressin- excitation in hippocampal slice preparations is associated
B, in nude mice with a transgene that overexpressed CRF, with reduction of the hyperpolarizing responses that follow
induces pigmentation and hair regrowth.56b bursts of spikes,60 and this is reminiscent of suppression of
Application of experimental methods for recording hyperpolarizing after potentials by certain neurotransmit-
electrical and synaptic behavior in single neurons in con- ters/modulators in ENS neurons (Chapter 21).64,65
cert with immunohistochemistry has greatly advanced CRF, when functioning as a neurotransmitter or neu-
insight into the role of CRF in the brain and in the ENS romodulator in the CNS, alters the plasticity of synaptic
(Figure 75.5). Application of electrophysiological methods transmission in forms that may be observed as long-term
generally finds that exposure to CRF stimulates firing of potentiation or long-term depression depending on the
single neurons in the hypothalamus,57 cerebral cortex,58 studied region of the brain or spinal cord.66–68 This is rele-
cerebellar Purkinje cells in culture,59 hippocampus,60,61 vant in terms of ramifications of prolonged stress, because
locus coeruleus,62 periaqueductal gray,63 and motor neu- long-term changes in synaptic strength are associated
rons in the lumbar spinal cord.58 CRF-evoked neuronal with memory functions in the CNS. Exposure to CRF
2006 SECTION  |  VI  Consequences of Disregulated Physiology

produces long-lasting enhancement of synaptic efficacy CRF receptors expressed by ENS neurons. Nevertheless,
in the hypothalamus.69,70 Antagonists at both CRF1R other lines of early evidence suggested that CRF might be
and CRF2R receptors depress the induction of long-term expressed in organs outside the brain and be released to act
potentiation in the CA1 region of the hippocampus.71 locally. Outside the brain, expression of CRF immunore-
In the cerebellum, CRF receptor antagonists inhibit the activity has been reported for the digestive system, entero-
induction of long-term depression of synaptic transmis- chromaffin cells, and by immune/inflammatory cells.90–94
sion.72 In the spinal cord, CRF converts long-term synaptic Current evidence suggests that intestinal manifesta-
suppression to facilitation.66 tions of stress reflect the release of CRF as a neurotrans-
mitter/neuromodulator in the integrative microcircuits of
the ENS well outside the hypothalamic-pituitary-adrenal
75.5.2  CRF in Stress
(HPA) axis. Earlier evidence supporting this was find-
Acutely applied forms of stress (e.g., water avoidance, ing that intraperitoneal administration of CRF mimics
handling, acoustic stimulation, abdominal surgery, move- stress-evoked changes that are mediated by a local action
ment restraint, swimming, or ether anesthesia) each inhibit inside the intestine. As early as 1992, Hanani and Wood95
gastric emptying in mice, rats, guinea pigs, and dogs.73 found that applications of CRF to ENS neurons evoked
Similarly in human subjects, anger, fear, preoperative anxi- firing in the guinea pig small intestinal myenteric plexus,
ety, and intense exercise are forms of stress that result in raising the possibility of an ENS signal function for CRF
slowing of gastric emptying and exacerbation of IBS reminiscent of its signaling function in the brain (Figure
symptoms.13,14 Acute stress, intracerebral injection of CRF, 75.6). The stress-like effects of peripheral administration
or urocortin 1 suppresses propulsive motility and transit in of CRF cannot be attributed to action in the brain and are
the small intestine of animals.74–78 Moreover, acute psy- consistent with a signaling function for CRF in the ENS
chological stress suppresses the small intestinal migrating analogous to its neurotransmitter function in the CNS.96
motor complex in humans.79 Unlike the inhibitory effects Intraperitoneal administration of CRF evokes delayed
on gastric and small intestinal motility, stress stimulates gastric emptying that is mediated by stimulation of CRF2R
contractile activity of the colonic musculature, accelerates receptors and stimulates colonic transit mediated by the
colon transit, and stimulates defecation in animal mod- CRF1R receptor.97 Microelectrode recording in concert
els, human volunteers, and IBS patients.14,75,80,81 Central with immunohistochemistry in ENS neurons identifies the
administration of CRF or urocortin 1 mimics effects of CRF1R receptor as the predominant subtype expressed by
stress by stimulating colonic motility, increasing colonic the ganglion cell bodies in the guinea pig intestine (see
transit, and accelerating fecal pellet output in rats.82 Figure 75.5).98
Urocortin 1 is included in the CRF family of mammalian Aside from effects on gastrointestinal motility, stress
messenger peptides,83 which also includes urocortin 284 and strongly influences intestinal glandular secretion and
urocortin 385 and two non-mammalian CRF-like peptides, mucosal barrier function in both animal models and human
sauvagine86 and urotensin 1.87 Intracerebral administra- subjects (see Chapter 74). For example, intravenous or
tion of non-selective or selective CRF1R receptor antago- intraperitoneal injection of CRF evokes acute watery
nists suppresses the effects of both stress and administration diarrhea in rats.11 In a related manner, psychological stress
of CRF or urocortin 1 on colonic motor activity.75,80,82,88 in human subjects reduces H2O absorption and converts
Although urocortins binds with high affinity to CRF2R absorption of Na and Cl to secretion of NaCl and H2O
receptors in the brain, CRF2R receptor antagonists do not in the jejunum.99,100 Elevated secretion in these cases
attenuate stress-induced acceleration of colonic transit or the is associated with increased blood flow in support of the
actions of CRF and urocortin-1 on colonic motility.82 secretory response, which is attributed to elevated firing of
secretomotor/vasodilator neurons in the individuals’ sub-
75.5.3  CRF Putative Neurotransmission in mucosal plexus. The result of elevated secretomotor neuro-
nal activity is increased liquidity of the intestinal contents
the ENS
and a predisposition to neurogenic secretory diarrhea
Downstream from the brain, CRF signaling inside the (Figure 75.7). In rats, exposure to restraint or water avoid-
ENS is implicated in stress-evoked alterations in motil- ance stress or intraperitoneal administration of CRF
ity and mucosal secretory and barrier functions. An early induces rapid onset watery diarrhea associated with stimu-
suggestion was that CRF, which does not enter the brain lation of mucosal secretion and increased epithelial barrier
from the blood, is actively transported out of the brain into permeability in the colon.11,12,101,102
the systemic circulation.89 This implies that CRF, after The connection between CRF and stress-evoked
release inside the brain during stress, might enter and be mucosal changes in animals has pathophysiological, clini-
carried by the blood, in hormonal fashion, to the gastroin- cal, and therapeutic implications. It suggests that CRF sig-
testinal tract where it produces its actions by binding to naling in the ENS might have direct involvement in the
Chapter  |  75  Enteric Neurobiology of Stress 2007

(A) AH-Neuron large organic molecules in mucosal biopsies from human


colon and that pretreatment with the CRF receptor antago-
nist, α-helical CRH (9-41), blocks this action of CRF.103
Cold temperature stress and the presence of the microflora
Post-spike AH
in the lumen are the only requirements for development of
inflammatory bowel disease in a primate model for ulcera-
tive colitis and colon cancer.6 Moreover, psychological
Post-spike AH stress exacerbates ulcerative colitis and leads to relapse in
CRF (30 nM) 20mV human patients is common knowledge.5,104,105
40s

(B) S-Neuron
75.5.4  CRF in Mucosal Secretion
Application of CRF to the serosal side of flat-sheet prepa-
rations of rat or guinea pig colon in Ussing flux chambers
stimulates electrogenic chloride secretion, which can be
measured as elevation of baseline short circuit current across
the mucosa. Increases in mucosal permeability to large mol-
ecules, such as horseradish peroxidase and mannitol, occur
in parallel with the CRF-evoked increases in chloride secre-
1st “Puff” CRF (25µm) tion.101 CRF-evoked increases in chloride secretion are
suppressed or abolished by application of nerve-blocking
agents (e.g., tetrodotoxin) or CRF receptor antagonists (e.g.,
α-helical CRH (9-41).101 Blockade of CRF-evoked mucosal
secretion by nerve-blocking agents is reliable evidence that
40mV this action of CRF results from stimulating the firing of
secretomotor neurons in the submucosal plexus.
10s
2nd “Puff” CRF (25µm)
FIGURE 75.6  CRF-evoked excitatory responses are different for
AH- and S-type enteric neurons when recorded intraneuronally with 75.5.5  CRF in the ENS
“sharp” microelectrodes. (A) Application of 30 nM CRF in the bath-
ing medium evoked a slow excitatory postsynaptic potential-like response
Aside from stimulation of neurogenic secretion, multiple
consisting of slowly activating membrane depolarization, increased input forms of evidence, analogous to the evidence for the “big
resistance, action potential discharge, and suppression of the hyperpo- brain,” point to the neural networks of the ENS “minibrain” as
larizing response (AH) at the end of each action potential. Downward the site where peripheral CRF-related mechanisms contribute
deflections on the trace are electrotonic potentials evoked by repetitive to stress-induced changes in intestinal motility and mucosal
injection of hyperpolarizing current pulses through the electrode. Increased
amplitude of the electrotonic potentials during the depolarizing response
function. The evidence can be summarized as follows:
reflects increase in input resistance due to closure of potassium channels. 1. Application of CRF to segments of guinea pig small
(B) Two continuous traces showing application of 25 µM CRF by micro
pressure ejection (100 ms “puffs”; see Figure 75.5). CRF evoked mem-
intestine in vitro evokes muscle contractions that are
brane depolarization accompanied by intense discharge of action potentials. blocked by tetrodotoxin, indicating that the CRF-
Application of a second puff 2 minutes later, apparent on the bottom trace, evoked contractions are mediated by stimulation of
evoked a much weaker response indicative of tachyphylaxis. S-type neu- excitatory musculomotor neurons.106
rons, but not AH-neurons show tachyphylaxis to CRF. 2. Exposure of rat isolated duodenal and colonic segments
to CRF enhances neurally mediated phasic contrac-
tions and neurally mediated peristaltic propulsion, and
stress-induced symptoms of watery diarrhea, urgency, and this action is prevented by the peptidergic CRF recep-
cramping lower abdominal pain that are present in FGIDs tor antagonist α-helical CRF (9-41) or the non-peptide
and mucosal inflammation in humans. CRF function as a receptor antagonist, CP-154,526.107,108
neurotransmitter/neuromodulator in the microcircuitry 3. Application of CRF to the myenteric or submucosal
that controls activity of secretomotor/vasodilator neurons plexus of guinea pig small and large intestine evokes exci-
is likely to be the underlying factor in effects of stress to tatory responses, which are blocked by selective CRF1R,
open the mucosal barrier and allow sensitizing antigens to but not CRF2R antagonists, in single neurons recorded
cross the barrier and evoke or exacerbate mucosal inflam- with intracellular microelectrodes (see Figure 75.6).95,98
matory responses in the colon. This is supported by evi- 4. H2O-avoidance stress or intraperitoneal injection of
dence that CRF acts to increase mucosal permeability to CRF induces elevated expression of c-fos, a marker of
2008 SECTION  |  VI  Consequences of Disregulated Physiology

