Skip to main content
Journal of Neurogastroenterology and Motility logoLink to Journal of Neurogastroenterology and Motility
. 2015 Jan;21(1):8–24. doi: 10.5056/jnm14162

Role of Corticotropin-releasing Factor Signaling in Stress-related Alterations of Colonic Motility and Hyperalgesia

Yvette Taché 1,*, Mulugeta Million 1
PMCID: PMC4288101  PMID: 25611064

Abstract

The corticotropin-releasing factor (CRF) signaling systems encompass CRF and the structurally related peptide urocortin (Ucn) 1, 2, and 3 along with 2 G-protein coupled receptors, CRF1 and CRF2. CRF binds with high and moderate affinity to CRF1 and CRF2 receptors, respectively while Ucn1 is a high-affinity agonist at both receptors, and Ucn2 and Ucn3 are selective CRF2 agonists. The CRF systems are expressed in both the brain and the colon at the gene and protein levels. Experimental studies established that the activation of CRF1 pathway in the brain or the colon recaptures cardinal features of diarrhea predominant irritable bowel syndrome (IBS) (stimulation of colonic motility, activation of mast cells and serotonin, defecation/watery diarrhea, and visceral hyperalgesia). Conversely, selective CRF1 antagonists or CRF1/CRF2 antagonists, abolished or reduced exogenous CRF and stress-induced stimulation of colonic motility, defecation, diarrhea and colonic mast cell activation and visceral hyperalgesia to colorectal distention. By contrast, the CRF2 signaling in the colon dampened the CRF1 mediated stimulation of colonic motor function and visceral hyperalgesia. These data provide a conceptual framework that sustained activation of the CRF1 system at central and/or peripheral sites may be one of the underlying basis of IBS-diarrhea symptoms. While targeting these mechanisms by CRF1 antagonists provided a relevant novel therapeutic venue, so far these promising preclinical data have not translated into therapeutic use of CRF1 antagonists. Whether the existing or newly developed CRF1 antagonists will progress to therapeutic benefits for stress-sensitive diseases including IBS for a subset of patients is still a work in progress.

Keywords: Colonic motility, Corticotropin-releasing factor, Irritable bowel syndrome, Stress, Visceral pain


Official abbreviations according to UPHARM guideline were used in this review series.

  • CRF, corticotrophin-releasing factor

  • CRF1, corticotrophin-releasing factor receptor 1

  • CRF2, corticotrophin-releasing factor receptor 2

Introduction

The influence of emotions on the gastrointestinal tract function have been described many centuries ago.1,2 The feelings of “butterflies” in the stomach as well as the “gut wrenching” and nauseous feelings and abdominal pain during or following emotional stress are some of the overt manifestations of the intimate brain-gut connections.3,4 The past decades have witnessed an increasing recognition of the role played by the bidirectional interaction between the brain and gut in functional bowel diseases.513 These interactions occurs through neuronal pathways via the efferent and afferent components of the parasympathetic and sympathetic nervous system,1416 hormonal route including the hypothalamic-pituitary-adrenal (HPA) axis17 and/or components of the immune system and the microbiota.18 Recent studies also identified a repertoire of neuromediators and receptors in the central nervous system and the gut that are involved in conveying the brain gut interactions in health and diseases.18 In particular one of the key mediator in the bodily response to various stressors and the brain-gut interactions is the corticotrophin releasing factor (CRF) pathways family.1921

In this review, we will first briefly outlined the components of the mammalian CRF signaling systems that encompasses CRF peptides, receptors and signaling pathways. We will review pre-clinical findings on the actions of CRF in the brain and the colon to mimic stress-related stimulation of colonic secretory-motor function and the development of visceral hyperalgesia as well as the role of CRF receptor subtypes in these colonic responses to stress. Lastly, we will address the translational application of these experimental studies to clinical settings with existing clinical trials using CRF receptor 1 (CRF1) antagonists for irritable bowel syndrome (IBS).

Corticotropin-releasing Facotor Peptides and Receptors

Corticotropin-releasing Factor Peptides

The mammalian CRF family of peptides consists of 4 distinct paralogs including CRF, and urocortin (Ucn) 1, Ucn2, and Ucn3.22 CRF is a 41-amino acid (a.a.) first isolated from ovine brain by Vale et al23 The peptide structure is highly conserved among a large variety of mammalian species including human, primate, carnivore and rodents.22,24 Similar to CRF, the structure of Ucn1 is highly conserved across mammalian species with rat, mouse and sheep primary sequences sharing 100% identity, and 95% with that of human.25 Subsequently, the simultaneous cloning of 2 novel putative CRF-related peptides by 2 independent groups26,27 resulted in divergent nomenclatures that were normalized in the UPHARM guideline.28 The mouse Ucn2, a 38-a.a. peptide, shares 34% homology with human/rat/mouse (h/r/m) CRF and 42% with r/mUcn1.29 Mouse Ucn3 and hUcn3 (also named stresscopin)26 share 90% identity with each other and are more distantly related to h/r/mCRF, h/rUcn1, and mUcn1 (18% and 21% homology respectively).27

