Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Oct 1.
Published in final edited form as: Exp Biol Med (Maywood). 2010 Oct;235(10):1168–1178. doi: 10.1258/ebm.2010.009347

Corticotropin-releasing factor signaling and visceral response to stress

Andreas Stengel 1, Yvette Taché 1
PMCID: PMC3169435  NIHMSID: NIHMS321402  PMID: 20881321

Abstract

Stress may cause behavioral and/or psychiatric manifestations such as anxiety and depression and also impact on the function of different visceral organs, namely the gastrointestinal and cardiovascular systems. During the past years substantial progress has been made in the understanding of the underlying mechanisms recruited by stressors. Activation of the corticotropin-releasing factor (CRF) signaling system is recognized to be involved in a large number of stress-related behavioral and somatic disorders. This review will outline the present knowledge on the distribution of the CRF system (ligands and receptors) expressed in the brain and peripheral viscera and its relevance in stress-induced alterations of gastrointestinal and cardiovascular functions and the therapeutic potential of CRF1 receptor antagonists.

Keywords: cardiovascular system, corticotropin-releasing factor (CRF), irritable bowel syndrome (IBS), stress, urocortin (Ucn)

Introduction

In 1936, Hans Selye pioneered the concept of biological stress based on macroscopic evidence that specific organs are commonly altered by various stressors. He showed that rats develop similar hypertrophy of the adrenals, involution of lymphatic organs and gastric erosions in response to exposure to noxious agents irrespectively of their chemical or physical nature.1,2 Thereafter, in the 1950s, Geoffrey Harris3,4 introduced the seminal concept of hypothalamic neurohumoral control of the pituitary adrenocorticotropin (ACTH) secretion that is activated by stress. Fifteen years later, the Nobel laureates, Drs Guillemin5 and Schally6 independently proved the existence of a corticotropin-releasing factor (CRF) in line with its stimulatory effect on ACTH release from the pituitary in rats. However, although CRF was one of the first hypothalamic releasing factors to be named, the elucidation of its structure lingered for nearly three decades until Vale et al.7 characterized CRF as a 41 amino acid (aa) peptide isolated from 490,000 ovine hypothalami. Subsequently, the same group cloned the CRF1 and CRF2 receptors, identified additional members of the CRF family812 and developed specific peptide CRF receptor antagonists1315 providing relevant tools to identify the molecular mechanisms of the stress response.

With great foresight, Selye2 had suspected early on that the yet to be chemically characterized CRF was ‘the first mediator that integrates the adaptive bodily response to stress’. Soon after the availability of the synthetic peptide, compelling reports established that CRF exerts a number of biological actions independently of the HPA activation. Existing experimental evidence showed that CRF injected into the brain mimics the overall behavioral (anxiety/depression and alteration of food consumption), autonomic (sympathetic and sacral parasympathetic activation), immune, metabolic and visceral adaptive changes produced by exposure to various systemic or cognitive stressors.1621 These initial findings built the foundation to explore the mechanisms of CRF’s actions in the brain and the role of CRF receptor activation in driving the stress-related visceral response. Consequently, curtailing the CRF signaling system was viewed as a potential pharmacological target in the drug treatment of various stress-related disorders.2224 Among those, irritable bowel syndrome (IBS) is a highly prevalent functional bowel disorder that affects up to 20% of the North American population.25 It is characterized by chronic abdominal pain or discomfort associated with changes in bowel habits (diarrhea, constipation or alternating pattern), generally in the absence of detectable organic alterations.2527 Stressful life events and psychosocial trauma, alone or combined with previous episodes of bowel infection/inflammation, have been identified as important risk factors that may account for the development, severity and/or maintenance of IBS symptoms.28 Moreover, IBS patients often show somatic29 as well as psychological comorbidities such as anxiety and depression reaching 40–90% in tertiary care centers.30 Therefore, the understanding of stress signaling pathways seems to be relevant in unraveling the contributing mechanisms of IBS and to identify possible new drug targets.

In this review, we will outline the state-of-knowledge on the CRF signaling system in the brain and visceral organs, and experimental evidence for a role of CRF pathways in mediating stress-related changes in behavioral and visceral functions with an emphasis on gastrointestinal and cardiovascular systems. Potential intervention strategies to dampen CRF signaling in stress-related disorders will also be highlighted.

Biochemical coding of stress signaling: CRF ligands and receptors

CRF peptide family

In addition to the 41 aa peptide CRF, three additional members of the CRF signaling family have been identified more recently, namely urocortin 1 (Ucn 1), urocortin 2 (Ucn 2) and urocortin 3 (Ucn 3).811 CRF has a well conserved primary structure among mammalian species including humans, primates, dogs and rodents.31 The mammalian Ucn 1 (also known as urocortin) is a 40 aa peptide isolated from the rat midbrain that displays 45% sequence identity with rat/human (r/h) CRF. As observed for CRF, the primary structure of Ucn 1 is highly conserved across mammalian species, including rat, mouse and sheep.8,32,33 Mouse Ucn 2 (mUcn 2) is a 38 aa peptide that shares 34% homology with r/h CRF and 42% with r/m Ucn 1,10 whereas human Ucn 3 has only little overlap with the structure of r/h CRF and r/h Ucn 1 (18% and 21% homology, respectively).9,11

CRF receptors

So far, two receptors, CRF1 and/or CRF2, have been cloned from two distinct genes that share 70% identity at the aa level.21,34 Both CRF receptor subtypes belong to the B1 subfamily of seven-transmembrane domain receptors that signal largely, but not exclusively, by coupling to G proteins resulting in the stimulation of adenylate cyclase.34 CRF1 and CRF2 receptors are subject to extensive alternative splicing resulting in various isoforms.3436 However, only the CRF1α isoform is coupled directly to adenylate cyclase, whereas the majority of other variants are lacking the ligand binding and/or signaling domains.34,35,37 Nonetheless, alternative splicing variants of CRF1 receptors are emerging to play an important role as modulators of downstream signaling, as recently thoroughly reviewed.38

With regards to CRF2 receptors, three functional splice variants differing in their N-terminal extracellular domains are known in humans (2α, 2β and 2γ) and two in rodents (2α and 2β).39 The C-terminus is common to CRF2α, CRF2β and CRF2γ receptor splice variants, while the N-terminal extracellular region, which interacts with the ligand, varies in length and is composed of 34, 61 and 20 aa, respectively.36,40,41 The sequence of the CRF2α is highly conserved across mammalian species and also found in amphibians, indicating an early development of this isoform which points towards its physiological importance.41 In contrast, the CRF2β is not as well conserved and only detected in mammals, indicating a more recent evolution.42 Four additional non-signaling CRF2α splice variants have been identified in the mouse brain and rat esophagus and some of them can act as a soluble binding protein (sCRF2α)36,43 or alter the cellular levels of full-length CRF2α mRNA and hence functional receptor levels.44 Recently a novel CRF2β isoform in the mouse heart has been found to act as a dominant-negative regulator of CRF2β membrane expression.45

The CRF1 and CRF2 receptors display different pharmacological binding characteristics. The CRF1α receptor has high affinity to CRF and Ucn 1 and no affinity to Ucn 2 and Ucn 3. In contrast, the CRF2α/β receptor displays high affinity to Ucn 1, Ucn 2 and Ucn 3 and lower affinity to CRF.8,10,39,41,46,47 While Ucn 2 and Ucn 3 have been identified as selective endogenous CRF2 agonists, so far there is no endogenous ligand exclusively binding to the CRF1 receptor. However, selective CRF1 peptide agonists such as cortagine and stressin1-A have been recently developed providing new tools to characterize the effects of selective activation of CRF1 receptors.48,49

Distribution of CRF peptides and receptors

CRF expressing neurons are widely distributed in the rat brain with major sites of CRF mRNA and CRF immunoreactivity in the paraventricular nucleus (PVN) of the hypothalamus, cerebral cortex, amygdalar-hippocampal complex and pontine Barrington’s nucleus.50,51 Of interest, the parvicellular part of the PVN contains a discrete group of CRF expressing neurons in the dorsal and ventral parts sending direct projections to spinal cord and brain stem nuclei, respectively, which regulate autonomic outflow to the viscera.52,53 Although being members of the same family, little neuroanatomical overlap exists between CRF and Ucns in the rat brain.8,10,5356 The central distribution of Ucn 1 is very limited with the highest expression in the Edinger–Westphal nucleus (EWN).55,56 Ucn 2 gene expression is localized in the PVN, supraoptic nucleus (SON), arcuate nucleus of the hypothalamus (Arc) and locus coeruleus (LC) as well as in several cranial motor nuclei (trigeminal, facial and hypoglossal) and the ventral horn of the spinal cord.10 Due to the lack of specific Ucn 2 antibodies, little is known about the brain distribution of Ucn 2 peptide. Ucn 3 mRNA is detected in the PVN mainly in the dorsal and ventral aspects, amygdala (basome-dial nucleus) and the basomedial nucleus of the stria terminalis.54

With regards to CRF receptors, dense CRF1 receptor expression is found in the forebrain, subcortical limbic structures such as the septal region and amygdala, whereas the expression in the hypothalamus is low under basal conditions but markedly up-regulated by stress.5759 Moreover, CRF1 receptors are prominently expressed in the anterior and intermediate lobe of the pituitary.60 CRF2 expression in the forebrain is restricted to subfornical structures with high expression in the lateral septum, amygdala and hypothalamus, including the ventromedial hypothalamus and SON.61 In the hindbrain, CRF2 receptors are expressed in the dorsal raphe nucleus, area postrema, nucleus of the solitary tract and choroid plexus.61

As for a variety of other neuropeptides, initially thought to be restricted to the brain and pituitary and later shown to be widely expressed in the periphery, CRF ligands and receptors are also detected in peripheral tissues including the gastrointestinal tract,62,63 heart, lung, spleen, testis and adipose tissue64 in animals and humans. In the gastrointestinal tract, immunostaining studies detected CRF immunoreactive neurons in the rat65 and guinea pig66 enteric nervous system. Ucn 1 is expressed in the rat colonic enteric nervous system at the gene67 and peptide65 level. In rodents, CRF receptors have been identified throughout the gastrointestinal tract. Whereas CRF1 receptor expression is localized in the myenteric and submucosal nervous plexus of the distal gut,68,69 CRF2 receptor expression was mainly identified on the luminal surface of the crypts, on blood vessels of the submucosal layer68 and also on myenteric neurons.70 Importantly, CRF2α is the main variant expressed in the rat brain, whereas CRF2β is expressed in non-neuronal tissues centrally as well as peripherally.71 In contrast, in humans CRF2β and CRF2γ are expressed mainly in brain neurons whereas CRF2α is found peripherally and centrally.72,73 In the human heart, Ucn 1 peptide has been shown by immunostaining74 along with the expression of CRF2α and CRF1 gene and protein expression.75 Whereas Ucn 1 concentrations were highest in the left ventricle as assessed by radioimmunoassay, CRF was very low or undetectable in the human heart.75