(A) (B) (C)

Cholinergic Secretomotor/ Cholinergic Secretomotor/ Non-cholinergic Secretomotor/


Vasodilator Neuron Vasodilator Neuron Vasodilator Neuron

Arteriole Arteriole Arteriole

Crypt Crypt Crypt

Secretion Secretion Secretion

FIGURE 75.7  Three kinds of secretomotor neurons in guinea pig small intestine.  (A) Cholinergic secretomotor/non-vasodilator neurons express
NPY and innervate the secretory glands (crypts of Lieberkühn), but not arterioles. (B) Cholinergic secretomotor/vasodilator neurons contain calretinin
and innervate the secretory glands and periglandular arterioles. (C) Non-cholinergic secretomotor/vasodilator neurons innervate the secretory glands and
periglandular arterioles. These neurons contain VIP in all species and are the only neurons in the submucosal plexus that express CRF.

neuronal activation, in colonic myenteric neurons, and (A) Myenteric (B) Submucosal
this is blocked by peripheral application of the CRF1/ Plexus Plexus
CRF2R antagonist, astressin, or the selective peptider-
gic CRF1R receptor antagonist, CP-154,526.109,110 Calbindin--------------------0% Calbindin--------------------0%
5. RT-PCR and Western blot analysis detects CRF1R Calretinin--------------------0% Calretinin--------------------0%
receptor mRNA transcripts, and protein and immu- ChAT ----------------------4.5% ChAT ------------------------0%
nohistochemical analysis reveals the expression of
CRF1 Receptor ------------0% CRF1 Receptor ------------0%
CRF1R protein by neurons in both myenteric and sub-
mucosal plexuses.108,111 NOS -----------------------5.0% NOS --------------------------0%

NPY --------------------------0% NPY --------------------------0%

75.5.6  CRF Localization in the ENS Serotonin -------------------0% Serotonin -------------------0%

Immunohistochemical localization of CRF in the ENS Somatostatin---------------0% Somatostatin---------------0%


adds to the lines of evidence suggesting that it is a key Substance P-------------4.2% Substance P----------------0%
neurotransmitter in the integrative microcircuitry that
VIP----------------------------0% VIP--------------------------77%
mediates stress-evoked changes in intestinal behavior. It
is expressed in neuronal cell bodies and nerve fibers in FIGURE 75.8  Proportions of neurons that express definitive chemi-
the myenteric and submucosal plexuses of the small and cal codes in combination with CRF immunoreactivity in guinea pig
large intestine and the myenteric plexus of the stomach in small bowel. (A) In the myenteric plexus, only cholinergic (ChAT)
neurons and nitrergic (NOS) and substance Pergic neurons coexpress
guinea pigs (Figure 75.8).112 Most, if not all, of the CRF
CRF. (B) In the submucosal plexus only VIPergic neurons express CRF.
immunoreactive nerve fibers, are presumed to be intrin- (Adapted from Liu et al. 112.)
sic because they remain in organotypic culture after time-
dependent degeneration of extrinsic sympathetic and spinal
afferent fibers. The numbers of ganglia and ganglion cells
with CRF-IR are low in the stomach and increase progres- several short lamellar dendrites). This kind of morphology
sively in the aboral direction with the largest numbers is characteristic for secretomotor/vasodilator, musculomo-
appearing in the distal colon.112 tor, and interneurons in the ENS.112 Less than 5% of CRF
Ninety-five percent of CRF immunoreactive neurons in immuno-positive myenteric neurons have Dogiel type II
the myenteric plexus have Dogiel type I morphology (i.e., morphology (i.e., a relatively large cell body with two or
an oval or round cell body with a single long process and more long processes; Figure 75.8). Neurons with Dogiel
Chapter  |  75  Enteric Neurobiology of Stress 2009

type II morphology belong to a special class of ENS the cholinergic neurons and 57% of CRF immunoreactive
interneurons, called AH-type, based on electrophysiologi- neurons that express the cholinergic code, suggesting that
cal properties that include long-lasting after-hyperpolariza- CRF might be released with acetylcholine when the neu-
tion associated with discharge of action potentials (Figure rons fire in the myenteric plexus in guinea pigs. Substance
75.6; see Chapter 21).65 About half of the CRF immuno- P is a marker for a subpopulation of excitatory musculo-
reactive neurons with Dogiel type I morphology in the motor neurons. CRF is expressed by 4% of substance P
myenteric plexus project their axons in the oral direction immunoreactive neurons and 55% of CRF immunoreac-
along the intestine and the other half project in the aboral tive neurons express the substance P code, suggesting that
direction. Divisions of projections in this manner would be CRF might be released with substance P when these exci-
expected if the oral projections were from excitatory mus- tatory musculomotor neurons fire. Immunoreactivity for
culomotor neurons and those going in the aboral direction nitric oxide synthase (NOS) is a marker code for inhibi-
were inhibitory musculomotor neurons. tory musculomotor neurons in the myenteric plexus. CRF
CRF is expressed most strongly in the submucosal is expressed by 5% of the neurons with NOS and 38% of
plexus. The proportion of the total number of ganglion CRF immunoreactive neurons express the (NO) code, sug-
cells expressing CRF in the submucosal plexus is about gesting that CRF might be co-released with the inhibitory
14-fold greater than the proportion in the myenteric musculomotor transmitter, NO, when the neurons fire.
plexus. Virtually all of the submucosal ganglia contained at In terms of chemical codes, morphology, and direc-
least one CRF-immunoreactive neuronal cell body. When tionality of axonal projections, a majority of the myenteric
evaluated as a percent of the total submucosal neuronal neurons, which express CRF, fit the profile for excitatory
population, similar percentages of CRF-immunoreactive or inhibitory musculomotor neurons. The neurons with
neurons are found in the duodenum, jejunum, ileum, and ChAT and substance P coexpression correspond to a sub-
proximal and distal colon in guinea pigs.112 All CRF- group of cholinergic/substance Pergic neurons, with single
immunoreactive submucosal neurons have Dogiel type I axonal morphology and orally projecting axons, identified
uniaxonal morphology. Dogiel type II submucosal neurons as excitatory musculomotor neurons to the circular muscle
do not express CRF. coat.114,115 These neurons may be responsible for some of
the CRF-positive nerve fibers that can be seen running par-
allel to the muscle fibers in the circular muscle layer of the
75.5.7  Distribution of CRF in Relation to
small and large intestine where they release their excitatory
ENS Chemical Codes neurotransmitters at neuromuscular junctions.98 The neu-
ENS neurons are classified by their shapes, function, and rons with NOS immunoreactivity correspond to a subset of
histochemical/immunohistochemical staining properties inhibitory musculomotor neurons with aboral projections to
and other characteristic features including the structures the circular muscle and descending interneurons involved
they innervate (e.g., longitudinal or circular muscle coats in local motility reflexes.114,115 It might be significant that
of the intestinal muscularis externa or intestinal secre- the CRF percentages are similar for motor neurons that
tory glands), the transmitters they release, and the synap- release excitatory or inhibitory neurotransmitters at intesti-
tic inputs they receive. Impressive progress has occurred nal neuromuscular junctions. This suggests that CRF might
in the last 25 years in terms of success in combining the be co-released with acetylcholine, substance P, or NO from
various neuronal biomarkers into individual profiles that subgroups of excitatory or inhibitory musculomotor neu-
distinguish each functionally identified neuronal class rons. Evidence that this could be involved in generation
in guinea pig small intestinal ENS (see Chapter 19).113 of motility patterns comes from studies in which periph-
Double labeling immunohistochemistry provides an effec- eral administration of CRF stimulates propulsive motility
tive means for identification of chemical biomarker codes in the colon as reflected by elevated contractile activity,
for subpopulations of CRFergic neuronal profiles (Figure accelerated transit, and ejection of fecal pellets in rats.116
75.5). The distribution of immunoreactive chemical codes Nevertheless, how this might relate to association of CRF
that are expressed with CRF in the myenteric and submu- and stress-induced changes in motility remains unresolved.
cosal plexuses of guinea pig ileum appear in Figure 75.8. Aside from acetylcholine, substance P, and NO, addi-
tional established chemical codes in the myenteric plexus
include somatostatin, serotonin, calbindin, and calretinin.
75.5.7.1  Myenteric Plexus
Small diameter uniaxonal neurons that express calretinin
Immunoreactivity for choline acetyltransferase (ChAT) make up about one-fourth of the neurons in the myenteric
is a code for neurons that express and probably release plexus of guinea pig small intestine.117,118 These neurons
acetylcholine as a neurotransmitter. It is a biomarker for are also cholinergic and identified as excitatory motor neu-
excitatory musculomotor neurons and two subsets of rons that innervate and contract the longitudinal muscle
secretomotor neurons. CRF is expressed by 4.5%% of coat when they fire. Calbindin immunoreactivity identifies
2010 SECTION  |  VI  Consequences of Disregulated Physiology