Corticotropin-releasing Factor Receptors

CRF and CRF-related peptides exert their biological actions by binding to 2 specific G-protein coupled receptors (GPCR), CRF1 and CRF2 (415 a.a. and 411 a.a., respectively).22,30 The 2 receptors are encoded by 2 distinct genes31 and display differential pharmacological and anatomical profiles indicative of distinct functional roles.28,32 CRF1 and CRF2 receptors share 70% identity within their species homologues.28 The most variable component is the binding domain that encompasses the N-terminal and the 3 extracellular coils that share only 40% homology between the 2 receptor subtypes. In their N extracellular domains, CRF receptors contain several potential points of N-glycosylation along with several Cys residues that form disulfide bonds closely associated with their functionality.33

As part of class B1 GPCRs containing exon-intron organization, CRF receptors are subject to extensive alternate splicing. A growing number of CRF1 receptor splice variants (> 10) with specific amino acid deletions has been identified in the brain and peripheral tissues including the skin, gut and placenta.3436 The main functional CRF1 receptor is CRF1a, which results from the excision of exon 6. This is a unique feature to humans since in most mammals, the exon 6 is absent and the fully active CRF1 receptor protein arises from transcription of 13 exons present within the CRF1 receptor gene sequence.37 In humans, the CRF2 gene consists of 12 exons and alternate splicing of exon 1 gives rise to three functional membrane isoforms: α (a), β (b) and γ (c) differing in the length of their N-terminal domains while in rodents, only the CRF2a and CRF2b are expressed.28 In addition, recently, five variants of the CRF2a receptors have been identified.38 CRF1 and CRF2 receptor variant expression is tissue specific, regulated by specific physiological conditions and affected by environmental stimuli.35,3840 There are growing evidence that these isoforms modulates CRF signaling.37,41,42

Corticotropin-releasing Factor Ligand-receptor Selectivity

Radioligand binding and functional studies, established that the CRF1 receptor has high affinity to h/r/mCRF (1.9 nM) and rUcn1 (0.3 nM) but no appreciable binding affinity (> 100 nM) to Ucn2 and Ucn3.28,33 In contrast, rUcn, h/mUcn2 and mUcn3 bind CRF2 receptor with greater affinity (0.3, 1.7/2.1, and 5.0 nM respectively) than h/r/mCRF (30.7 nM), making this receptor subtype highly selective for Ucns signaling.25,29,30,33,43 Although CRF is a preferential CRF1 agonist, no endogenous ligand with selectivity for CRF1 receptors have been identified. Recently, non-natural peptides, namely cortagine and stressin1-A have been developed which display selective binding to CRF1 receptors.44,45

The structural and mechanistic basis of natural peptide agonists interaction with the CRF1 receptor has been delineated and much advancement has been made as recently reviewed in details.33,46 In particular, it is well established that the α-helical conformation of CRF peptides in hydrophobic or amphiphilic milieu plays an important role to convey the biological activity of these peptides. The structure-activity relationship based on CRF1 mediated ACTH release established that the first 21 a.a. of the N-terminal and 8 residues of the C-terminal regions of CRF are required for the peptide to be biologically active.33 The current state-of-knowledge posits a “two-step” model of ligand-receptor interaction whereby the C-terminal portion of CRF (about 33–40 a.a.) is essential to bind the extracellular N-terminal domain of the CRF1 receptor (N-domain). This brings the N-terminus (about 10 a.a.) of the peptide in the optimal orientation to bind to the J-domain which is formed by the seven transmembrane helices and three extracellular loops responsible for the coupling to Gs- and Gi-proteins.33,34 Recent studies also indicate that the molecular basis for ligand selectivity involves a single amino acid difference in the N-terminal extracellular domains of the receptors (CRF1 Glu104/ CRF2a Pro-100) at a site proximate to peptide residue 35 (Arg in CRF/Ucn1, Ala in Ucn2/3).47

In contrast to the CRF1 receptor isoforms,34,36 binding characteristics of CRF receptor splice variants, CRF2a, CRF2b, and CRF2c are almost identical with high affinity for Ucn1, Ucn2, and Ucn3, and lower affinity for r/hCRF.27,4850 Unexpectedly, the mouse soluble CRFR2a isoform displays very low affinity for Ucn2 and Ucn3 while binding to Ucn1 (Ki 6.6 nM) and to a lesser extent to CRF (23 nM). Activation of this isoform also inhibits cAMP and extracellular signal-regulated kinases 1 (ERK1/2)-p42,44 responses to Ucn1 and CRF.41 In contrast, rat CRF2a-tr binds with low affinity to CRF (Kd 12.7 nM) while displaying no affinity to Ucn1.51