CRF signaling and stress-related behavioral responses

The effects of CRF ligands on behavior have been extensively characterized. Injection of CRF and Ucn 1 directly into the brain induces a variety of behavioral changes. For instance, CRF injected at low doses into the cerebrospinal fluid of non-stressed animals activates locomotor activity as well as rearing and grooming behavior which are unrelated to the activation of the pituitary adrenal axis.17,76,77 Central administration of CRF also induces arousal as reflected in typical activation patterns assessed by electroencephalography.78 Under basal conditions, central injection of CRF at low doses also stimulates memory and learning, whereas higher doses have an opposite effect.79 This enhancement of learning induced by central administration of CRF is CRF1 receptor-mediated.80 Likewise, Ucn 1 increases memory retention at low doses but decreases learning at high doses.81 In several tests including open field or the elevated plus maze, CRF or Ucn 1 induce CRF1-mediated, anxiogenic-like behavioral responses.76,8284 Moreover, exogenous central injection of either CRF or Ucn 1 decreases food intake, which is mediated by both CRF1 and CRF2 receptors in the early and late phases of the anorexigenic response, respectively.85,86 CRF has also been implicated in drug dependence as reflected in CRF dysregulation in abstinent alcohol dependent rats. Consistent reports showed that CRF1 receptor antagonists decrease the amount of self-administered drugs such as ethanol or heroin during a period of stress-related escalation.87

CRF signaling and stress-related changes in the gastrointestinal tract

Brain CRF receptors are involved in stress-related inhibition of gastric motor function

Several stressors are known to delay gastric emptying in animals as well as in healthy humans.20 Likewise, it is well established that CRF, Ucn 1 and Ucn 2 injected into the cerebrospinal fluid inhibit gastric emptying of a non-caloric or caloric liquid or solid meal and suppress propagative contractions through activation of CRF2 receptors.63 Moreover, pretreatment with peptide CRF receptor antagonists administered into the brain ventricle blocked the inhibition of gastric motor function induced by various stressors.20,88 However, under conditions of surgical stress (abdominal surgery with cecal palpation), central CRF1 receptors play a major role as indicated by the absence of the postoperative delay of gastric emptying 2 h after surgery in CRF1 knockout mice.89 The central action of CRF to delay gastric transit is not mediated by the associated stimulation of the HPA axis but by the autonomic nervous system since the gastric inhibitory motor response can still be observed in hypophysectomized or adrenal-ectomized rats and no longer in vagotomized rats.90,91 Key structures that influence autonomic outflow to the stomach, namely the PVN and the dorsal vagal complex in the brainstem, have been identified as the brain nuclei mediating the CRF-induced inhibition of gastric emptying and motility in rats.63,92,93

CRF injection into the brain and stress inhibit small intestinal motor function

Much less is known about the effects of stress on small intestinal motor function compared with the large amount of data on the other segments of the gut.88 Nonetheless, as observed in the stomach, acute stress as well as central injection of CRF or Ucn 1 inhibits duodenal and small intestinal transit and motility.94,95 However, the slowing of CRF-induced small intestinal transit is less pronounced compared with the stomach, which could be attributed to the more prominent enteric than autonomic control of the small intestinal motor function compared with the stomach.96 So far, the central CRF receptor subtype involved in the mediation of the inhibitory response in the small intestine remains to be described.

Brain CRF receptors are involved in stress-related stimulation of colonic motor function

Whereas several stressors delay gastric emptying and small intestinal transit, colonic motility is increased in stressed experimental animals and healthy subjects.20 The stimulatory response on colonic transit and defecation is mimicked by central injection of CRF or Ucn 1 primarily through activation of brain CRF1 receptors in rodents.63,97,98 In contrast to gastric motor functions, the CRF2 antagonist, astressin2-B, did not prevent the colonic response to central injection of CRF in rodents.20,63 As observed in the stomach, the CRF- and stress-induced stimulation of colonic motor function occurs independently from the activation of the HPA axis.91 The peripheral pathways involve increased parasympathetic outflow to the colon via vagal celiac branches innervating the proximal colon and more prominently the sacral parasympathetic fibers innervating the distal colon and rectum.99

Neuroanatomical and functional evidence support that activation of the CRF1 signaling pathway in the PVN and LC integrates the behavioral and autonomic–colonic responses to stress.100,101 CRF neurons are localized in the dorsal cap of the parvicellular PVN known to have transsynaptic connections to the colon.102 Moreover, CRF-containing neurons in the Barrington’s nucleus project to the LC and to the intermediolateral column of the sacral spinal cord, thereby providing input to catecholamine neurons projecting to the forebrain and sacral parasympathetic nervous system innervating the descending colon.96,102 Other studies established that water avoidance stress induces CRF gene transcription in the PVN and activates neurons in the PVN and LC/Barrington’s nuclei as assessed by the neuronal marker Fos and the response is blunted by the CRF receptor antagonist, α-helical CRF(9–41) injected into the lateral brain ventricle.103,104 Moreover, microinjection of CRF into the PVN or LC complex results in a stimulation of colonic motor function and α-helical CRF(9–41) microinjected directly into the PVN prevents the stimulated colonic transit and defecation induced by water avoidance or restraint stress,93 indicating the importance of the PVN in orchestrating these responses. Furthermore, CRF injected intracerebroventricularly (icv) increases the activity of noradrenergic neurons in the LC, resulting in the release of noradrenalin into the brain cortex mediating arousal and anxiogenic behavior.101,105

CRF system in the gut and stress-related alteration of gut motor function

The expression of both CRF receptors and ligands in the gastrointestinal tract points towards a role of a local CRF signaling system in the gut.68,70,106108 Peripheral injection of CRF or Ucn 1 in rats inhibits gastric emptying, delays small intestinal transit and stimulates colonic transit resulting in increased defecation.109111 The mediation of the delay in gastric emptying involves CRF2 receptors, whereas the stimulation of colonic motility is CRF1 receptor-mediated.62 Therefore, peripheral injection of Ucn 1 or CRF, which interact with both CRF1 and CRF2 receptors, simultaneously inhibits gastric motor function while increasing colonic motility in rodents.95,111,112 Consistent with a differential inhibitory and stimulatory effect of CRF2 and CRF1 on gastric and colonic motor function, respectively, peripherally injected Ucn 2 selectively delays gastric emptying without altering colonic motility,111,112 whereas the selective CRF1 agonists, stressin1-A and cortagine, stimulate defecation and have no effect on gastric emptying.49,113 Moreover, upon peripheral injection, the selective CRF2 antagonists, astressin2-B and antisauvagine-30, selectively block CRF- and Ucn 1-induced delay of gastric emptying,111,112 whereas the selective CRF1 antagonists, CP-154,526 and NBI 27914, selectively abolish CRF- or Ucn 1-induced stimulation of colonic motor function in rodents.110114 Likewise, the wrap restraint-induced delay of gastric emptying is blocked by peripheral injection of peptide non-selective CRF1/CRF2 antagonists115117 and selective CRF2 antagonist.112 Moreover, the restraint- or water avoidance stress-induced stimulation of colonic transit and fecal pellet output can be abolished by peripherally administered antagonists, α-helical CRF(9–41) or astressin, which are peptides not crossing the blood–brain barrier.109,110,118,119 These observations highlight the involvement of the peripheral CRF signaling system in mediating the gut motor response to stress. Collectively these data support the concept that activation of the CRF signaling system in the gut serves as the local efferent effector of the brain–gut interaction during stress.120 There is also evidence that the gut CRF system locally modulates gut inflammatory processes as recently reviewed.121

Implication of CRF1 receptors in the pathophysiology of IBS symptoms

Exogenous administration of CRF or Ucn 1 can reproduce cardinal features of diarrhea predominant IBS including anxiogenic behavior, enhanced visceral pain to colorectal distention, increased colonic mucus secretion, propulsive motility, development of watery stool/diarrhea and increased colonic mucosal permeability facilitating bacterial translocation into colonic tissue (Figure 1).62,98,113,120,122 Colonic mast cells activated by peripheral CRF in response to stress are involved in the enhanced permeability, bacterial uptake and pain response as shown by the mast cell stabilizer, doxantrazole, that prevents the central CRF-and stress-induced colonic hypersensitivity to colorectal distention in experimental animals and humans (Figure 1).119,123,124 Acute or chronic stress-induced visceral hyperalgesia is also mediated by the activation of CRF1 receptors.24,98 This is supported by pharmacological studies showing that central injection of CRF mimics stress-induced colonic hyperalgesia after colorectal distention22 and that CRF1 antagonists block colonic hypersensitivity to colorectal distention induced by various stressors in different rodent models of visceral hypersensitivity.22,24,98 In contrast, activation of the CRF2 receptor induced by peripheral injection of Ucn 2 or the CRF2 preferring agonist, sauvagine, reduces visceral pain following colorectal distention in rats.125,126 These data indicate that CRF1 and CRF2 receptors exert counteracting effects as shown by activation of CRF1 receptors facilitating visceral sensitization and activation of CRF2 receptors preventing visceral hyperalgesia.

Figure 1.

Figure 1

Activation of CRF1 signaling pathways and IBS-like manifestations, which can be blocked by CRF1 receptor antagonists. ANS, autonomic nervous system; ENS, enteric nervous system; IBS, irritable bowel syndrome (A color version of this figure is available in the online journal)

In addition to convergent preclinical evidence, data obtained in human studies indicate that systemic injection of CRF induces similar changes in the colon as observed in experimental animals. The peripheral administration of CRF decreases the threshold for visceral pain to colorectal distention in healthy human subjects.127,128 Other studies also showed that CRF activates subepithelial mast cells and stimulates transcellular uptake of protein antigens in colonic mucosal biopsies of healthy volunteers.124 In addition, systemic injection of CRF induces a colonic motility response more prominently in subjects with IBS compared with healthy controls along with the induction of abdominal pain and discomfort in IBS patients but not in healthy controls (Table 1).129 Conversely, peripheral injection of α-helical CRF(9–41) prevents rectal electrical stimulation-induced enhanced colonic motility, visceral perception and anxiety in IBS patients compared with healthy subjects without altering the HPA axis (Table 1).130 Moreover, peptide CRF antagonists also result in near normalization of the altered electroencephalogram activities in IBS patients under basal conditions and in response to colorectal distention.131 Taken together, these clinical reports support that the activation of the CRF pathway with preferential CRF1 agonists recaptures cardinal features of IBS symptoms which can be dampened by systemic injection of peptide CRF receptor antagonists. Preclinical studies clearly demonstrated that the blockade of CRF1 receptors can prevent/attenuate the development of those stress-related colonic functional or cellular alterations. Therefore, the conceptual framework supports that sustained activation of the CRF1 system at central and/or peripheral sites may be one component underlying stress-related manifestations of IBS symptoms. However, two recent double-blind randomized clinical trials failed to show efficacy of CRF1 antagonist treatment on colonic motility in patients with diarrhea-predominant IBS (Table 1)132 or on visceral pain in male and female IBS patients.133 As several other CRF1 candidates are being tested, results from these clinical trials should provide additional insight regarding the potential therapeutic benefit of CRF1 receptor antagonists for anxiety, depression and IBS as established in a large array of preclinical in vivo studies.134

Table 1.