ENS neurons with multipolar Dogiel type II morphology and continue on through several ganglia without forming
and AH-type electrophysiological behavior (see Chapters contacts with other ganglion cells.121 Immunoreactive CRF
19, 21, 22, 81). Somatostatin immunoreactivity marks a fibers in the interganglionic fiber tracts are most likely pro-
group of descending interneurons in the guinea pig mye- jections from CRF/VIP secretomotor/vasodilator neurons,
nteric plexus, and serotonin is expressed by a different because they appear alongside submucosal arterioles and
population of descending interneurons.113 None of these both VIP and CRF persist in isolated submucosal prepa-
four subpopulations express CRF. rations after degeneration of extrinsic innervation in orga-
notypic culture.112 Persistence of CRF fibers in isolated
submucosal preparations after several days in culture indi-
75.5.7.2  Submucosal Plexus
cates that few, if any, of the CRF neurons in the myenteric
CRF is expressed exclusively by neuronal cell bodies plexus project their axons to the submucosal plexus.
that coexpress vasoactive intestinal peptide (VIP) in the
submucosal plexus (Figures 75.7 and 75.8). VIP is an
75.6  CRF RECEPTORS
established neurotransmitter released by non-cholinergic
secretomotor neurons at neuroepithelial junctions.119 CRF CRF and CRF-like peptides exert their signaling functions
immunoreactivity is not found in submucosal neurons through specific G-protein-coupled receptors. Two types of
that express ChAT, a neurochemical code for cholinergic CRF receptors have been cloned, characterized, and named
secretomotor innervation of the mucosal epithelium. VIP CRF1R and CRF2R.122 Each receptor is associated with
is a chemical code for secretomotor/vasodilator neurons distinctly different physiological events that reflect unique
(Figure 75.7).112,119 Colocalization with VIP suggests that distributions in various tissues and differing profiles of
CRF is released together with VIP when secretomotor pharmacological specificity. The mRNA transcript for the
neurons fire. Like the implications for coexpression with CRF1R is widely expressed in the mammalian brain and
excitatory and inhibitory neurotransmitters in musculomo- pituitary gland and reduced levels are found in peripheral
tor neurons to circular muscle, coincident release at neu- organs, including testis, ovaries, and adrenal glands.123–125
roepithelial and neurovascular junctions might function to CRF2R is expressed in the brain and peripheral tissues,
modulate the responses of the secretory glands and blood including heart, lung, gastrointestinal tract, skeletal mus-
vessels individually in response to input from a single cle, testis, and ovaries.126–128 CRF1R and CRF2R are
population of secretomotor/vasodilator neurons. In view of expressed in the intestinal tract where mRNA transcripts
established significance of CRF in stress-related opening and Western blot protein expression can be found for crude
of the mucosal barrier, it might be questioned if VIP/CRF extracts from rat colon.110,129 CRF1R and CRF2R immu-
co-release is significant in terms of secretomotor influence noreactivity is colocalized with neuron specific enolase
on mucosal permeability. and neurofilament protein in the rat colon. This was a
Cholinergic secretomotor neurons express immuno- seminal indication (seen in 2004) that CRF receptors are
reactivity for neuropeptide Y (NPY) in the submucosal expressed by neurons in the ENS.111a
plexus, while cholinergic secretomotor/vasodilator neurons CRF binds with high affinity to CRF1R and with lower
express calretinin.113,115 Substance P and NeuN are marker affinity to CRF2R. Urocortin 1 binds with high affinity to
codes for submucosal neurons with Dogiel type II mor- CRF1R and CRF2R.130 Urocortin 2 and urocortin 3 are
phology and AH-type electrical and synaptic behavior.120 putative endogenous ligands for CRF2R based on exclusive
None of these neuronal populations in the submucosal binding to CRF2R and absence of CRF1R binding.84,85,131
plexus express CRF. Currently available, selective CRF receptor antagonists
There are far fewer CRF-positive nerve fibers in sub- now enable quantitative investigational identification of
mucosal ganglia than in ganglia in the myenteric plexus. CRF receptor types involved in specific actions of CRF
There are no CRF fibers in over half of the ganglia in the at the tissue, organ, and whole animal levels of biologi-
submucosal plexus.112 On the other hand, CRF nerve fib- cal organization. Earlier receptor antagonists were CRF
ers are abundant in the interganglionic fiber tracts. It family peptides with modified amino acid sequences that
seems that rather than forming synapses within the gan- were eventually followed by development of non-peptide
glia, most of the CRF neurons appear to send their axons antagonists that avoided many of the pitfalls associated
outside their home ganglia to release neurotransmitters at with peptides, especially in human therapeutics. Among
synapses with neurons and at neuroeffector junctions. The the currently available antagonists for research are the pep-
microanatomy of VIP immunoreactive neurons in the sub- tides, astressin, and α-helical CRF (9-41), which are non-
mucosal plexus is essentially the same as for the neurons selective CRF antagonists that have high affinity for both
that contain CRF. VIP and CRF neurons have few axonal CRF1R and CRF2R.132 Non-peptide CRF antagonists,
projections to synapses with other neurons inside their CP154526, NBI27914, NBI35965, and antalarmin, each
ganglia of residence. Instead, their axons leave the ganglia have strong selectivity for CRF1R; whereas the peptidergic
Chapter  |  75  Enteric Neurobiology of Stress 2011

antagonists antisauvagine-30 and the long-acting analog (A) Myenteric (B) Submucosal
astressin2-B have high selectivity for CRF2R.10,132 Plexus Plexus

Calbindin--------------------86% Calbindin------------------100%
75.6.1  CRF Receptor Expression in Relation Calretinin ---------------------0% Calretinin---------------------0%
to ENS Codes ChAT ------------------------74% ChAT -----------------------60%
Early results from work on CRF receptor gene expres- NOS ---------------------------0% NPY -------------------------34%
sion suggested that CRF2R is the predominant receptor
NPY ---------------------------0% Somatostatin --------------29%
subtype expressed in the gastrointestinal tract and that the
CRF1R transcript is expressed mainly in the brain and pitu- Somatostatin ----------------0% Substance P -------------100%
itary gland.123,124,126,127 Immunoreactivity for CRF1R and VIP -----------------------------0% VIP ----------------------------0%
CRF2R was reported, subsequently, for the upper gastroin-
testinal tract.111a CRF1R is found in a narrow zone in the FIGURE 75.9  Proportions of each neurochemical neuronal type
upper oxyntic gland region of the gastric corpus. CRF2R that express immunoreactivity for CRF1R in guinea pig small
is localized to gastric parietal cells and to enteroendocrine bowel.  (A) In the myenteric plexus, only calbindin-positive neurons
cells in the stomach and duodenal mucosa and to the lumi- (i.e., AH-type) and cholinergic (ChAT-positive) neurons express CRF1R.
(B) In the submucosal plexus, all calbindin- and substance P-positive
nal surfaces of the crypts and around blood vessels in rat
neurons express immunoreactivity for CRF1R. VIP-positive neurons do
colon. Double staining finds CRF1R coexpressed with the not express CRF1R. (Adapted from Liu et al. 112.)
neuronal markers, neuron-specific enolase and neurofila-
ment protein, in the myenteric and submucosal plexuses.111a
Immunofluorescence, RT-PCR, and Western blot evidence 75.6.2.1  Myenteric Plexus
now suggests that CRFR1 is the predominant stress-asso- Immunoreactivity for the calcium-binding protein, calbi-
ciated receptor type expressed by neurons in guinea pig ndin, is a marker for neurons with AH-type electrophysi-
and rat ENS. CRFR2 stimulation, in rat colon, offsets to ological behavior and multipolar Dogiel II morphology
a degree stress-evoked changes mediated by CRFR1.111b in the ENS.113,114,133 Most calbindin neurons (86%) in
Moreover, RT-PCR reveals mRNA transcripts for CRF1R the myenteric plexus express CRF1R (Figures 75.9 and
gene expression in both myenteric and submucosal plex- 75.10). This is consistent with electrophysiological results
uses.98,111,129 Immunoreactivity for CRF1R is reported to be showing that CRF evokes slowly activating membrane
expressed also by mucosal epithelial and enteroendocrine depolarization and action potential discharge in AH-type
cells in rat,110,129 but not guinea pig.98 In general, immuno- neurons with Dogiel II morphology (Figure 75.6). CRF1R
histochemical localization of CRFR1 protein is consistent is expressed by 74% of all cholinergic ENS neurons in
with RT-PCR and Western blot evidence for expression of the myenteric plexus, as revealed by colocalization with
CRF1R by ENS neurons. Moreover, pharmacological data ChAT. Based on ChAT expression, at least ten differ-
obtained with selective CRF receptor antagonists in electro- ent functionally identified subpopulations of guinea pig
physiological studies (see the following section) show the myenteric neurons can release acetylcholine at neuron-to-
excitatory actions of CRF and urocortin 1 on ENS neurons neuron synapses or at neuroeffector junctions; included are
to be mediated exclusively by CRF1R.98 ascending and descending interneurons, calbindin-positive
AH-type neurons, and excitatory motor neurons to the
musculature and mucosal secretory glands.113
75.6.2  Double-labeling Immunofluorescence
Intestinal smooth muscle, in the guinea pig, does not
Double-labeling immunohistochemical methods are pro- express CRF1R, which suggests that CRF1R does not
ductive for identification of the classes of ENS neurons mediate any direct myogenic effect of musculomotor CRF
that express CRF1R (Figure 75.5). Figure 75.9 shows release on contractility and therefore propulsive or mixing
data for the distribution of CRF1R in relation to CRF and motility.105 Moreover, finding of a lack of expression of
some of the known chemical codes for myenteric and sub- CRF1R by interstitial cells of Cajal implies that alteration
mucosal plexuses in guinea pig small intestine. CRF1R is of intestinal motor responses seen during exogenous appli-
found in neurons that are neighbors to neurons that express cations of CRF or during exposure of an animal to stress
CRF in both the myenteric and submucosal plexuses. does not involve any action of CRF at CRF1R receptors on
However, immunoreactivity for CRF is never found in neu- functions of interstitial cells of Cajal.105 On the other hand,
rons that express CRF1R, which implies that specific sub- exposure of intestinal preparations to CRF in vitro evokes
sets of neurons express CRF as a neurotransmitter, while excitatory junction potentials in the circular muscle of
other specific subsets express the postsynaptic receptor for guinea pig small intestine that reflect input from excitatory
CRF neurotransmission (Figure 75.10). musculomotor neurons.134 This probably reflects activation
2012 SECTION  |  VI  Consequences of Disregulated Physiology