Corticotropin-releasing Factor Receptor Signaling

As members of the class B1 class of GPCR, CRF receptors are primarily coupled to Gαs, the hallmark feature known to this GPCR subfamily. Ligand receptor interaction initiates several intracellular cAMP-dependent signaling cascades leading to acute post-translational modification of target proteins by protein kinase A (PKA) in the cytoplasm and gene transcription regulation by cAMP response element-binding proteins activation in the nucleus.46 However, CRF ligand-receptor interaction can diverge to a plethora of downstream intracellular effectors, such as guanyl cyclase, ERK1/2, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) transcription factor, ion channels, tyrosine hydroxylase phosphorylation and glycogen synthase kinase 3 beta/Wnt/b-catenin pathway.34,46,52 In general, while CRF1 and CRF2 receptor signaling is mediated primarily through Gαs and thus cAMP as the second messenger,33,34 a low affinity binding site for the peptide ligand can also activate Gi- and Gq/11-coupled state.46,53 The occurrence of various signaling pathways is cell type and ligand specific.46 The several described signaling pathways of CRF ligand-receptor interaction have been delineated in several cell lines or in specific organs,46 and more recently, in colonic endocrine, neuronal or epithelial cells.36,54,55

Corticotropin-releasing Factor Receptor Antagonists

Key to the assessment of the role of endogenous CRF ligands and CRF receptors in the stress response was the early development of several N-terminally truncated and substituted CRF analogs developed by Jean E Rivier.56 Overall, four groups of CRF receptor antagonists have been developed which include the specific, nonselective peptide CRF1 and CRF2 receptor antagonists namely α-helical CRF9–41, D-Phe12CRF12–41, astressin and the acting astressin-B; the selective peptide CRF2 receptor antagonists, anti-sauvagine and astressin2-B;56 the recently developed selective peptide CRF1 antagonist, YAM19 which represents the only peptide CRF1 receptor antagonist57; and the numerous non-peptide CRF1 antagonists.33,58,59 All these antagonists have been widely used as tools to determine the CRF receptor sub-types mediating stress-induced behavioral, endocrine and autonomic responses.58,60,61 While upon peripheral administration, the non-peptide CRF1 receptor antagonists readily cross the blood-brain barrier as recently reviewed,59,62 the peptide CRF antagonists have no or very low ability to cross the barrier.63 This allows us to delineate the central and/or peripheral role of CRF receptor upon injection of antagonists into the brain or the periphery respectively. For instance, we showed that intravenous (iv) injection of astressin, at a dose that blocked intravenous CRF-induced delayed gastric emptying, did not influence CRF injected into cisterna magna-induced delayed gastric transit in rats.64 Likewise, α-helical CRF9–41 infused intravenously did not influence the suppression of pentagastrin stimulated acid secretion evoked by intracerebroventricular (icv) injection of CRF, while, injected icv, the antagonist blocked the icv CRF action in rats.65

Role of Brain Corticotropin-releasing Factor Receptor 1 in Stress-related Stimulation of Colonic Motor Function

Brain Distribution of Corticotropin-releasing Factor–Cordicotropin-releasing Factor Receptor 1

CRF peptides and receptors are widely distributed in the brain including specific regions linked with anxiogenic and digestive behaviors.32,6669 In particular, major sites of CRF mRNA and CRF immunoreactivity are located in the paraventricular nucleus (PVN) of the hypothalamus, cerebral cortex, amygdalar-hippocampal complex and pontine Barrington’s nucleus in rodents and humans.67,68 With regards to the brain distribution of CRF receptors, dense CRF1 receptor expression is found in the forebrain, subcortical limbic structures (more prominently in the septal region) and amygdala, whereas the expression in the hypothalamus is low under basal conditions but markedly up-regulated by stress.32,70,71 Moreover, CRF1 receptors at the gene and protein levels are prominently expressed in the anterior and intermediate lobe of the pituitary.60

Activation of Brain Corticotropin-releasing Factor Receptor 1 Is Involved in the Colonic Response to Stress