Overlap of symptoms/parameters induced by CRF and dampened by CRF1 antagonists

Symptoms/parameters Effects of systemic CRF in healthy subjects Effects of systemic CRF1 antagonists in IBS patients
Anxiety 168 130,169
Colonic motility 122 ↓, =122,132
Colonic permeability 124 124
Gut inflammation 170 140
Visceral sensitivity to colorectal distention 128 ↓, =122,133

increase; =, no change; ↓, decrease

CRF, corticotropin-releasing factor; IBS, irritable bowel syndrome

Implication of CRF1 receptors in the pathophysiology of inflammatory bowel disease

Although stress does not cause inflammatory bowel disease (IBD) encompassing ulcerative colitis as well as Crohn’s disease, it does aggravate the condition. Particularly, psychological stress increases the risk to exacerbate existing IBD symptoms and/or to induce the flare-up of symptoms.135 Consequently, owing to its characterized role in stress-response, the CRF system has been investigated in relation with the pathophysiology of IBD in experimental models and clinical settings.136

The various modalities through which the local CRF system is regulated in the gut during inflammation and conversely influences inflammatory processes have been recently reviewed in detail.121 CRF increases the release of proinflammatory cytokines such as tumor necrosis factor (TNF)-α and interleukin (IL)-6 from macrophages in vitro.137 In vivo studies showed that the CRF1 receptor antagonist, antalarmin, inhibits the endotoxin-induced inflammatory response as reflected by the suppression of elevated cytokines, namely TNF-α, IL-1β, and IL-6, resulting in prolonged survival after endotoxin treatment in BALB/c mice.137 In the ileum, the inflammatory response to intraluminal perfusion of Clostridium difficile toxin is markedly reduced in mice with deletion of the CRF gene138 or after treatment with double-stranded RNA locally silencing the CRF expression in the ileum.139 Clinical observations showed that the expression of CRF is increased in enterochromaffin cells and macrophages in mucosal biopsies of patients with IBD and may exert a proinflammatory action.140 Although the existing experimental and clinical data are encouraging, clinical trials are still to be performed to assess whether the modulation of CRF signaling impacts on IBD symptoms.

CRF signaling and the cardiovascular system

CRF administered into the lateral brain ventricle in rats elicits widespread effects on the cardiovascular system, namely increasing heart rate, mean arterial pressure and cardiac output that are similar to those induced by various stressors (Table 2).141 In addition, the icv administration of the CRF1/CRF2 antagonist, α-helical CRF(9–41) attenuates icv IL-1β-induced tachycardia and hypertension.142 In contrast, intravenous injection of CRF, and more so Ucns, causes vasodilation and concomitantly decreases blood pressure accompanied by compensatory tachycardia.143,144 Under in vitro conditions using an isolated preparation of rat heart, CRF increases coronary blood flow, releases atrial natriuretic peptide and exerts a positive inotropic effect (Table 2).145,146 These findings suggest that CRF influences cardiovascular functions not only centrally but also via a local action. In line with this assumption, CRF and more abundantly Ucns along with CRF2 receptors are expressed in the heart of rodents and humans.74,75,147,148

Table 2.

Effects of activation of CRF2 receptors on cardiovascular functions in animal models

Site of injection Species Effect Reference
icv Rat ↑ Heart Rate 141
Rat ↑ Mean arterial pressure 141
Rat ↑ Cardiac output 141
iv Dog Vasodilation 171
Dog ↓ Blood pressure 171
Dog ↑ Heart Rate 171
In vitro Rat ↑ Coronary blood flow 146
Rat Vasodilation 146
Rat ↑ Aortic pressure 146
Rat ↑ Oxygen consumption 146
Rat Positive inotropic effect 146
Rat ↑ Atrial natriuretic peptide 145

increase; ↓, decrease; icv, intracerebroventricular; iv, intravenous

Convincing evidence established that the CRF2 receptor mediates the peripheral effects of the CRF signaling system on the heart and blood vessels.149151 CRF receptors, particularly CRF2β, are densely expressed in the heart and in blood vessels.152154 Ucn 1 and CRF2 receptor expression are modulated by stress as shown by systemic endotoxin administration and restraint that increase Ucn 1 mRNA expression but decrease CRF2 mRNA in the rat atria and ventricles.155,156 The predominant role of peripheral CRF2 receptors in cardiovascular functions is further underlined by the observation that CRF2 KO mice display an elevated basal systolic and mean arterial pressure.157 Moreover, CRF2 KO mice do not respond to peripheral injection of Ucn 1, whereas their wild-type littermates show a pronounced decrease in blood pressure following peripheral injection of the peptide.157 Due to the high binding affinity of Ucns for CRF2 compared with CRF, the vasodilatory and inotropic effects of Ucns are more pronounced than those induced by CRF.158,159 These observations led to the assumption of a cardioprotective role of peripheral Ucns and their potential therapeutic use under conditions of heart failure. In preclinical studies, systemic injection of Ucn 1, Ucn 2 or Ucn 3 decreases peripheral resistance and atrial pressure and concomitantly increases cardiac output in animals with heart failure.160162 Moreover, exogenous Ucn 1 reduced the number of cell deaths following hypoxia163 resulting in decreased infarction size.164,165 Likewise, intravenous infusion of Ucn 2 decreased systemic vascular resistance while increasing cardiac output, heart rate and left ventricular ejection fraction in healthy human subjects.166 Other clinical studies indicate that patients with systolic heart failure have elevated plasma Ucn 1 levels in the early stage of the disease (New York Heart Association, NYHA 1–2), whereas with progression (NYHA 3–4), the levels decrease167 suggesting a possible predictive value of circulating Ucn 1 as a biomarker. Collectively, existing evidence supports the potential clinical relevance of using Ucns as a new therapeutic intervention for heart failure or following hypoxic stress such as myocardial infarction. However, clinical trials are needed to corroborate the preclinical and proof of concept clinical findings.

Summary

The CRF signaling system composed of CRF, Ucn 1, Ucn 2 and Ucn 3 along with CRF1 and CRF2 receptors is largely expressed in the brain and visceral organs such as the gut and cardiovascular system, all known to be affected by stress. Pharmacological approaches using selective CRF antagonists in experimental animals have demonstrated that the activation of CRF1 receptors plays a major role in the development of anxiogenic and colonic propulsive responses to stress independently from the HPA axis. In the brain and gut, the activation of CRF1 receptors recapitulates key features of IBS-diarrhea predominant patients such as watery stool, visceral hyperalgesia, an increase in colonic motility, mucus secretion and also induces hypervigilance. Moreover, these alterations can be abolished by CRF1 antagonists in preclinical IBS-like models suggesting therapeutic potential for CRF1 antagonists. However, two recent clinical trials failed to demonstrate efficacy of CRF1 antagonists in the treatment of IBS symptoms. Additional clinical studies are still ongoing with new CRF1 antagonist candidates to establish their potential therapeutic value in the drug treatment of IBS. Another feature of the CRF system is its proinflammatory action in the gut that may have a bearing with the underlying mechanism of stress-related exacerbation of IBDs. Lastly, central activation of CRF receptors results in a hypertensive response that may contribute to the development of stress-related hypertension. By contrast, in the periphery the activation of Ucns-CRF2 signaling exerts potent vasodilatory effects. Preclinical and clinical studies point towards the use of CRF family members, especially Ucn 1, as possible biomarkers in the diagnosis and progress monitoring of systolic heart failure as well as in the drug treatment of heart failure or following hypoxic stress such as myocardial infarction.

Acknowledgments

YT is in receipt of a VA Research Career Scientist Award and NIH R01 grants DK 33061 and DK 57238. AS is supported by the German Research Foundation Grant STE 1765/1-1. We thank Eugenia Hu for careful reading of the manuscript.

Footnotes

Author contributions: AS provided the first draft of the manuscript, and YT structured and critically reviewed and edited the manuscript.