Positive Feed-forward
Synaptic Network
Directions of Transmission

(CRF)

Calbindin-Positive
AH-Dogiel II Neurons

(CRF)

Non-Cholinergic Secretomotor/
Vasodilator Neurons
(VIP-Positive Neurons)

Secretomotor
Neurons

Secretory Glands

FIGURE 75.10  Calbindin-positive AH-Dogiel morphological type II interneurons are networked into positive feed-forward synaptic cir-
cuits.  Feed-forward synaptic excitation is a mechanism for rapid initiation of synchronous discharge in multiple neural elements of the circuit. Output
from the feed-forward circuit drives populations of secretomotor neurons, which in turn drive the behavior of the secretory epithelium and vasculature.
Positive feed-forward synaptic networks synchronize simultaneous firing of large numbers of motor neurons to ensure coordination of secretion around
and along an extended length of bowel. VIP-positive, non-cholinergic secretomotor vasodilator neurons are the only neurons that express CRF in the
submucosal plexus. When they fire, these neurons are postulated to release CRF as an excitatory neurotransmitter at postsynaptic CRF1R receptors
expressed on calbindin-positive interneurons in positive feed-forward synaptic networks that drive mucosal secretion.

of CRF1R on cholinergic and/or substance Pergic muscu- Calbindin-positive neurons with Dogiel II multipolar
lomotor neurons. morphology and AH-type electrophysiological behavior
innervate and activate subsets of secretomotor neurons.
A single nicotinic synapse connects AH-type axons with
75.6.2.2  Submucosal Plexus
secretomotor neurons.121,135–137
Nearly all of the neurons in the submucosal plexus of All calbindin-containing submucosal neurons with
guinea pig small intestine fit into one or the other of four Dogiel II morphology, like their counterparts in the mye-
neurochemical classes (Figure 75.7).112,113 These include: nteric plexus, express CRF1R (Figure 75.9). CRF1R
(1) neurons with multipolar Dogiel II morphology and appears also to be expressed by cholinergic/NPY secreto-
coexpressed immunoreactivity for calbindin, ChAT, and motor neurons that do not innervate the vasculature. On
SP; (2) neurons with a single axon and immunoreactiv- the other hand, the receptor is not expressed by VIPergic,
ity for calretinin and ChAT; (3) neurons with single axon non-cholinergic secretomotor/vasodilator neurons nor is it
morphology that coexpress NPY and ChAT; and (4) single expressed by cholinergic/calretinin-containing secretomo-
axonal neurons immunoreactive for VIP. Three of these tor/vasodilator neurons.
subclasses are motor neurons that stimulate mucosal secre- Calbindin-positive AH-type neurons are interconnected
tion when they fire. The first are VIP-expressing, non- to form positive feed-forward circuits in which firing of
cholinergic secretomotor/vasodilator neurons that innervate one member of the circuit evokes simultaneous firing in
the mucosal epithelium and small arterioles. Second are all neighboring members (Figure 75.10). This is a form of
cholinergic secretomotor/vasodilator neurons that contain synaptic connectivity in which the neurons of the circuit
calretinin, and the third subclass includes cholinergic secre- make recurrent excitatory synaptic connections with each
tomotor neurons that contain NPY and do not innervate the other.138–140 Feed-forward connectivity in the circuit causes
vasculature (Figure 75.7). excitation to build rapidly to firing threshold in each of the
Chapter  |  75  Enteric Neurobiology of Stress 2013

neurons that make up the circuit. Rapid buildup of firing in Outside the ENS, CRF is expressed near the base of
the individually interconnected neurons in the circuit ensures crypts in human colon and coexpressed with serotonin in
simultaneous activation of the whole network around the mucosal enterochromaffin cells.93 A large proportion of
circumference and along the length of a segment of bowel. calbindin-positive, AH-Dogiel II neurons in the myenteric
Neurons in the circuit have excitatory synaptic connections plexus express CRF1R, which might become a target for
with adjacent motor neurons. In this arrangement, output of CRF release from enterochromaffin cells (Figure 75.9).
the circuit functions to simultaneously activate large pools Neurites from AH-Dogiel II neurons project downward
of motor neurons around the circumference and up and through the circular muscle coat to end in submucosal/
down an intestinal segment. When submucosal secretomo- mucosal regions.144 The terminals in the submucosa
tor neurons are activated in this manner, the effect is to dis- express serotonergic 5-HT3 receptors. Stimulation by
tribute secretion of electrolytes, H2O, and mucin uniformly 5-HT, released from enterochromaffin cells, fires the ter-
around the circumference and along a length of bowel.141 minals.147 Once initiated, the action potentials propagate
The observation that calbindin-containing neurons (AH-type along the neurites in a retrograde manner to the cell body
neurons) express receptors for CRF, but no CRF, while VIP- and may fire the cell body depending on its synaptic/para-
expressing, non-cholinergic secretomotor/vasodilator neu- crine input and level of excitability when they arrive (see
rons express CRF, but no receptors for CRF, suggests that Chapter 19). Based on its immunohistochemical locali-
these neurons might be the source of CRF synaptic input to zation, CRF1R is expected to be operative alongside the
AH-type neurons that make up the feed-forward circuit that 5-HT3 receptors at the AH-Dogiel II terminals.
drives the cholinergic secretomotor/vasodilator neurons that CRF also is released locally in sites of inflammation in
contain calretinin and the cholinergic secretomotor neurons animal models and inflamed joints of patients with rheuma-
that contain NPY and do not innervate the vasculature. toid arthritis, which suggests that CRF might have proin-
flammatory actions in the gut.145–148 Immunoreactivity
for CRF expression is significantly increased in mucosal
75.7  SOURCES FOR CRF
inflammatory cells in ulcerative colitis and is correlated
VIP and acetylcholine are firmly established as submu- with the intensity of the inflammation.149 Expression is
cosal neurotransmitters at neuroepithelial and neurovas- restricted to inflammatory cells and is not increased in ente-
cular junctions that increase blood flow in concert with rochromaffin cells or mucosal epithelial cells in ulcerative
stimulation of mucosal secretion.119,142 The CRF hypoth- colitis. The association of CRF with inflammatory bowel
esis for stress-evoked stimulation of mucosal secretion and disease is reminiscent of evidence that subepithelial mast
increased barrier permeability is supported also by firm cells in mucosal biopsies from human colon express immu-
evidence. Localization of CRF1R in the submucosal ENS noreactivity for CRF1R and CRF2R, and our preliminary
suggests that the neuronal site of action for CRF-evoked results show coexpression of CRF1R and CRF2R recep-
effects of stress-induced diarrhea and opening of the tors by mast cells in guinea pig colon and human jejunum
mucosal barrier to large molecules is restricted to cholin- and CRF-evoked release of histamine from mast cells in
ergic/NPY secretomotor neurons that do not innervate guinea pig colon.102,150 Release of chemoattractant factors
the vasculature. That this might be the case is suggested for inflammatory/immune cells occurs in concert with the
by reports that pretreatment with the muscarinic receptor collection of other mast cell mediators in response to CRF
antagonist, atropine, suppresses stress-induced stimula- input. This might well be a significant factor in the associa-
tion of mucosal secretion and increased permeability of the tion of mucosal inflammation with stress (see Figure 75.2).
mucosal barrier in animals12,101,102,143 and humans.99
The expression of receptors for CRF by AH-Dogiel
75.7.1  CRF Action on ENS Neurons
type II neurons, putative cholinergic secretomotor neu-
rons, and enteric mast cells raises a question of the ori- Intracellular recording with “sharp” microelectrodes is
gin of the CRF that could act to stimulate the receptors. used to study CRF1R receptor pharmacology in neurons
Based on the immunohistochemical localization, cholin- in the ENS (Figure 75.5).98 Electrophysiological actions
ergic and substance P excitatory musculomotor neurons of CRF on ENS neurons are essentially the same for the
or nitrergic inhibitory musculomotor neurons could be a guinea pig small and large intestine. Application of CRF
source of CRF release. Calbindin-positive AH-Dogiel type in vitro evokes, with an EC50 of 25 nM, slowly activat-
II neurons, which comprise about 20% of the neurons in ing slow EPSP-like depolarizing responses associated
the myenteric plexus,112 do not express CRF and therefore with increased excitability in myenteric neurons (Figure
are not a likely source. In the submucosal plexus, the only 75.6). CRF stimulates excitability in this manner for both
apparent neuronal source for CRF release is the population neurons with single axons and S-type electrophysiologi-
of VIPergic, non-cholinergic secretomotor/vasodilator neu- cal behavior and Dogiel type II multipolar neurons with
rons (Figures 75.8 and 75.10). AH-type electrophysiological behavior. An increase in
2014 SECTION  |  VI  Consequences of Disregulated Physiology

membrane input resistance attributed to closure of K   3. Collins SM, Barbara G, Vallance B. Stress, inflammation and the irrita-
channels is associated with the depolarizing responses in ble bowel syndrome. Can J Gastroenterol. 1999;13(suppl A):47A–49A.
AH-type neurons.151 Changes in input resistance in S-type   4. Naliboff BD, Mayer M, Fass R, Fitzgerald LZ, Chang L, Bolus R,
et al. The effect of life stress on symptoms of heartburn. Psychosom
neurons are inconsistent, with input resistance decreased
Med. 2004;66:426–434.
or unchanged. The action of CRF reflects direct activa-
  5. Peck OC, Wood JD. Brain-gut interactions in ulcerative colitis.
tion of receptors on the neuron, rather than indirect syn- Gastroenterology. 2000;118:807–808.
aptic input from CRF-sensitive neurons elsewhere in the   6. Wood JD, Peck OC, Tefend KS, Stonerook MJ, Caniano DA,
preparation, because blockade of synaptic transmission by Mutabagani KH, et  al. Evidence that colitis is initiated by environ-
tetrodotoxin or lowering of Ca2 and elevation of Mg2 mental stress and sustained by fecal factors in the cotton-top tamarin
in the bathing medium does not change the CRF-evoked (Saguinus oedipus). Dig Dis Sci. 2000;45:385–393.
depolarizing responses.98 Tachyphylaxis to CRF occurs   7. Fone DR, Horowitz M, Maddox A, Akkermans LM, Read NW, Dent
rapidly in S-type neurons (Figure 75.6) and does not occur J. Gastroduodenal motility during the delayed gastric emptying
in AH-type neurons. This may be significant in terms of induced by cold stress. Gastroenterology. 1990;98:1155–1161.
the putative neurotransmitter role for CRF in the model in   8. Tache Y, Martinez V, Million M, Rivier J. Corticotropin-
releasing factor and the brain-gut motor response to stress. Can J
Figure 75.10.
Gastroenterol. 1999;13(suppl A):18A–25A.
The excitatory responses to CRF in ENS neurons are
  9. Tache Y, Martinez V, Million M, Wang L. Stress and the gastroin-
mediated exclusively by CRF1R. Astressin, a non-selective testinal tract III. Stress-related alterations of gut motor function:
CRF1R/CRF2R receptor antagonist, or the selective role of brain corticotropin-releasing factor receptors. Am J Physiol
CRF1R receptor antagonist, NBI 27914, both block the Gastrointest Liver Physiol. 2001;280:G173–G177.
excitatory responses to CRF. The CRF2R antagonist, anti- 10. Million M, Maillot C, Saunders P, Rivier J, Vale W, Tache Y. Human
sauvagine-30, has been tested and does not suppress or urocortin II, a new CRF-related peptide, displays selective CRF(2)-
otherwise change depolarizing responses to CRF, which is mediated action on gastric transit in rats. Am J Physiol Gastrointest
consistent with other evidence that CRF1R mediates exci- Liver Physiol. 2002;282:G34–G40.
tatory responses to CRF in ENS neurons in guinea pig. 11. Saunders PR, Maillot C, Million M, Tache Y. Peripheral corticotro-
pin-releasing factor induces diarrhea in rats: role of CRF1 receptor in
fecal watery excretion. Eur J Pharmacol. 2002;435:231–235.
75.7.2  CRF–Mast Cell Connection 12. Santos J, Saunders PR, Hanssen NP, Yang PC, Yates D, Groot
Evidence is accumulating that enteric mast cells are inner- JA, et  al. Corticotropin-releasing hormone mimics stress-induced
colonic epithelial pathophysiology in the rat. Am J Physiol. 1999;
vated by CRFergic ENS neurons. Firing of the neurons
277:G391–G399.
and release of CRF at postsynaptic receptors on the mast
13. Fukudo S, Nomura T, Hongo M. Impact of corticotropin-releasing
cells is expected to release a deluge of mast cell-signaling hormone on gastrointestinal motility and adrenocorticotropic hor-
molecules. These may be mediators stored in granules mone in normal controls and patients with irritable bowel syndrome.
(e.g., histamine, proteases, and bradykinin) or obtained by Gut. 1998;42:845–849.
synthesis from membrane lipids (e.g., prostaglandins and 14. Rao SS, Hatfield RA, Suls JM, Chamberlain MJ. Psychological and
platelet-activating factor). This expectation is reinforced physical stress induce differential effects on human colonic motility.
by findings of immunoreactivity for CRF1R and CRF2R Am J Gastroenterol. 1998;93:985–990.
receptor subtypes expressed by subepithelial mast cells in 15. Payne A, Blanchard EB, Holt CS, Schwarz SP. Physiological reac-
mucosal biopsies from human colon102 and by mast cells tivity to stressors in irritable bowel syndrome patients, inflammatory
in guinea pig colon and human jejunum.150 Application of bowel disease patients and non-patient controls. Behav Res Ther.
1992;30:293–300.
CRF to segments of guinea pig and human small intestine
16. Smith SM, Vale WW. The role of the hypothalamic-pituitary-adrenal
in vitro evokes release of mast cell histamine and protease
axis in neuroendocrine responses to stress. Dialogues Clin Neurosci.
II into the bathing medium as determined by ELISA.150 2006;8:383–395.
These findings are an incentive for adding innervation of 17. Wood JD. Neurotransmission at the interface of sympathetic and
enteric mast cells by CRF secretomotor neurons to a pos- enteric divisions of the autonomic nervous system. Chin J Physiol.
tulated circuit diagram for CRF neurotransmission in the 1999;42:201–210.
submucosal plexus in a scenario for stress, ENS, and mast 18. Wood JD. The synaptic interface between the sympathetic and
cell interaction (see Figure 75.2). The outcome of which enteric divisions of the autonomic nervous system. In: Chey WY,
results in stimulation of neurogenic secretion, compromise ed. Functional Bowel Disorders. New York: Raven Press; 1982. p.
of the mucosal barrier, and enhanced propulsive motility. 87–91.
19. North RA, Surprenant A. Inhibitory synaptic potentials result-
ing from alpha 2-adrenoceptor activation in guinea-pig submucous
REFERENCES
plexus neurones. J Physiol. 1985;358:17–33.
  1. Mayer EA. The neurobiology of stress and gastrointestinal disease. 20. Wood JD, Mayer CJ. Adrenergic inhibition of serotonin release from
Gut. 2000;47:861–869. neurons in guinea pig Auerback’s plexus. J Neurophysiol. 1979;
  2. Mayer EA. Psychological stress and colitis. Gut. 2000;46:595–596. 42:594–603.
Chapter  |  75  Enteric Neurobiology of Stress 2015