References

  • 1.Selye H. A syndrome produced by diverse nocuous agents. Nature. 1936;138:32. doi: 10.1176/jnp.10.2.230a. [DOI] [PubMed] [Google Scholar]
  • 2.Selye H. Theories. Boston-London: Butterworths; 1976. [Google Scholar]
  • 3.De Groot J, Harris GW. Hypothalmic control of the anterior pituitary gland and blood lymphocytes. J Physiol. 1950;111:335–46. doi: 10.1113/jphysiol.1950.sp004483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Harris GW. The hypothalamus and endocrine glands. Br Med Bull. 1950;6:345–50. doi: 10.1093/oxfordjournals.bmb.a073628. [DOI] [PubMed] [Google Scholar]
  • 5.Guillemin R, Rosenberg B. Humoral hypothalamic control of anterior pituitary: a study with combined tissue cultures. Endocrinology. 1955;57:599–607. doi: 10.1210/endo-57-5-599. [DOI] [PubMed] [Google Scholar]
  • 6.Saffran M, Schally AV, Benfey BG. Stimulation of the release of corticotropin from the adenohypophysis by a neurohypophysial factor. Endocrinology. 1955;57:439–44. doi: 10.1210/endo-57-4-439. [DOI] [PubMed] [Google Scholar]
  • 7.Vale W, Spiess J, Rivier C, Rivier J. Characterization of a 41-residue ovine hypothalamic peptide that stimulates secretion of corticotropin and beta-endorphin. Science. 1981;213:1394–7. doi: 10.1126/science.6267699. [DOI] [PubMed] [Google Scholar]
  • 8.Vaughan J, Donaldson C, Bittencourt J, Perrin MH, Lewis K, Sutton S, Chan R, Turnbull AV, Lovejoy D, Rivier C, Rivier J, Sawchenko PE, Vale W. Urocortin, a mammalian neuropeptide related to fish urotensin I and to corticotropin-releasing factor. Nature. 1995;378:287–92. doi: 10.1038/378287a0. [DOI] [PubMed] [Google Scholar]
  • 9.Hsu SY, Hsueh AJ. Human stresscopin and stresscopin-related peptide are selective ligands for the type 2 corticotropin-releasing hormone receptor. Nat Med. 2001;7:605–11. doi: 10.1038/87936. [DOI] [PubMed] [Google Scholar]
  • 10.Reyes TM, Lewis K, Perrin MH, Kunitake KS, Vaughan J, Arias CA, Hogenesch JB, Gulyas J, Rivier J, Vale WW, Sawchenko PE. Urocortin II: a member of the corticotropin-releasing factor (CRF) neuropeptide family that is selectively bound by type 2 CRF receptors. Proc Natl Acad Sci USA. 2001;98:2843–8. doi: 10.1073/pnas.051626398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lewis K, Li C, Perrin MH, Blount A, Kunitake K, Donaldson C, Vaughan J, Reyes TM, Gulyas J, Fischer W, Bilezikjian L, Rivier J, Sawchenko PE, Vale WW. Identification of urocortin III, an additional member of the corticotropin-releasing factor (CRF) family with high affinity for the CRF2 receptor. Proc Natl Acad Sci USA. 2001;98:7570–5. doi: 10.1073/pnas.121165198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Perrin MH, Vale WW. Corticotropin releasing factor receptors and their ligand family. Ann NY Acad Sci. 1999;885:312–28. doi: 10.1111/j.1749-6632.1999.tb08687.x. [DOI] [PubMed] [Google Scholar]
  • 13.Gulyas J, Rivier C, Perrin M, Koerber SC, Sutton S, Corrigan A, Lahrichi SL, Craig AG, Vale W, Rivier J. Potent, structurally constrained agonists and competitive antagonists of corticotropin-releasing factor. Proc Natl Acad Sci USA. 1995;92:10575–9. doi: 10.1073/pnas.92.23.10575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rivier J, Gulyas J, Corrigan A, Martinez V, Craig AG, Taché Y, Vale W, Rivier C. Astressin analogues (corticotropin-releasing factor antagonists) with extended duration of action in the rat. J Med Chem. 1998;41:5012–19. doi: 10.1021/jm980426c. [DOI] [PubMed] [Google Scholar]
  • 15.Rivier J, Gulyas J, Kirby D, Low W, Perrin MH, Kunitake K, DiGruccio M, Vaughan J, Reubi JC, Waser B, Koerber SC, Martinez V, Wang L, Taché Y, Vale W. Potent and long-acting corticotropin releasing factor (CRF) receptor 2 selective peptide competitive antagonists. J Med Chem. 2002;45:4737–47. doi: 10.1021/jm0202122. [DOI] [PubMed] [Google Scholar]
  • 16.Taché Y, Goto Y, Gunion M, Rivier J, Debas H. Inhibition of gastric acid secretion in rats and in dogs by corticotropin-releasing factor. Gastroenterology. 1984;86:281–6. [PubMed] [Google Scholar]
  • 17.Dunn AJ, Berridge CW. Physiological and behavioral responses to corticotropin-releasing factor administration: is CRF a mediator of anxiety or stress responses? Brain Res Brain Res Rev. 1990;15:71–100. doi: 10.1016/0165-0173(90)90012-d. [DOI] [PubMed] [Google Scholar]
  • 18.De Souza EB. Corticotropin-releasing factor receptors: physiology, pharmacology, biochemistry and role in central nervous system and immune disorders. Psychoneuroendocrinology. 1995;20:789–819. doi: 10.1016/0306-4530(95)00011-9. [DOI] [PubMed] [Google Scholar]
  • 19.Habib KE, Weld KP, Rice KC, Pushkas J, Champoux M, Listwak S, Webster EL, Atkinson AJ, Schulkin J, Contoreggi C, Chrousos GP, McCann SM, Suomi SJ, Higley JD, Gold PW. Oral administration of a corticotropin-releasing hormone receptor antagonist significantly attenuates behavioral, neuroendocrine, and autonomic responses to stress in primates. Proc Natl Acad Sci USA. 2000;97:6079–84. doi: 10.1073/pnas.97.11.6079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Taché Y, Martinez V, Million M, Wang L. Stress and the gastrointestinal tract III. Stress-related alterations of gut motor function: role of brain corticotropin-releasing factor receptors. Am J Physiol Gastrointest Liver Physiol. 2001;280:G173–7. doi: 10.1152/ajpgi.2001.280.2.G173. [DOI] [PubMed] [Google Scholar]
  • 21.Bale TL, Vale WW. CRF and CRF receptors: role in stress responsivity and other behaviors. Annu Rev Pharmacol Toxicol. 2004;44:525–57. doi: 10.1146/annurev.pharmtox.44.101802.121410. [DOI] [PubMed] [Google Scholar]
  • 22.Taché Y, Brunnhuber S. From Hans Selye’s discovery of biological stress to the identification of corticotropin-releasing factor signaling pathways: implication in stress-related functional bowel diseases. Ann NY Acad Sci. 2008;1148:29–41. doi: 10.1196/annals.1410.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chrousos GP. Stress and disorders of the stress system. Nat Rev Endocrinol. 2009;5:374–81. doi: 10.1038/nrendo.2009.106. [DOI] [PubMed] [Google Scholar]
  • 24.Taché Y, Kiank C, Stengel A. A role for corticotropin-releasing factor in functional gastrointestinal disorders. Curr Gastroenterol Rep. 2009;11:270–7. doi: 10.1007/s11894-009-0040-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Talley NJ. Functional gastrointestinal disorders as a public health problem. Neurogastroenterol Motil. 2008;20(Suppl 1):121–9. doi: 10.1111/j.1365-2982.2008.01097.x. [DOI] [PubMed] [Google Scholar]
  • 26.Camilleri M. Management of the irritable bowel syndrome. Gastroenterology. 2001;120:652–68. doi: 10.1053/gast.2001.21908. [DOI] [PubMed] [Google Scholar]
  • 27.Halpert A, Drossman D. Biopsychosocial issues in irritable bowel syndrome. J Clin Gastroenterol. 2005;39:665–9. doi: 10.1097/01.mcg.0000174024.81096.44. [DOI] [PubMed] [Google Scholar]
  • 28.Santos J, Alonso C, Vicario M, Ramos L, Lobo B, Malagelada JR. Neuropharmacology of stress-induced mucosal inflammation: implications for inflammatory bowel disease and irritable bowel syndrome. Curr Mol Med. 2008;8:258–73. doi: 10.2174/156652408784533788. [DOI] [PubMed] [Google Scholar]
  • 29.Riedl A, Schmidtmann M, Stengel A, Goebel M, Wisser AS, Klapp BF, Mönnikes H. Somatic comorbidities of irritable bowel syndrome: a systematic analysis. J Psychosom Res. 2008;64:573–82. doi: 10.1016/j.jpsychores.2008.02.021. [DOI] [PubMed] [Google Scholar]
  • 30.North CS, Hong BA, Alpers DH. Relationship of functional gastrointestinal disorders and psychiatric disorders: implications for treatment. World J Gastroenterol. 2007;13:2020–7. doi: 10.3748/wjg.v13.i14.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lovejoy DA, Balment RJ. Evolution and physiology of the corticotropin-releasing factor (CRF) family of neuropeptides in vertebrates. Gen Comp Endocrinol. 1999;115:1–22. doi: 10.1006/gcen.1999.7298. [DOI] [PubMed] [Google Scholar]
  • 32.Zhao L, Donaldson CJ, Smith GW, Vale WW. The structures of the mouse and human urocortin genes (Ucn and UCN) Genomics. 1998;50:23–33. doi: 10.1006/geno.1998.5292. [DOI] [PubMed] [Google Scholar]
  • 33.Cepoi D, Sutton S, Arias C, Sawchenko P, Vale WW. Ovine genomic urocortin: cloning, pharmacologic characterization, and distribution of central mRNA. Brain Res Mol Brain Res. 1999;68:109–18. doi: 10.1016/s0169-328x(99)00076-5. [DOI] [PubMed] [Google Scholar]
  • 34.Grammatopoulos DK, Chrousos GP. Functional characteristics of CRH receptors and potential clinical applications of CRH-receptor antagonists. Trends Endocrinol Metab. 2002;13:436–44. doi: 10.1016/s1043-2760(02)00670-7. [DOI] [PubMed] [Google Scholar]
  • 35.Pisarchik A, Slominski AT. Alternative splicing of CRH-R1 receptors in human and mouse skin: identification of new variants and their differential expression. FASEB J. 2001;15:2754–6. doi: 10.1096/fj.01-0487fje. [DOI] [PubMed] [Google Scholar]
  • 36.Wu SV, Yuan PQ, Wang L, Peng YL, Chen CY, Taché Y. Identification and characterization of multiple corticotropin-releasing factor type 2 receptor isoforms in the rat esophagus. Endocrinology. 2007;148:1675–87. doi: 10.1210/en.2006-0565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Pisarchik A, Slominski A. Molecular and functional characterization of novel CRFR1 isoforms from the skin. Eur J Biochem. 2004;271:2821–30. doi: 10.1111/j.1432-1033.2004.04216.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zmijewski MA, Slominski AT. Emerging role of alternative splicing of CRF1 receptor in CRF signaling. Acta Biochim Pol. 2010;57:1–13. [PMC free article] [PubMed] [Google Scholar]