21. Surprenant A, North RA. Mechanism of synaptic inhibition by 41. Baird AW, Cuthbert AW. Neuronal involvement in type 1 hyper-
noradrenaline acting at alpha 2-adrenoceptors. Proc R Soc Lond B sensitivity reactions in gut epithelia. Br J Pharmacol. 1987;92:
Biol Sci. 1988;234:85–114. 647–655.
22. Kamath PS, Phillips SF, O’Connor MK, Brown ML, Zinsmeister AR. 42. McKay DM, Perdue MH. Intestinal epithelial function: the case for
Colonic capacitance and transit in man: modulation by luminal con- immunophysiological regulation. Implications for disease (2). Dig
tents and drugs. Gut. 1990;31:443–449. Dis Sci. 1993;38:1735–1745.
23. Drossman DA. The functional gastrointestinal disorders and the 43. Raghavan M, Bjorkman PJ. Fc receptors and their interactions
Rome III process. Gastroenterology. 2006;130:1377–1390. with immunoglobulins. Annu Rev Cell Dev Biol. 1996;12:
24. Ringel Y, Sperber AD, Drossman DA. Irritable bowel syndrome. 181–220.
Annu Rev Med. 2001;52:319–338. 44. Vergnolle N. Proteinase-activated receptors (PARs) in infection
25. Pan G, Lu S, Ke M, Han S, Guo H, Fang X. Epidemiologic study of and inflammation in the gut. Int J Biochem Cell Biol. 2008;40:
the irritable bowel syndrome in Beijing: stratified randomized study 1219–1227.
by cluster sampling. Chin Med J (Engl). 2000;113:35–39. 45. MacQueen G, Marshall J, Perdue M, Siegel S, Bienenstock J.
26. Gwee KA, Wee S, Wong ML, Png DJ. The prevalence, symptom Pavlovian conditioning of rat mucosal mast cells to secrete rat mast
characteristics, and impact of irritable bowel syndrome in an asian cell protease II. Science. 1989;243:83–85.
urban community. Am J Gastroenterol. 2004;99:924–931. 46. Santos J, Saperas E, Nogueiras C, Mourelle M, Antolin M, Cadahia
27. Quigley EM. Changing face of irritable bowel syndrome. World J A, et  al. Release of mast cell mediators into the jejunum by cold
Gastroenterol. 2006;12:1–5. pain stress in humans. Gastroenterology. 1998;114:640–648.
28. Thompson WG, Longstreth GL, Drossman DA, Heaton K, Irvine EJ, 47. Williams RM, Berthoud HR, Stead RH. Vagal afferent nerve
Muller-lissner S. Functional bowel disorders and functional abdomi- fibres contact mast cells in rat small intestinal mucosa.
nal pain. In: Drossman DA, Corazziari E, Talley NJ, Thrompson WG, Neuroimmunomodulation. 1997;4:266–270.
Whitehead WE, eds. The Functional Gastrointestinal Disorders. 48. Gottwald TP, Hewlett BR, Lhotak S, Stead RH. Electrical stimu-
McLean VA: Degnon Associates; 2000. p. 351–432. lation of the vagus nerve modulates the histamine content of mast
29. Drossman DA, Creed FH, Olden KW, Svedlund J, Toner BB, cells in the rat jejunal mucosa. Neuroreport. 1995;7:313–317.
Whitehead WE. Psychosocial aspects of the functional gastrointesti- 49. Santos J, Saperas E, Mourelle M, Antolin M, Malagelada JR.
nal disorders. Gut. 1999;45(suppl 2):II25–II30. Regulation of intestinal mast cells and luminal protein release by
30. Ringel Y, Drossman DA, Turkington TG, Bradshaw B, Hawk TC, cerebral thyrotropin-releasing hormone in rats. Gastroenterology.
Bangdiwala S, et  al. Regional brain activation in response to rectal 1996;111:1465–1473.
distension in patients with irritable bowel syndrome and the effect of 50. Gue M, Del Rio-Lacheze C, Eutamene H, Theodorou V, Fioramonti
a history of abuse. Dig Dis Sci. 2003;48:1774–1781. J, Bueno L. Stress-induced visceral hypersensitivity to rectal disten-
31. Drossman DA, Ringel Y, Vogt BA, Leserman J, Lin W, Smith JK, sion in rats: role of CRF and mast cells. Neurogastroenterol Motil.
et al. Alterations of brain activity associated with resolution of emo- 1997;9:271–279.
tional distress and pain in a case of severe irritable bowel syndrome. 51. Tache Y, Million M. Central corticotropin-releasing factor and the
Gastroenterology. 2003;124:754–761. hypothalamic-pituitary-acrenal axis in gastrointestinal physiology.
32. Levy RL, Olden KW, Naliboff BD, Bradley LA, Francisconi C, In: Johnson LR, Barrett KE, Ghishan FK, Merchant JL, Said HM,
Drossman DA, et al. Psychosocial aspects of the functional gastroin- Wood JD, eds. Physiology of the Gastrointestinal Tract. San Diego:
testinal disorders. Gastroenterology. 2006;130:1447–1458. Academic Press; 2006. p. 791–816.
33. Frieling T, Cooke HJ, Wood JD. Neuroimmune communication in the 52. Joseph SA, Pilcher WH, Knigge KM. Anatomy of the corticotropin-
submucous plexus of guinea pig colon after sensitization to milk anti- releasing factor and opiomelanocortin systems of the brain. Fed
gen. Am J Physiol. 1994;267:G1087–G1093. Proc. 1985;44:100–107.
34. Frieling T, Palmer JM, Cooke HJ, Wood JD. Neuroimmune commu- 53. Berridge CW, Dunn AJ. CRF and restraint-stress decrease explora-
nication in the submucous plexus of guinea pig colon after infection tory behavior in hypophysectomized mice. Pharmacol Biochem
with Trichinella spiralis. Gastroenterology. 1994;107:1602–1609. Behav. 1989;34:517–519.
35. Liu S, Hu HZ, Gao N, Gao C, Wang G, Wang X, et al. Neuroimmune 54. Martinez V, Tache Y. CRF1 receptors as a therapeutic target for irri-
interactions in guinea pig stomach and small intestine. Am J Physiol table bowel syndrome. Curr Pharm Des. 2006;12:4071–4088.
Gastrointest Liver Physiol. 2003;284:G154–G164. 55. Chatzaki E, Minas V, Zoumakis E, Makrigiannakis A. CRF recep-
36. Wood JD. Enteric neuroimmunophysiology and pathophysiology. tor antagonists: utility in research and clinical practice. Curr Med
Gastroenterology. 2004;127:635–657. Chem. 2006;13:2751–2760.
37. Cowles VE, Sarna SK. Effect of T. spiralis infection on intesti- 56a. Stengel A, Tache Y. Corticotropin-releasing factor signaling and
nal motor activity in the fasted state. Am J Physiol. 1990;259: visceral response to stress. Exp Biol Med (Maywood). 2010;235:
G693–G701. 1168–1178.
38. Broaddus RR, Castro GA. Mast cell-mediated colonic immune 56b. Wang L, Million M, Rivier J, Rivier C, Craft N, Stenzel-Poore MP,
function and its inhibition by dietary aspirin in mice infected with et al. CRF receptor antagonist astressin-B reverses and prevents alo-
Trichinella spiralis. Int Arch Allergy Immunol. 1994;105:135–142. pecia in CRF over-expressing mice. PLoS One. 2011;6:e16377.
39. Harari Y, Russell DA, Castro GA. Anaphylaxis-mediated epithe- 57. Yamashita H, Kasai M, Inenaga K. Effects of corticotropin-releas-
lial Cl- secretion and parasite rejection in rat intestine. J Immunol. ing factor on neurons in the hypothalamic paraventricular nucleus in
1987;138:1250–1255. vitro. Brain Res Bull. 1991;27:321–325.
40. Wang YZ, Palmer JM, Cooke HJ. Neuroimmune regulation of colonic 58. Owens MJ, Nemeroff CB. Physiology and pharmacology of cortico-
secretion in guinea pigs. Am J Physiol. 1991;260:G307–G314. tropin-releasing factor. Pharmacol Rev. 1991;43:425–473.
2016 SECTION  |  VI  Consequences of Disregulated Physiology

59. Fox EA, Gruol DL. Corticotropin-releasing factor suppresses the endogenous corticotropin-releasing factor. Gastroenterology. 1988;
afterhyperpolarization in cerebellar Purkinje neurons. Neurosci Lett. 95:1510–1517.
1993;149:103–107. 77. Wittmann T, Crenner F, Angel F, Hanusz L, Ringwald C, Grenier JF.
60. Aldenhoff JB, Gruol DL, Rivier J, Vale W, Siggins GR. Corticotropin Long-duration stress. Immediate and late effects on small and large
releasing factor decreases postburst hyperpolarizations and excites bowel motility in rat. Dig Dis Sci. 1990;35:495–500.
hippocampal neurons. Science. 1983;221:875–877. 78. Kihara N, Fujimura M, Yamamoto I, Itoh E, Inui A, Fujimiya M.
61. Blank T, Nijholt I, Grammatopoulos DK, Randeva HS, Hillhouse Effects of central and peripheral urocortin on fed and fasted gas-
EW, Spiess J. Corticotropin-releasing factor receptors couple to mul- troduodenal motor activity in conscious rats. Am J Physiol Gastroin-
tiple G-proteins to activate diverse intracellular signaling pathways test Liver Physiol. 2001;280:G406–G419.
in mouse hippocampus: role in neuronal excitability and associative 79. Kellow JE, Langeluddecke PM, Eckersley GM, Jones MP, Tennant
learning. J Neurosci. 2003;23:700–707. CC. Effects of acute psychologic stress on small-intestinal motility
62. Borsody MK, Weiss JM. Influence of corticotropin-releasing hor- in health and the irritable bowel syndrome. Scand J Gastroenterol.
mone on electrophysiological activity of locus coeruleus neurons. 1992;27:53–58.
Brain Res. 1996;724:149–168. 80. Gue M, Junien JL, Bueno L. Conditioned emotional response in rats
63. Bowers LK, Swisher CB, Behbehani MM. Membrane and synaptic enhances colonic motility through the central release of corticotropin-
effects of corticotropin-releasing factor on periaqueductal gray neu- releasing factor. Gastroenterology. 1991;100:964–970.
rons of the rat. Brain Res. 2003;981:52–57. 81. Sagami Y, Shimada Y, Tayama J, Nomura T, Satake M, Endo Y, et al.
64. Wood JD. Integrative functions of the enteric nervous system. In: Effect of a corticotropin releasing hormone receptor antagonist on
Johnson LR, Barrett KE, Ghishan FK, Merchant JL, Said HM, colonic sensory and motor function in patients with irritable bowel
Wood JD, eds. Physiology of the Gastrointestinal Tract. San Diego: syndrome. Gut. 2004;53:958–964.
Academic Press; 2006. p. 665–683. 82. Martinez V, Wang L, Rivier J, Grigoriadis D, Tache Y. Central CRF,
65. Wood JD. Cellular neurophysiology of enteric neurons. In: Johnson urocortins and stress increase colonic transit via CRF1 receptors
LR, Barrett KE, Ghishan FK, Merchant. JL, Said HM, Wood JD, eds. while activation of CRF2 receptors delays gastric transit in mice. J
Physiology of the Gastrointestinal Tract. San Diego: Academic Press; Physiol. 2004;556:221–234.
2006. p. 629–664. VI 83. Vaughan J, Donaldson C, Bittencourt J, Perrin MH, Lewis K,
66. Ikeda H, Kusudo K, Ryu PD, Murase K. Effects of corticotropin- Sutton S, et  al. Urocortin, a mammalian neuropeptide related to
releasing factor on plasticity of optically recorded neuronal activity fish urotensin I and to corticotropin-releasing factor. Nature. 1995;
in the substantia gelatinosa of rat spinal cord slices. Pain. 2003;106: 378:287–292.
197–207. 84. Reyes TM, Lewis K, Perrin MH, Kunitake KS, Vaughan J, Arias CA,
67. Fu Y, Pollandt S, Liu J, Krishnan B, Genzer K, Orozco-Cabal L, et  al. Urocortin II: a member of the corticotropin-releasing factor
et  al. Long-term potentiation (LTP) in the central amygdala (CeA) (CRF) neuropeptide family that is selectively bound by type 2 CRF
is enhanced after prolonged withdrawal from chronic cocaine and receptors. Proc Natl Acad Sci USA. 2001;98:2843–2848.
requires CRF1 receptors. J Neurophysiol. 2007;97:937–941. 85. Lewis K, Li C, Perrin MH, Blount A, Kunitake K, Donaldson C,
68. Schierloh A, Deussing J, Wurst W, Zieglgansberger W, Rammes G. et  al. Identification of urocortin III, an additional member of the
Corticotropin-releasing factor (CRF) receptor type 1-dependent mod- corticotropin-releasing factor (CRF) family with high affinity for the
ulation of synaptic plasticity. Neurosci Lett. 2007;416:82–86. CRF2 receptor. Proc Natl Acad Sci USA. 2001;98:7570–7575.
69. Wang HL, Wayner MJ, Chai CY, Lee EH. Corticotrophin-releasing 86. Montecucchi PC, Henschen A. Amino acid composition and
factor produces a long-lasting enhancement of synaptic efficacy in sequence analysis of sauvagine, a new active peptide from the
the hippocampus. Eur J Neurosci. 1998;10:3428–3437. skin of Phyllomedusa sauvagei. Int J Pept Protein Res. 1981;18:
70. Wang HL, Tsai LY, Lee EH. Corticotropin-releasing factor produces 113–120.
a protein synthesis — dependent long-lasting potentiation in dentate 87. Lederis K, Letter A, McMaster D, Moore G, Schlesinger D.
gyrus neurons. J Neurophysiol. 2000;83:343–349. Complete amino acid sequence of urotensin I, a hypotensive and cor-
71. Rebaudo R, Melani R, Balestrino M, Izvarina N. Electrophysiological ticotropin-releasing neuropeptide from Catostomus. Science. 1982;
effects of sustained delivery of CRF and its receptor agonists in hip- 218:162–165.
pocampal slices. Brain Res. 2001;922:112–117. 88. Martinez V, Rivier J, Wang L, Tache Y. Central injection of a new
72. Miyata M, Okada D, Hashimoto K, Kano M, Ito M. Corticotropin- corticotropin-releasing factor (CRF) antagonist, astressin, blocks
releasing factor plays a permissive role in cerebellar long-term CRF- and stress-related alterations of gastric and colonic motor func-
depression. Neuron. 1999;22:763–775. tion. J Pharmacol Exp Ther. 1997;280:754–760.
73. Tache Y, Bonaz B. Corticotropin-releasing factor receptors and 89. Martins JM, Kastin AJ, Banks WA. Unidirectional specific and mod-
stress-related alterations of gut motor function. J Clin Invest. 2007; ulated brain to blood transport of corticotropin-releasing hormone.
117:33–40. Neuroendocrinology. 1996;63:338–348.
74. Bueno L, Fioramonti J. Effects of corticotropin-releasing factor, cor- 90. Petrusz P, Merchenthaler I, Ordronneau P, Maderdrut JL, Vigh S,
ticotropin and cortisol on gastrointestinal motility in dogs. Peptides. Schally AV. Corticotropin-releasing factor (CRF)-like immunore-
1986;7:73–77. activity in the gastro-entero-pancreatic endocrine system. Peptides.
75. Williams CL, Peterson JM, Villar RG, Burks TF. Corticotropin- 1984;5(suppl 1):71–78.
releasing factor directly mediates colonic responses to stress. Am J 91. Suda T, Tomori N, Tozawa F, Mouri T, Demura H, Shizume K.
Physiol. 1987;253:G582–G586. Distribution and characterization of immunoreactive corticotropin-
76. Lenz HJ, Raedler A, Greten H, Vale WW, Rivier JE. Stress-induced releasing factor in human tissues. J Clin Endocrinol Metab. 1984;
gastrointestinal secretory and motor responses in rats are mediated by 59:861–866.
Chapter  |  75  Enteric Neurobiology of Stress 2017

  92. Kawai K, Hotate K, Chiba Y, Munekata E, Ohashi S, Wakabayashi 109. Miampamba M, Maillot C, Million M, Tache Y. Peripheral CRF
I, et  al. The distribution of corticotrophin-releasing factor immu- activates myenteric neurons in the proximal colon through CRF(1)
noreactivity in various ovine tissues. Acta Endocrinol (Copenh). receptor in conscious rats. Am J Physiol Gastrointest Liver Physiol.
1985;108:433–439. 2002;282:G857–G865.
  93. Kawahito Y, Sano H, Kawata M, Yuri K, Mukai S, Yamamura Y, et al. 110. Miampamba M, Million M, Yuan PQ, Larauche M, Tache Y. Water
Local secretion of corticotropin-releasing hormone by enterochroma- avoidance stress activates colonic myenteric neurons in female rats.
ffin cells in human colon. Gastroenterology. 1994;106:859–865. Neuroreport. 2007;18:679–682.
  94. Karalis K, Muglia LJ, Bae D, Hilderbrand H, Majzoub JA. CRH 111a. Chatzaki E, Crowe PD, Wang L, Million M, Tache Y, Grigoriadis
and the immune system. J Neuroimmunol. 1997;72:131–136. DE. CRF receptor type 1 and 2 expression and anatomical distribu-
  95. Hanani M, Wood JD. Corticotropin-releasing hormone excites mye- tion in the rat colon. J Neurochem. 2004;90:309–316.
nteric neurons in the guinea-pig small intestine. Eur J Pharmacol. 111b. Gourcerol G, Wu SV, Yuan PQ, Pham H, Miampamba M, Larauche
1992;211:23–27. M, et  al. Activation of corticotropin-releasing factor receptor 2
  96. Maillot C, Million M, Wei JY, Gauthier A, Tache Y. Peripheral mediates the colonic motor coping response to acute stress in
corticotropin-releasing factor and stress-stimulated colonic motor rodents. Gastroenterology. 2011;140:1586–1596.
activity involve type 1 receptor in rats. Gastroenterology. 2000;119: 112. Liu S, Gao N, Hu HZ, Wang X, Wang GD, Fang X, et  al.
1569–1579. Distribution and chemical coding of corticotropin-releasing factor-
  97. Martinez V, Wang L, Rivier JE, Vale W, Tache Y. Differential immunoreactive neurons in the guinea pig enteric nervous system.
actions of peripheral corticotropin-releasing factor (CRF), urocortin J Comp Neurol. 2006;494:63–74.
II, and urocortin III on gastric emptying and colonic transit in mice: 113. Furness JB. The Enteric Nervous System. Oxford: Blackwell; 2006.
role of CRF receptor subtypes 1 and 2. J Pharmacol Exp Ther. 114. Furness JB. Types of neurons in the enteric nervous system. J Auton
2002;301:611–617. Nerv Syst. 2000;81:87–96.
  98. Liu S, Gao X, Gao N, Wang X, Fang X, Hu HZ, et al. Expression 115. Brookes SJ. Classes of enteric nerve cells in the guinea-pig small
of type 1 corticotropin-releasing factor receptor in the guinea pig intestine. Anat Rec. 2001;262:58–70.
enteric nervous system. J Comp Neurol. 2005;481:284–298. 116. Castagliuolo I, Lamont JT, Qiu B, Fleming SM, Bhaskar KR,
  99. Barclay GR, Turnberg LA. Effect of cold-induced pain on salt and Nikulasson ST, et  al. Acute stress causes mucin release from rat
water transport in the human jejunum. Gastroenterology. 1988; colon: role of corticotropin releasing factor and mast cells. Am J
94:994–998. Physiol. 1996;271:G884–G892.
100. Barclay GR, Turnberg LA. Effect of psychological stress on salt and 117. Brookes SJ, Song ZM, Steele PA, Costa M. Identification of
water transport in the human jejunum. Gastroenterology. 1987;93: motor neurons to the longitudinal muscle of the guinea pig ileum.
91–97. Gastroenterology. 1992;103:961–973.
101. Saunders PR, Kosecka U, McKay DM, Perdue MH. Acute stressors 118. Costa M, Brookes SJ, Steele PA, Gibbins I, Burcher E, Kandiah CJ.
stimulate ion secretion and increase epithelial permeability in rat Neurochemical classification of myenteric neurons in the guinea-
intestine. Am J Physiol. 1994;267:G794–G799. pig ileum. Neuroscience. 1996;75:949–967.
102. Saunders PR, Santos J, Hanssen NP, Yates D, Groot JA, Perdue MH. 119. Vanner S, Surprenant A. Cholinergic and noncholinergic submu-
Physical and psychological stress in rats enhances colonic epithelial cosal neurons dilate arterioles in guinea pig colon. Am J Physiol.
permeability via peripheral CRH. Dig Dis Sci. 2002;47:208–215. 1991;261:G136–G144.
103. Wallon C, Yang PC, Keita AV, Ericson AC, McKay DM, Sherman 120. Poole DP, Castelucci P, Robbins HL, Chiocchetti R, Furness JB.
PM, et  al. Corticotropin-releasing hormone (CRH) regulates mac- The distribution of P2X3 purine receptor subunits in the guinea pig
romolecular permeability via mast cells in normal human colonic enteric nervous system. Auton Neurosci. 2002;101:39–47.
biopsies in vitro. Gut. 2008;57:50–58. 121. Furness JB, Alex G, Clark MJ, Lal VV. Morphologies and projec-
104. Vidal A, Gomez-Gil E, Sans M, Portella MJ, Salamero M, Pique tions of defined classes of neurons in the submucosa of the guinea-
JM, et al. Life events and inflammatory bowel disease relapse: a pro- pig small intestine. Anat Rec A Discov Mol Cell Evol Biol. 2003;272:
spective study of patients enrolled in remission. Am J Gastroenterol. 475–483.
2006;101:775–781. 122. Perrin MH, Vale WW. Corticotropin releasing factor receptors and
105. Langhorst J, Cobelens PM, Kavelaars A, Heijnen CJ, Benson S, their ligand family. Ann N Y Acad Sci. 1999;885:312–328.
Rifaie N, et  al. Stress-related peripheral neuroendocrine-immune 123. Chen R, Lewis KA, Perrin MH, Vale WW. Expression cloning of a
interactions in women with ulcerative colitis. Psychoneuroendo- human corticotropin-releasing-factor receptor. Proc Natl Acad Sci U
crinology. 2007;32:1086–1096. S A. 1993;90:8967–8971.
106. Lazar Z, Benko R, Bolcskei K, Rumbus Z, Wolf M, Holzer P, et al. 124. Perrin MH, Donaldson CJ, Chen R, Lewis KA, Vale WW. Cloning
Actions of endothelin and corticotropin releasing factor in the and functional expression of a rat brain corticotropin releasing fac-
guinea-pig ileum: no evidence for an interaction with capsaicin-sen- tor (CRF) receptor. Endocrinology. 1993;133:3058–3061.
sitive neurons. Neuropeptides. 2003;37:220–232. 125. Palchaudhuri MR, Wille S, Mevenkamp G, Spiess J, Fuchs E,
107. Mancinelli R, Azzena GB, Diana M, Forgione A, Fratta W. In vitro Dautzenberg FM. Corticotropin-releasing factor receptor type 1
excitatory actions of corticotropin-releasing factor on rat colonic from Tupaia belangeri--cloning, functional expression and tissue
motility. J Auton Pharmacol. 1998;18:319–324. distribution. Eur J Biochem. 1998;258:78–84.
108. Porcher C, Juhem A, Peinnequin A, Sinniger V, Bonaz B. 126. Perrin M, Donaldson C, Chen R, Blount A, Berggren T, Bilezikjian
Expression and effects of metabotropic CRF1 and CRF2 recep- L, et  al. Identification of a second corticotropin-releasing factor
tors in rat small intestine. Am J Physiol Gastrointest Liver Physiol. receptor gene and characterization of a cDNA expressed in heart.
2005;288:G1091–G1103. Proc Natl Acad Sci USA. 1995;92:2969–2973.
2018 SECTION  |  VI  Consequences of Disregulated Physiology