  • 39.Hauger RL, Grigoriadis DE, Dallman MF, Plotsky PM, Vale WW, Dautzenberg FM. International Union of Pharmacology. XXXVI. Current status of the nomenclature for receptors for corticotropin-releasing factor and their ligands. Pharmacol Rev. 2003;55:21–6. doi: 10.1124/pr.55.1.3. [DOI] [PubMed] [Google Scholar]
  • 40.Miyata I, Shiota C, Chaki S, Okuyama S, Inagami T. Localization and characterization of a short isoform of the corticotropin-releasing factor receptor type 2alpha (CRF(2)alpha-tr) in the rat brain. Biochem Biophys Res Commun. 2001;280:553–7. doi: 10.1006/bbrc.2000.4112. [DOI] [PubMed] [Google Scholar]
  • 41.Dautzenberg FM, Higelin J, Wille S, Brauns O. Molecular cloning and functional expression of the mouse CRF2(a) receptor splice variant. Regul Pept. 2004;121:89–97. doi: 10.1016/j.regpep.2004.04.009. [DOI] [PubMed] [Google Scholar]
  • 42.Dautzenberg FM, Kilpatrick GJ, Hauger RL, Moreau J. Molecular biology of the CRH receptors – in the mood. Peptides. 2001;22:753–60. doi: 10.1016/s0196-9781(01)00388-6. [DOI] [PubMed] [Google Scholar]
  • 43.Chen AM, Perrin MH, Digruccio MR, Vaughan JM, Brar BK, Arias CM, Lewis KA, Rivier JE, Sawchenko PE, Vale WW. A soluble mouse brain splice variant of type 2alpha corticotropin-releasing factor (CRF) receptor binds ligands and modulates their activity. Proc Natl Acad Sci USA. 2005;102:2620–5. doi: 10.1073/pnas.0409583102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Evans RT, Seasholtz AF. Soluble corticotropin-releasing hormone receptor 2alpha splice variant is efficiently translated but not trafficked for secretion. Endocrinology. 2009;150:4191–202. doi: 10.1210/en.2009-0285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sztainberg Y, Kuperman Y, Issler O, Gil S, Vaughan J, Rivier J, Vale W, Chen A. A novel corticotropin-releasing factor receptor splice variant exhibits dominant negative activity: a putative link to stress-induced heart disease. FASEB J. 2009;23:2186–96. doi: 10.1096/fj.08-128066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Dautzenberg FM, Hauger RL. The CRF peptide family and their receptors: yet more partners discovered. Trends Pharmacol Sci. 2002;23:71–7. doi: 10.1016/s0165-6147(02)01946-6. [DOI] [PubMed] [Google Scholar]
  • 47.Smart D, Coppell A, Rossant C, Hall M, McKnight AT. Characterisation using microphysiometry of CRF receptor pharmacology. Eur J Pharmacol. 1999;379:229–35. doi: 10.1016/s0014-2999(99)00506-3. [DOI] [PubMed] [Google Scholar]
  • 48.Tezval H, Jahn O, Todorovic C, Sasse A, Eckart K, Spiess J. Cortagine, a specific agonist of corticotropin-releasing factor receptor subtype 1, is anxiogenic and antidepressive in the mouse model. Proc Natl Acad Sci USA. 2004;101:9468–73. doi: 10.1073/pnas.0403159101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Rivier J, Gulyas J, Kunitake K, DiGruccio M, Cantle JP, Perrin MH, Donaldson C, Vaughan J, Million M, Gourcerol G, Adelson DW, Rivier C, Taché Y, Vale W. Stressin1-A, a potent corticotropin releasing factor receptor 1 (CRF1)-selective peptide agonist. J Med Chem. 2007;50:1668–74. doi: 10.1021/jm0613875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Swanson LW, Sawchenko PE, Lind RW. Regulation of multiple peptides in CRF parvocellular neurosecretory neurons: implications for the stress response. Prog Brain Res. 1986;68:169–90. doi: 10.1016/s0079-6123(08)60238-1. [DOI] [PubMed] [Google Scholar]
  • 51.Valentino RJ, Page ME, Luppi PH, Zhu Y, Van Bockstaele E, Aston-Jones G. Evidence for widespread afferents to Barrington’s nucleus, a brainstem region rich in corticotropin-releasing hormone neurons. Neuroscience. 1994;62:125–43. doi: 10.1016/0306-4522(94)90320-4. [DOI] [PubMed] [Google Scholar]
  • 52.Sawchenko PE, Swanson LW. Immunohistochemical identification of neurons in the paraventricular nucleus of the hypothalamus that project to the medulla or to the spinal cord in the rat. J Comp Neurol. 1982;205:260–72. doi: 10.1002/cne.902050306. [DOI] [PubMed] [Google Scholar]
  • 53.Sawchenko PE, Imaki T, Potter E, Kovacs K, Imaki J, Vale W. The functional neuroanatomy of corticotropin-releasing factor. Ciba Found Symp. 1993;172:5–21. doi: 10.1002/9780470514368.ch2. [DOI] [PubMed] [Google Scholar]
  • 54.Venihaki M, Sakihara S, Subramanian S, Dikkes P, Weninger SC, Liapakis G, Graf T, Majzoub JA. Urocortin III, a brain neuropeptide of the corticotropin-releasing hormone family: modulation by stress and attenuation of some anxiety-like behaviours. J Neuroendocrinol. 2004;16:411–22. doi: 10.1111/j.1365-2826.2004.01170.x. [DOI] [PubMed] [Google Scholar]
  • 55.Kozicz T, Yanaihara H, Arimura A. Distribution of urocortin-like immunoreactivity in the central nervous system of the rat. J Comp Neurol. 1998;391:1–10. doi: 10.1002/(sici)1096-9861(19980202)391:1<1::aid-cne1>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
  • 56.Bittencourt JC, Vaughan J, Arias C, Rissman RA, Vale WW, Sawchenko PE. Urocortin expression in rat brain: evidence against a pervasive relationship of urocortin-containing projections with targets bearing type 2 CRF receptors. J Comp Neurol. 1999;415:285–312. [PubMed] [Google Scholar]
  • 57.Bonaz B, Rivest S. Effect of a chronic stress on CRF neuronal activity and expression of its type 1 receptor in the rat brain. Am J Physiol. 1998;275:R1438–49. doi: 10.1152/ajpregu.1998.275.5.R1438. [DOI] [PubMed] [Google Scholar]
  • 58.Imaki T, Katsumata H, Miyata M, Naruse M, Imaki J, Minami S. Expression of corticotropin-releasing hormone type 1 receptor in paraventricular nucleus after acute stress. Neuroendocrinology. 2001;73:293–301. doi: 10.1159/000054646. [DOI] [PubMed] [Google Scholar]
  • 59.Konishi S, Kasagi Y, Katsumata H, Minami S, Imaki T. Regulation of corticotropin-releasing factor (CRF) type-1 receptor gene expression by CRF in the hypothalamus. Endocr J. 2003;50:21–36. doi: 10.1507/endocrj.50.21. [DOI] [PubMed] [Google Scholar]
  • 60.Turnbull AV, Rivier C. Corticotropin-releasing factor (CRF) and endocrine responses to stress: CRF receptors, binding protein, and related peptides. Proc Soc Exp Biol Med. 1997;215:1–10. doi: 10.3181/00379727-215-44108. [DOI] [PubMed] [Google Scholar]
  • 61.Bittencourt JC, Sawchenko PE. Do centrally administered neuropeptides access cognate receptors? : an analysis in the central corticotropin-releasing factor system. J Neurosci. 2000;20:1142–56. doi: 10.1523/JNEUROSCI.20-03-01142.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Taché Y, Perdue MH. Role of peripheral CRF signalling pathways in stress-related alterations of gut motility and mucosal function. Neurogastroenterol Motil. 2004;16(Suppl 1):137–42. doi: 10.1111/j.1743-3150.2004.00490.x. [DOI] [PubMed] [Google Scholar]
  • 63.Taché Y, Bonaz B. Corticotropin-releasing factor receptors and stress-related alterations of gut motor function. J Clin Invest. 2007;117:33–40. doi: 10.1172/JCI30085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Boorse GC, Denver RJ. Widespread tissue distribution and diverse functions of corticotropin-releasing factor and related peptides. Gen Comp Endocrinol. 2006;146:9–18. doi: 10.1016/j.ygcen.2005.11.014. [DOI] [PubMed] [Google Scholar]
  • 65.Kimura T, Amano T, Uehara H, Ariga H, Ishida T, Torii A, Tajiri H, Matsueda K, Yamato S. Urocortin I is present in the enteric nervous system and exerts an excitatory effect via cholinergic and serotonergic pathways in the rat colon. Am J Physiol Gastrointest Liver Physiol. 2007;293:G903–10. doi: 10.1152/ajpgi.00066.2007. [DOI] [PubMed] [Google Scholar]
  • 66.Liu S, Gao N, Hu HZ, Wang X, Wang GD, Fang X, Gao X, Xia Y, Wood JD. Distribution and chemical coding of corticotropin-releasing factor-immunoreactive neurons in the guinea pig enteric nervous system. J Comp Neurol. 2006;494:63–74. doi: 10.1002/cne.20781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Harada S, Imaki T, Naruse M, Chikada N, Nakajima K, Demura H. Urocortin mRNA is expressed in the enteric nervous system of the rat. Neurosci Lett. 1999;267:125–8. doi: 10.1016/s0304-3940(99)00349-3. [DOI] [PubMed] [Google Scholar]
  • 68.Chatzaki E, Crowe PD, Wang L, Million M, Taché Y, Grigoriadis DE. CRF receptor type 1 and 2 expression and anatomical distribution in the rat colon. J Neurochem. 2004;90:309–16. doi: 10.1111/j.1471-4159.2004.02490.x. [DOI] [PubMed] [Google Scholar]
  • 69.Yuan PQ, Million M, Wu SV, Rivier J, Taché Y. Peripheral corticotropin releasing factor (CRF) and a novel CRF1 receptor agonist, stressin1-A activate CRF1 receptor expressing cholinergic and nitrergic myenteric neurons selectively in the colon of conscious rats. Neurogastroenterol Motil. 2007;19:923–36. doi: 10.1111/j.1365-2982.2007.00978.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Porcher C, Juhem A, Peinnequin A, Sinniger V, Bonaz B. Expression and effects of metabotropic CRF1 and CRF2 receptors in rat small intestine. Am J Physiol Gastrointest Liver Physiol. 2005;288:G1091–103. doi: 10.1152/ajpgi.00302.2004. [DOI] [PubMed] [Google Scholar]
  • 71.Chalmers DT, Lovenberg TW, De Souza EB. Localization of novel corticotropin-releasing factor receptor (CRF2) mRNA expression to specific subcortical nuclei in rat brain: comparison with CRF1 receptor mRNA expression. J Neurosci. 1995;15:6340–50. doi: 10.1523/JNEUROSCI.15-10-06340.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Valdenaire O, Giller T, Breu V, Gottowik J, Kilpatrick G. A new functional isoform of the human CRF2 receptor for corticotropin-releasing factor. Biochim Biophys Acta. 1997;1352:129–32. doi: 10.1016/s0167-4781(97)00047-x. [DOI] [PubMed] [Google Scholar]
  • 73.Kostich WA, Chen A, Sperle K, Largent BL. Molecular identification and analysis of a novel human corticotropin-releasing factor (CRF) receptor: the CRF2gamma receptor. Mol Endocrinol. 1998;12:1077–85. doi: 10.1210/mend.12.8.0145. [DOI] [PubMed] [Google Scholar]
  • 74.Nishikimi T, Miyata A, Horio T, Yoshihara F, Nagaya N, Takishita S, Yutani C, Matsuo H, Matsuoka H, Kangawa K. Urocortin, a member of the corticotropin-releasing factor family, in normal and diseased heart. Am J Physiol Heart Circ Physiol. 2000;279:H3031–9. doi: 10.1152/ajpheart.2000.279.6.H3031. [DOI] [PubMed] [Google Scholar]
  • 75.Kimura Y, Takahashi K, Totsune K, Muramatsu Y, Kaneko C, Darnel AD, Suzuki T, Ebina M, Nukiwa T, Sasano H. Expression of urocortin and corticotropin-releasing factor receptor subtypes in the human heart. J Clin Endocrinol Metab. 2002;87:340–6. doi: 10.1210/jcem.87.1.8160. [DOI] [PubMed] [Google Scholar]
  • 76.Sutton RE, Koob GF, Le Moal M, Rivier J, Vale W. Corticotropin releasing factor produces behavioural activation in rats. Nature. 1982;297:331–3. doi: 10.1038/297331a0. [DOI] [PubMed] [Google Scholar]
  • 77.Sherman JE, Kalin NH. The effects of ICV-CRH on novelty-induced behavior. Pharmacol Biochem Behav. 1987;26:699–703. doi: 10.1016/0091-3057(87)90599-5. [DOI] [PubMed] [Google Scholar]
  • 78.Ehlers CL, Henriksen SJ, Wang M, Rivier J, Vale W, Bloom FE. Corticotropin releasing factor produces increases in brain excitability and convulsive seizures in rats. Brain Res. 1983;278:332–6. doi: 10.1016/0006-8993(83)90266-4. [DOI] [PubMed] [Google Scholar]
  • 79.Koob GF, Bloom FE. Corticotropin-releasing factor and behavior. Fed Proc. 1985;44:259–63. [PubMed] [Google Scholar]
  • 80.Radulovic J, Ruhmann A, Liepold T, Spiess J. Modulation of learning and anxiety by corticotropin-releasing factor (CRF) and stress: differential roles of CRF receptors 1 and 2. J Neurosci. 1999;19:5016–25. doi: 10.1523/JNEUROSCI.19-12-05016.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zorrilla EP, Schulteis G, Ormsby A, Klaassen A, Ling N, McCarthy JR, Koob GF, De Souza EB. Urocortin shares the memory modulating effects of corticotropin-releasing factor (CRF): mediation by CRF1 receptors. Brain Res. 2002;952:200–10. doi: 10.1016/s0006-8993(02)03345-0. [DOI] [PubMed] [Google Scholar]
  • 82.Takahashi LK, Kalin NH, Vanden Burgt JA, Sherman JE. Corticotropin-releasing factor modulates defensive-withdrawal and exploratory behavior in rats. Behav Neurosci. 1989;103:648–54. doi: 10.1037//0735-7044.103.3.648. [DOI] [PubMed] [Google Scholar]
  • 83.Moreau JL, Kilpatrick G, Jenck F. Urocortin, a novel neuropeptide with anxiogenic-like properties. Neuroreport. 1997;8:1697–701. doi: 10.1097/00001756-199705060-00027. [DOI] [PubMed] [Google Scholar]
  • 84.Spina MG, Merlo-Pich E, Akwa Y, Balducci C, Basso AM, Zorrilla EP, Britton KT, Rivier J, Vale WW, Koob GF. Time-dependent induction of anxiogenic-like effects after central infusion of urocortin or corticotropin-releasing factor in the rat. Psychopharmacology (Berl) 2002;160:113–21. doi: 10.1007/s00213-001-0940-y. [DOI] [PubMed] [Google Scholar]
  • 85.Richard D, Huang Q, Timofeeva E. The corticotropin-releasing hormone system in the regulation of energy balance in obesity. Int J Obes Relat Metab Disord. 2000;24(Suppl 2):S36–9. doi: 10.1038/sj.ijo.0801275. [DOI] [PubMed] [Google Scholar]
  • 86.Cullen MJ, Ling N, Foster AC, Pelleymounter MA. Urocortin, corticotropin releasing factor-2 receptors and energy balance. Endocrinology. 2001;142:992–9. doi: 10.1210/endo.142.3.7989. [DOI] [PubMed] [Google Scholar]
  • 87.Koob GF, Zorrilla EP. Neurobiological mechanisms of addiction: focus on corticotropin-releasing factor. Curr Opin Invest Drugs. 2010;11:63–71. [PMC free article] [PubMed] [Google Scholar]
  • 88.Taché Y, Million M. Central corticotropin-releasing factor and the hypothalamic–pituitary–adrenal axis in gastrointestinal physiology. In: Johnson LR, Wood J, editors. Physiology of the Gastrointestinal Tract. 4. San Diego: Elsevier Academic Press; 2006. pp. 791–816. [Google Scholar]
  • 89.Luckey A, Wang L, Jamieson PM, Basa NR, Million M, Czimmer J, Vale W, Taché Y. Corticotropin-releasing factor receptor 1-deficient mice do not develop postoperative gastric ileus. Gastroenterology. 2003;125:654–9. doi: 10.1016/s0016-5085(03)01069-2. [DOI] [PubMed] [Google Scholar]
  • 90.Taché Y, Maeda-Hagiwara M, Turkelson CM. Central nervous system action of corticotropin-releasing factor to inhibit gastric emptying in rats. Am J Physiol. 1987;253:G241–5. doi: 10.1152/ajpgi.1987.253.2.G241. [DOI] [PubMed] [Google Scholar]
  • 91.Lenz HJ, Burlage M, Raedler A, Greten H. Central nervous system effects of corticotropin-releasing factor on gastrointestinal transit in the rat. Gastroenterology. 1988;94:598–602. doi: 10.1016/0016-5085(88)90229-6. [DOI] [PubMed] [Google Scholar]
  • 92.Heymann-Mönnikes I, Taché Y, Trauner M, Weiner H, Garrick T. CRF microinjected into the dorsal vagal complex inhibits TRH analog- and kainic acid-stimulated gastric contractility in rats. Brain Res. 1991;554:139–44. doi: 10.1016/0006-8993(91)90181-t. [DOI] [PubMed] [Google Scholar]
  • 93.Mönnikes H, Schmidt BG, Raybould HE, Taché Y. CRF in the paraventricular nucleus mediates gastric and colonic motor response to restraint stress. Am J Physiol. 1992;262:G137–43. doi: 10.1152/ajpgi.1992.262.1.G137. [DOI] [PubMed] [Google Scholar]
  • 94.Lenz HJ, Raedler A, Greten H, Vale WW, Rivier JE. Stress-induced gastrointestinal secretory and motor responses in rats are mediated by endogenous corticotropin-releasing factor. Gastroenterology. 1988;95:1510–7. doi: 10.1016/s0016-5085(88)80070-2. [DOI] [PubMed] [Google Scholar]
  • 95.Kihara N, Fujimura M, Yamamoto I, Itoh E, Inui A, Fujimiya M. Effects of central and peripheral urocortin on fed and fasted gastroduodenal motor activity in conscious rats. Am J Physiol Gastrointest Liver Physiol. 2001;280:G406–19. doi: 10.1152/ajpgi.2001.280.3.G406. [DOI] [PubMed] [Google Scholar]
  • 96.Taché Y. The parasympathetic nervous system in the pathophysiology of the gastrointestinal tract. In: Bolis CL, Licinio J, Govoni S, editors. Handbook of Autonomic Nervous System in Health and Disease. New York: Dekker Inc; 2002. pp. 463–503. [Google Scholar]
  • 97.Martinez V, Wang L, Rivier J, Grigoriadis D, Taché Y. Central CRF, urocortins and stress increase colonic transit via CRF1 receptors while activation of CRF2 receptors delays gastric transit in mice. J Physiol. 2004;556:221–34. doi: 10.1113/jphysiol.2003.059659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Martinez V, Taché Y. CRF1 receptors as a therapeutic target for irritable bowel syndrome. Curr Pharm Des. 2006;12:4071–88. doi: 10.2174/138161206778743637. [DOI] [PubMed] [Google Scholar]
  • 99.Million M, Wang L, Martinez V, Taché Y. Differential Fos expression in the paraventricular nucleus of the hypothalamus, sacral parasympathetic nucleus and colonic motor response to water avoidance stress in Fischer and Lewis rats. Brain Res. 2000;877:345–53. doi: 10.1016/s0006-8993(00)02719-0. [DOI] [PubMed] [Google Scholar]
  • 100.Mönnikes H, Schmidt BG, Tebbe J, Bauer C, Taché Y. Microinfusion of corticotropin releasing factor into the locus coeruleus/subcoeruleus nuclei stimulates colonic motor function in rats. Brain Res. 1994;644:101–8. doi: 10.1016/0006-8993(94)90352-2. [DOI] [PubMed] [Google Scholar]
  • 101.Valentino RJ, Miselis RR, Pavcovich LA. Pontine regulation of pelvic viscera: pharmacological target for pelvic visceral dysfunctions. Trends Pharmacol Sci. 1999;20:253–60. doi: 10.1016/s0165-6147(99)01332-2. [DOI] [PubMed] [Google Scholar]
  • 102.Valentino RJ, Kosboth M, Colflesh M, Miselis RR. Transneuronal labeling from the rat distal colon: anatomic evidence for regulation of distal colon function by a pontine corticotropin-releasing factor system. J Comp Neurol. 2000;417:399–414. [PubMed] [Google Scholar]
  • 103.Bonaz B, Taché Y. Water-avoidance stress-induced c-fos expression in the rat brain and stimulation of fecal output: role of corticotropin-releasing factor. Brain Res. 1994;641:21–8. doi: 10.1016/0006-8993(94)91810-4. [DOI] [PubMed] [Google Scholar]
  • 104.Kresse AE, Million M, Saperas E, Taché Y. Colitis induces CRF expression in hypothalamic magnocellular neurons and blunts CRF gene response to stress in rats. Am J Physiol Gastrointest Liver Physiol. 2001;281:G1203–13. doi: 10.1152/ajpgi.2001.281.5.G1203. [DOI] [PubMed] [Google Scholar]
  • 105.Koob GF. Corticotropin-releasing factor, norepinephrine, and stress. Biol Psychiatry. 1999;46:1167–80. doi: 10.1016/s0006-3223(99)00164-x. [DOI] [PubMed] [Google Scholar]