127. Lovenberg TW, Chalmers DT, Liu C, De Souza EB. CRF2 alpha 140. Thomas EA, Bornstein JC. Inhibitory cotransmission or after-
and CRF2 beta receptor mRNAs are differentially distributed hyperpolarizing potentials can regulate firing in recurrent net-
between the rat central nervous system and peripheral tissues. works with excitatory metabotropic transmission. Neuroscience.
Endocrinology. 1995;136:4139–4142. 2003;120:333–351.
128. Muramatsu Y, Sugino N, Suzuki T, Totsune K, Takahashi K, 141. Chambers JD, Bornstein JC, Sjovall H, Thomas EA. Recurrent
Tashiro A, et al. Urocortin and corticotropin-releasing factor recep- networks of submucous neurons controlling intestinal secretion:
tor expression in normal cycling human ovaries. J Clin Endocrinol a modeling study. Am J Physiol Gastrointest Liver Physiol. 2005;
Metab. 2001;86:1362–1369. 288:G887–G896.
129. Chatzaki E, Murphy BJ, Wang L, Million M, Ohning GV, Crowe 142. Cooke HJ. Neural and humoral regulation of small intestinal elec-
PD, et al. Differential profile of CRF receptor distribution in the rat trolyte transport. In: Johnson LR, Christensen J, Jackson MJ,
stomach and duodenum assessed by newly developed CRF receptor Jacobson ED, Walsh JH, eds. Physiology of the Gastrointestinal
antibodies. J Neurochem. 2004;88:1–11. Tract. New York: Raven Press; 1987. p. 1307–1350.
130. Hauger RL, Grigoriadis DE, Dallman MF, Plotsky PM, Vale WW, 143. Kiliaan AJ, Saunders PR, Bijlsma PB, Berin MC, Taminiau JA,
Dautzenberg FM. International Union of Pharmacology. XXXVI. Groot JA, et  al. Stress stimulates transepithelial macromolecular
Current status of the nomenclature for receptors for corticotropin- uptake in rat jejunum. Am J Physiol. 1998;275:G1037–G1044.
releasing factor and their ligands. Pharmacol Rev. 2003;55:21–26. 144. Bertrand PP, Kunze WA, Furness JB, Bornstein JC. The terminals
131. Hsu SY, Hsueh AJ. Human stresscopin and stresscopin-related pep- of myenteric intrinsic primary afferent neurons of the guinea-pig
tide are selective ligands for the type 2 corticotropin-releasing hor- ileum are excited by 5-hydroxytryptamine acting at 5-hydroxytryp-
mone receptor. Nat Med. 2001;7:605–611. tamine-3 receptors. Neuroscience. 2000;101:459–469.
132. Dautzenberg FM, Hauger RL. The CRF peptide family and their 145. Crofford LJ, Sano H, Karalis K, Friedman TC, Epps HR, Remmers
receptors: yet more partners discovered. Trends Pharmacol Sci. EF, et  al. Corticotropin-releasing hormone in synovial fluids and
2002;23:71–77. tissues of patients with rheumatoid arthritis and osteoarthritis. J
133. Furness JB, Trussell DC, Pompolo S, Bornstein JC, Smith TK. Immunol. 1993;151:1587–1596.
Calbindin neurons of the guinea-pig small intestine: quantita- 146. Crofford LJ, Sano H, Karalis K, Webster EA, Friedman TC,
tive analysis of their numbers and projections. Cell Tissue Res. Chrousos GP, et  al. Local expression of corticotropin-releasing
1990;260:261–272. hormone in inflammatory arthritis. Ann N Y Acad Sci. 1995;771:
134. Liu S, Hu H-Z, Gao N, Wang G-D, Wang X-Y, Fang X, et al. CRF1 459–471.
receptors mediate excitatory actions of urocortin on neurons in 147. Crofford LJ, Sano H, Karalis K, Webster EL, Goldmuntz EA,
guinea-pig small intestinal myenteric plexus. Gastroenterology. Chrousos GP, et  al. Local secretion of corticotropin-releasing hor-
2004;126(4 suppl 2):A411. mone in the joints of Lewis rats with inflammatory arthritis. J Clin
135. Lomax AE, Bertrand PP, Furness JB. Electrophysiological charac- Invest. 1992;90:2555–2564.
teristics distinguish three classes of neuron in submucosal ganglia 148. Karalis K, Sano H, Redwine J, Listwak S, Wilder RL, Chrousos
of the guinea-pig distal colon. Neuroscience. 2001;103:245–255. GP. Autocrine or paracrine inflammatory actions of corticotropin-
136. Reed DE, Vanner SJ. Converging and diverging cholinergic inputs releasing hormone in vivo. Science. 1991;254:421–423.
from submucosal neurons amplify activity of secretomotor neurons 149. Kawahito Y, Sano H, Mukai S, Asai K, Kimura S, Yamamura Y,
in guinea-pig ileal submucosa. Neuroscience. 2001;107:685–696. et al. Corticotropin releasing hormone in colonic mucosa in patients
137. Pan H, Gershon MD. Activation of intrinsic afferent pathways in with ulcerative colitis. Gut. 1995;37:544–551.
submucosal ganglia of the guinea pig small intestine. J Neurosci. 150. Wang XY, Wang GD, Xia Y, Ren W, Needleman BJ, Mikami DJ,
2000;20:3295–3309. et al. Mast Cell Activation by Corticotropin Releasing Factor
138. Kunze WA, Furness JB, Bornstein JC. Simultaneous intracellular (CRF) in Guinea Pig and Human Intestine. Digestive Disease Week
recordings from enteric neurons reveal that myenteric AH neurons Abstract T2407; mddwadmin@gastro.org, 2010.
transmit via slow excitatory postsynaptic potentials. Neuroscience. 151. Grafe P, Mayer CJ, Wood JD. Synaptic modulation of calcium-
1993;55:685–694. dependent potassium conductance in myenteric neurones in the
139. Thomas EA, Bertrand PP, Bornstein JC. A computer simulation guinea-pig. J Physiol. 1980;305:235–248.
of recurrent, excitatory networks of sensory neurons of the gut in
guinea-pig. Neurosci Lett. 2000;287:137–140.

S-ar putea să vă placă și