  • 106.Chatzaki E, Murphy BJ, Wang L, Million M, Ohning GV, Crowe PD, Petroski R, Taché Y, Grigoriadis DE. Differential profile of CRF receptor distribution in the rat stomach and duodenum assessed by newly developed CRF receptor antibodies. J Neurochem. 2004;88:1–11. doi: 10.1046/j.1471-4159.2003.02078.x. [DOI] [PubMed] [Google Scholar]
  • 107.Liu S, Gao X, Gao N, Wang X, Fang X, Hu HZ, Wang GD, Xia Y, Wood JD. Expression of type 1 corticotropin-releasing factor receptor in the guinea pig enteric nervous system. J Comp Neurol. 2005;481:284–98. doi: 10.1002/cne.20370. [DOI] [PubMed] [Google Scholar]
  • 108.Porcher C, Peinnequin A, Pellissier S, Meregnani J, Sinniger V, Canini F, Bonaz B. Endogenous expression and in vitro study of CRF-related peptides and CRF receptors in the rat gastric antrum. Peptides. 2006;27:1464–75. doi: 10.1016/j.peptides.2005.10.023. [DOI] [PubMed] [Google Scholar]
  • 109.Williams CL, Peterson JM, Villar RG, Burks TF. Corticotropin-releasing factor directly mediates colonic responses to stress. Am J Physiol. 1987;253:G582–6. doi: 10.1152/ajpgi.1987.253.4.G582. [DOI] [PubMed] [Google Scholar]
  • 110.Maillot C, Million M, Wei JY, Gauthier A, Taché Y. Peripheral corticotropin-releasing factor and stress-stimulated colonic motor activity involve type 1 receptor in rats. Gastroenterology. 2000;119:1569–79. doi: 10.1053/gast.2000.20251. [DOI] [PubMed] [Google Scholar]
  • 111.Martinez V, Wang L, Rivier JE, Vale W, Taché Y. Differential actions of peripheral corticotropin-releasing factor (CRF), urocortin II, and urocortin III on gastric emptying and colonic transit in mice: role of CRF receptor subtypes 1 and 2. J Pharmacol Exp Ther. 2002;301:611–17. doi: 10.1124/jpet.301.2.611. [DOI] [PubMed] [Google Scholar]
  • 112.Million M, Maillot C, Saunders P, Rivier J, Vale W, Taché Y. Human urocortin II, a new CRF-related peptide, displays selective CRF(2)-mediated action on gastric transit in rats. Am J Physiol Gastrointest Liver Physiol. 2002;282:G34–40. doi: 10.1152/ajpgi.00283.2001. [DOI] [PubMed] [Google Scholar]
  • 113.Larauche MH, Gourcerol G, Wang L, Pambukchian K, Brunnhuber S, Adelson DW, Rivier JE, Million M, Taché Y. Cortagine: a CRF1 agonist induces stress-like alterations of colonic function and visceral hypersensitivity in rodents primarily through peripheral pathways. Am J Physiol Gastrointest Liver Physiol. 2009;297:G215–27. doi: 10.1152/ajpgi.00072.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Saunders PR, Maillot C, Million M, Taché Y. Peripheral corticotropin-releasing factor induces diarrhea in rats: role of CRF1 receptor in fecal watery excretion. Eur J Pharmacol. 2002;435:231–5. doi: 10.1016/s0014-2999(01)01574-6. [DOI] [PubMed] [Google Scholar]
  • 115.Barquist E, Zinner M, Rivier J, Taché Y. Abdominal surgery-induced delayed gastric emptying in rats: role of CRF and sensory neurons. Am J Physiol. 1992;262:G616–20. doi: 10.1152/ajpgi.1992.262.4.G616. [DOI] [PubMed] [Google Scholar]
  • 116.Martinez V, Rivier J, Taché Y. Peripheral injection of a new corticotropin-releasing factor (CRF) antagonist, astressin, blocks peripheral CRF- and abdominal surgery-induced delayed gastric emptying in rats. J Pharmacol Exp Ther. 1999;290:629–34. [PubMed] [Google Scholar]
  • 117.Nozu T, Martinez V, Rivier J, Taché Y. Peripheral urocortin delays gastric emptying: role of CRF receptor 2. Am J Physiol. 1999;276:G867–74. doi: 10.1152/ajpgi.1999.276.4.G867. [DOI] [PubMed] [Google Scholar]
  • 118.Miyata K, Ito H, Fukudo S. Involvement of the 5-HT3 receptor in CRH-induce defecation in rats. Am J Physiol. 1998;274:G827–31. doi: 10.1152/ajpgi.1998.274.5.G827. [DOI] [PubMed] [Google Scholar]
  • 119.Castagliuolo I, Lamont JT, Qiu B, Fleming SM, Bhaskar KR, Nikulasson ST, Kornetsky C, Pothoulakis C. Acute stress causes mucin release from rat colon: role of corticotropin releasing factor and mast cells. Am J Physiol. 1996;271:G884–92. doi: 10.1152/ajpgi.1996.271.5.G884. [DOI] [PubMed] [Google Scholar]
  • 120.Stengel A, Taché Y. Neuroendocrine control of the gut during stress: corticotropin-releasing factor signaling pathways in the spotlight. Annu Rev Physiol. 2009;71:219–39. doi: 10.1146/annurev.physiol.010908.163221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kiank C, Taché Y, Larauche M. Stress-related modulation of inflammation in experimental models of bowel disease and post-infectious irritable bowel syndrome: role of corticotropin-releasing factor receptors. Brain Behav Immun. 2010;24:41–8. doi: 10.1016/j.bbi.2009.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Fukudo S. Role of corticotropin-releasing hormone in irritable bowel syndrome and intestinal inflammation. J Gastroenterol. 2007;42(Suppl 17):48–51. doi: 10.1007/s00535-006-1942-7. [DOI] [PubMed] [Google Scholar]
  • 123.Gue M, Del Rio-Lacheze C, Eutamene H, Theodorou V, Fioramonti J, Bueno L. Stress-induced visceral hypersensitivity to rectal distension in rats: role of CRF and mast cells. Neurogastroenterol Motil. 1997;9:271–9. doi: 10.1046/j.1365-2982.1997.d01-63.x. [DOI] [PubMed] [Google Scholar]
  • 124.Wallon C, Yang PC, Keita AV, Ericson AC, McKay DM, Sherman PM, Perdue MH, Soderholm JD. Corticotropin-releasing hormone (CRH) regulates macromolecular permeability via mast cells in normal human colonic biopsies in vitro. Gut. 2008;57:50–8. doi: 10.1136/gut.2006.117549. [DOI] [PubMed] [Google Scholar]
  • 125.Million M, Maillot C, Adelson DA, Nozu T, Gauthier A, Rivier J, Chrousos GP, Bayati A, Mattsson H, Taché Y. Peripheral injection of sauvagine prevents repeated colorectal distension-induced visceral pain in female rats. Peptides. 2005;26:1188–95. doi: 10.1016/j.peptides.2005.02.004. [DOI] [PubMed] [Google Scholar]
  • 126.Million M, Wang L, Wang Y, Adelson DW, Yuan PQ, Maillot C, Coutinho SV, McRoberts JA, Bayati A, Mattsson H, Wu V, Wei JY, Rivier J, Vale W, Mayer EA, Taché Y. CRF2 receptor activation prevents colorectal distension induced visceral pain and spinal ERK1/2 phosphorylation in rats. Gut. 2006;55:172–81. doi: 10.1136/gut.2004.051391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Lembo T, Plourde V, Shui Z, Fullerton S, Mertz H, Taché Y, Sytnik B, Munakata J, Mayer E. Effects of the corticotropin-releasing factor (CRF) on rectal afferent nerves in humans. Neurogastroenterol Motil. 1996;8:9–18. doi: 10.1111/j.1365-2982.1996.tb00237.x. [DOI] [PubMed] [Google Scholar]
  • 128.Nozu T, Kudaira M. Corticotropin-releasing factor induces rectal hypersensitivity after repetitive painful rectal distention in healthy humans. J Gastroenterol. 2006;41:740–4. doi: 10.1007/s00535-006-1848-4. [DOI] [PubMed] [Google Scholar]
  • 129.Fukudo S, Nomura T, Hongo M. Impact of corticotropin-releasing hormone on gastrointestinal motility and adrenocorticotropic hormone in normal controls and patients with irritable bowel syndrome. Gut. 1998;42:845–9. doi: 10.1136/gut.42.6.845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Sagami Y, Shimada Y, Tayama J, Nomura T, Satake M, Endo Y, Shoji T, Karahashi K, Hongo M, Fukudo S. Effect of a corticotropin releasing hormone receptor antagonist on colonic sensory and motor function in patients with irritable bowel syndrome. Gut. 2004;53:958–64. doi: 10.1136/gut.2003.018911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Tayama J, Sagami Y, Shimada Y, Hongo M, Fukudo S. Effect of alpha-helical CRH on quantitative electroencephalogram in patients with irritable bowel syndrome. Neurogastroenterol Motil. 2007;19:471–83. doi: 10.1111/j.1365-2982.2007.00903.x. [DOI] [PubMed] [Google Scholar]
  • 132.Sweetser S, Camilleri M, Linker Nord SJ, Burton DD, Castenada L, Croop R, Tong G, Dockens R, Zinsmeister AR. Do corticotropin releasing factor-1 receptors influence colonic transit and bowel function in females with irritable bowel syndrome? Am J Physiol Gastrointest Liver Physiol. 2009;296:G1299–306. doi: 10.1152/ajpgi.00011.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Dukes GE, Mayer EA, Kelleher DL, Hicks KJ, Boardley KL, Alpers DH. A randomised, double blind, placebo (PLA) controlled, crossover study to evaluate the efficacy and safety of the corticotrophin releasing factor 1 (CRF1) receptor antagonist (RA) GW876008 in irritable bowel syndrome (IBS) patients (Pts) Neurogastroenterol Motil. 2009;21:279. [Google Scholar]
  • 134.Zorrilla EP, Koob GF. Progress in corticotropin-releasing factor-1 antagonist development. Drug Discov Today. 2010;15:371–83. doi: 10.1016/j.drudis.2010.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Mawdsley JE, Rampton DS. Psychological stress in IBD: new insights into pathogenic and therapeutic implications. Gut. 2005;54:1481–91. doi: 10.1136/gut.2005.064261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Paschos KA, Kolios G, Chatzaki E. The corticotropin-releasing factor system in inflammatory bowel disease: prospects for new therapeutic approaches. Drug Discov Today. 2009;14:713–20. doi: 10.1016/j.drudis.2009.04.002. [DOI] [PubMed] [Google Scholar]
  • 137.Agelaki S, Tsatsanis C, Gravanis A, Margioris AN. Corticotropin-releasing hormone augments proinflammatory cytokine production from macrophages in vitro and in lipopolysaccharide-induced endotoxin shock in mice. Infect Immun. 2002;70:6068–74. doi: 10.1128/IAI.70.11.6068-6074.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Anton PM, Gay J, Mykoniatis A, Pan A, O’Brien M, Brown D, Karalis K, Pothoulakis C. Corticotropin-releasing hormone (CRH) requirement in Clostridium difficile toxin A-mediated intestinal inflammation. Proc Natl Acad Sci USA. 2004;101:8503–8. doi: 10.1073/pnas.0402693101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.la Fleur SE, Wick EC, Idumalla PS, Grady EF, Bhargava A. Role of peripheral corticotropin-releasing factor and urocortin II in intestinal inflammation and motility in terminal ileum. Proc Natl Acad Sci USA. 2005;102:7647–52. doi: 10.1073/pnas.0408531102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Gross KJ, Pothoulakis C. Role of neuropeptides in inflammatory bowel disease. Inflamm Bowel Dis. 2007;13:918–32. doi: 10.1002/ibd.20129. [DOI] [PubMed] [Google Scholar]
  • 141.Fisher LA, Jessen G, Brown MR. Corticotropin-releasing factor (CRF): mechanism to elevate mean arterial pressure and heart rate. Regul Pept. 1983;5:153–61. doi: 10.1016/0167-0115(83)90123-4. [DOI] [PubMed] [Google Scholar]
  • 142.Nakamori T, Morimoto A, Murakami N. Effect of a central CRF antagonist on cardiovascular and thermoregulatory responses induced by stress or IL-1 beta. Am J Physiol. 1993;265:R834–9. doi: 10.1152/ajpregu.1993.265.4.R834. [DOI] [PubMed] [Google Scholar]
  • 143.MacCannell KL, Hamilton PL, Lederis K, Newton CA, Rivier J. Corticotropin releasing factor-like peptides produce selective dilatation of the dog mesenteric circulation. Gastroenterology. 1984;87:94–102. [PubMed] [Google Scholar]
  • 144.Chen CY, Doong ML, Rivier JE, Taché Y. Intravenous urocortin II decreases blood pressure through CRF(2) receptor in rats. Regul Pept. 2003;113:125–30. doi: 10.1016/s0167-0115(03)00003-x. [DOI] [PubMed] [Google Scholar]
  • 145.Grunt M, Haug C, Duntas L, Pauschinger P, Maier V, Pfeiffer EF. Dilatory and inotropic effects of corticotropin-releasing factor (CRF) on the isolated heart. Effects on atrial natriuretic peptide (ANP) release. Horm Metab Res. 1992;24:56–9. doi: 10.1055/s-2007-1003255. [DOI] [PubMed] [Google Scholar]
  • 146.Grunt M, Glaser J, Schmidhuber H, Pauschinger P, Born J. Effects of corticotropin-releasing factor on isolated rat heart activity. Am J Physiol. 1993;264:H1124–9. doi: 10.1152/ajpheart.1993.264.4.H1124. [DOI] [PubMed] [Google Scholar]
  • 147.Muglia LJ, Jenkins NA, Gilbert DJ, Copeland NG, Majzoub JA. Expression of the mouse corticotropin-releasing hormone gene in vivo and targeted inactivation in embryonic stem cells. J Clin Invest. 1994;93:2066–72. doi: 10.1172/JCI117201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Okosi A, Brar BK, Chan M, D’Souza L, Smith E, Stephanou A, Latchman DS, Chowdrey HS, Knight RA. Expression and protective effects of urocortin in cardiac myocytes. Neuropeptides. 1998;32:167–71. doi: 10.1016/s0143-4179(98)90033-6. [DOI] [PubMed] [Google Scholar]
  • 149.Coste SC, Quintos RF, Stenzel-Poore MP. Corticotropin-releasing hormone-related peptides and receptors: emergent regulators of cardiovascular adaptations to stress. Trends Cardiovasc Med. 2002;12:176–82. doi: 10.1016/s1050-1738(02)00157-3. [DOI] [PubMed] [Google Scholar]
  • 150.Hashimoto K, Nishiyama M, Tanaka Y, Noguchi T, Asaba K, Hossein PN, Nishioka T, Makino S. Urocortins and corticotropin releasing factor type 2 receptors in the hypothalamus and the cardiovascular system. Peptides. 2004;25:1711–21. doi: 10.1016/j.peptides.2004.05.024. [DOI] [PubMed] [Google Scholar]
  • 151.Takahashi K, Totsune K, Murakami O, Shibahara S. Urocortins as cardiovascular peptides. Peptides. 2004;25:1723–31. doi: 10.1016/j.peptides.2004.04.018. [DOI] [PubMed] [Google Scholar]
  • 152.Perrin M, Donaldson C, Chen R, Blount A, Berggren T, Bilezikjian L, Sawchenko P, Vale W. Identification of a second corticotropin-releasing factor receptor gene and characterization of a cDNA expressed in heart. Proc Natl Acad Sci USA. 1995;92:2969–73. doi: 10.1073/pnas.92.7.2969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Kageyama K, Gaudriault GE, Bradbury MJ, Vale WW. Regulation of corticotropin-releasing factor receptor type 2 beta messenger ribonucleic acid in the rat cardiovascular system by urocortin, glucocorticoids, and cytokines. Endocrinology. 2000;141:2285–93. doi: 10.1210/endo.141.7.7572. [DOI] [PubMed] [Google Scholar]
  • 154.Baigent SM, Lowry PJ. mRNA expression profiles for corticotrophin-releasing factor (CRF), urocortin, CRF receptors and CRF-binding protein in peripheral rat tissues. J Mol Endocrinol. 2000;25:43–52. doi: 10.1677/jme.0.0250043. [DOI] [PubMed] [Google Scholar]
  • 155.Heldwein KA, Duncan JE, Stenzel P, Rittenberg MB, Stenzel-Poore MP. Endotoxin regulates corticotropin-releasing hormone receptor 2 in heart and skeletal muscle. Mol Cell Endocrinol. 1997;131:167–72. doi: 10.1016/s0303-7207(97)00103-2. [DOI] [PubMed] [Google Scholar]
  • 156.Pournajafi Nazarloo H, Tanaka Y, Dorobantu M, Hashimoto K. Modulation of corticotropin-releasing hormone receptor type 2 mRNA expression by CRH deficiency or stress in the mouse heart. Regul Pept. 2003;115:131–8. doi: 10.1016/s0167-0115(03)00150-2. [DOI] [PubMed] [Google Scholar]
  • 157.Coste SC, Kesterson RA, Heldwein KA, Stevens SL, Heard AD, Hollis JH, Murray SE, Hill JK, Pantely GA, Hohimer AR, Hatton DC, Phillips TJ, Finn DA, Low MJ, Rittenberg MB, Stenzel P, Stenzel-Poore MP. Abnormal adaptations to stress and impaired cardiovascular function in mice lacking corticotropin-releasing hormone receptor-2. Nat Genet. 2000;24:403–9. doi: 10.1038/74255. [DOI] [PubMed] [Google Scholar]
  • 158.Parkes DG, Vaughan J, Rivier J, Vale W, May CN. Cardiac inotropic actions of urocortin in conscious sheep. Am J Physiol. 1997;272:H2115–22. doi: 10.1152/ajpheart.1997.272.5.H2115. [DOI] [PubMed] [Google Scholar]
  • 159.Terui K, Higashiyama A, Horiba N, Furukawa KI, Motomura S, Suda T. Coronary vasodilation and positive inotropism by urocortin in the isolated rat heart. J Endocrinol. 2001;169:177–83. doi: 10.1677/joe.0.1690177. [DOI] [PubMed] [Google Scholar]
  • 160.Rademaker MT, Charles CJ, Espiner EA, Fisher S, Frampton CM, Kirkpatrick CM, Lainchbury JG, Nicholls MG, Richards AM, Vale WW. Beneficial hemodynamic, endocrine, and renal effects of urocortin in experimental heart failure: comparison with normal sheep. J Am Coll Cardiol. 2002;40:1495–505. doi: 10.1016/s0735-1097(02)02170-8. [DOI] [PubMed] [Google Scholar]
  • 161.Rademaker MT, Charles CJ, Espiner EA, Frampton CM, Lainchbury JG, Richards AM. Four-day urocortin-I administration has sustained beneficial haemodynamic, hormonal, and renal effects in experimental heart failure. Eur Heart J. 2005;26:2055–62. doi: 10.1093/eurheartj/ehi351. [DOI] [PubMed] [Google Scholar]
  • 162.Rademaker MT, Cameron VA, Charles CJ, Richards AM. Urocortin 3: haemodynamic, hormonal, and renal effects in experimental heart failure. Eur Heart J. 2006;27:2088–98. doi: 10.1093/eurheartj/ehl138. [DOI] [PubMed] [Google Scholar]
  • 163.Brar BK, Jonassen AK, Stephanou A, Santilli G, Railson J, Knight RA, Yellon DM, Latchman DS. Urocortin protects against ischemic and reperfusion injury via a MAPK-dependent pathway. J Biol Chem. 2000;275:8508–14. doi: 10.1074/jbc.275.12.8508. [DOI] [PubMed] [Google Scholar]
  • 164.Schulman D, Latchman DS, Yellon DM. Urocortin protects the heart from reperfusion injury via upregulation of p42/p44 MAPK signaling pathway. Am J Physiol Heart Circ Physiol. 2002;283:H1481–8. doi: 10.1152/ajpheart.01089.2001. [DOI] [PubMed] [Google Scholar]
  • 165.Liu CN, Yang C, Liu XY, Li S. In vivo protective effects of urocortin on ischemia-reperfusion injury in rat heart via free radical mechanisms. Can J Physiol Pharmacol. 2005;83:459–65. doi: 10.1139/y05-033. [DOI] [PubMed] [Google Scholar]
  • 166.Davis ME, Pemberton CJ, Yandle TG, Fisher SF, Lainchbury JG, Frampton CM, Rademaker MT, Richards AM. Urocortin 2 infusion in healthy humans: hemodynamic, neurohormonal, and renal responses. J Am Coll Cardiol. 2007;49:461–71. doi: 10.1016/j.jacc.2006.09.035. [DOI] [PubMed] [Google Scholar]
  • 167.Ng LL, Loke IW, O’Brien RJ, Squire IB, Davies JE. Plasma urocortin in human systolic heart failure. Clin Sci (Lond) 2004;106:383–8. doi: 10.1042/CS20030311. [DOI] [PubMed] [Google Scholar]
  • 168.Steiger A. Neuroendocrinology of Sleep. In: Lajtha A, Blaustein JD, editors. Handbook of Neurochemistry and Molecular Neurobiology. Berlin/Heidelberg: Springer US; 2007. pp. 895–937. [Google Scholar]
  • 169.Ising M, Holsboer F. CRH-sub-1 receptor antagonists for the treatment of depression and anxiety. Exp Clin Psychopharmacol. 2007;15:519–28. doi: 10.1037/1064-1297.15.6.519. [DOI] [PubMed] [Google Scholar]
  • 170.Wallon C, Soderholm JD. Corticotropin-releasing hormone and mast cells in the regulation of mucosal barrier function in the human colon. Ann N Y Acad Sci. 2009;1165:206–10. doi: 10.1111/j.1749-6632.2009.04030.x. [DOI] [PubMed] [Google Scholar]
  • 171.Lenz HJ, Fisher LA, Vale WW, Brown MR. Corticotropin-releasing factor, sauvagine, and urotensin I: effects on blood flow. Am J Physiol. 1985;249:R85–90. doi: 10.1152/ajpregu.1985.249.1.R85. [DOI] [PubMed] [Google Scholar]

RESOURCES