Consistent with the brain localization of CRF/CRF1 signaling pathway, convergent preclinical reports point to the role of central CRF1 receptors in stress-related stimulation of colonic secretory motor function.61 First, CRF and Ucn1 injected into the cerebrospinal fluid induces the occurrence of colonic spike burst activity, acceleration of colonic transit, decreased in colonic fluid absorption, stimulation of defecation, and induction of diarrhea in freely moving rats, mice and gerbils, mimicking the effects of exposure to acute stressors whereas Ucn2 and Ucn3 under the same conditions have no effect.7280 Secondly, pharmacological blockade of CRF receptors using icv injection of peptide CRF1/CRF2 receptor antagonists, α-helical CRF9–41, D-Phe12CRF12–41, or astressin inhibited the stimulation of defecation, as well as the increase in wet stool and colonic spike-bursts induced by icv CRF, wrap restraint, water avoidance stress (WAS) and/or conditioned fear.72,75,76,8186 Furthermore, the CRF1 receptor antagonists, CP-154,526, CRA 1000, NBI-27914, NBI-35965, antalarmin, JTC-017, NGD 98-2, and NGD 9002 injected either centrally or peripherally (as they cross the blood brain barrier), dampened icv CRF and various stressors including acute restraint, WAS, elevated plus maze, social intruder or exposure to chronic heterotypic stressors (WAS, forced swim, cold restraint, and restraint-induced stimulation of colonic motor function).74,80,84,8790 By contrast the selective CRF2 antagonist, astressin2-B injected centrally at doses that blocked CRF2 receptor-mediated inhibition of gastric emptying, did not alter restraint stress-related defecation in mice or chronic heterotypic stress.74,89 In addition CRF1 receptor knockout mice showed significantly less defecation in an open field test than did the wild-type littermates.91 Importantly, brain CRF1 receptors are not involved in the basal and postprandial regulation of colonic motor function under non-stress conditions.92

Brain Sites of Action of Corticotropin-releasing Factor-induced Stimulation of Colonic Motor Function

Brain nuclei responsive to CRF leading to the stimulation of colonic motor function (increased in tonic and phasic colonic motility, decreased colonic transit time and induction of watery fecal output) have been localized in the hypothalamus (PVN, arcuate nucleus) and pontine areas, namely the locus coeruleus (LC)/Barrington nucleus complex,9396 which are also brain nuclei involved in central CRF-induced anxiety and depression.97,98 Anatomical support for the role of pontine and PVN CRF signaling pathways in the regulation of pelvic organ functions came from tracing studies showing that a proportion of CRF immunoreactive neurons in the Barrington’s nucleus and PVN are linked transynaptically to the colon.99,100 Of interest is the growing pharmacological evidence that a number of brain peptides influencing food intake, such as neuropeptide Y, glucagon-like peptide, cocaine- and amphetamine-regulated transcript (CART), and ghrelin, act in the brain to stimulate colonic motor function through CRF receptor activation-dependent mechanisms in the hypothalamus.101106

Brain Corticotropin-releasing Factor and Colonic Response: Peripheral Mechanisms

Peripheral mechanisms involved in colonic secretory motor stimulation in response to central CRF or stress (restraint or WAS) is independent from the activation of the HPA axis and brought about through the activation of sacral parasympathetic outflow and related input to the enteric nervous system (ENS) activity.73,82,85 The ganglionic blocker, chlorisondamine prevented and subdiaphragmatic vagotomy attenuated the stimulation of colonic transit, phasic and tonic contractions, and defecation induced by CRF injected icv or into the PVN while bretylium (noradrenergic blocker) had no effect.73,95,96,103 Effector mechanisms within the colon involved parasympathetic cholinergic mediated activation of colonic serotonin (5-HT) interacting with 5-HT3 and 5-HT4 receptors. This is supported by the increase in 5-HT content in the feces of the rat proximal colon in response to icv CRF107 and pharmacological approach using 5-HT3 antagonists (granistron, ramosteron, ondansetron and azasetron), the 5-HT4 antagonist, SB-204070 and the muscarinic antagonist, atropine that all blocked icv CRF-induced colonic motor stimulation and decreases in colonic fluid loss.76,78,88,107

Role of Colonic Corticotropin-releasing Factor Receptors in Stress-related Stimulation of Colonic Motor Function

Activation of Peripheral Corticotropin-releasing Factor Receptor 1 Contributes to the Colonic Response to Stress

Functional studies showed that CRF and Ucn1 injected peripherally are as potent as injected centrally to increase propagative clustered spike-burst activity in the proximal colon and to stimulate distal colonic transit, defecation and to induce prominent diarrhea in rodents.54,72,76,108115 The stimulation of colonic secretory-motor function after peripheral administration of CRF or Ucn1 involves CRF1 receptors in rats and mice. This is supported by the fact that elective CRF1 agonist, cortagine or stressin1 injected intraperitoneally stimulates colonic motor function and induces diarrhea while the CRF2 agonists, Ucn2 has not effect under the same conditions.54,108,110,111,116,117

The colonic CRF-CRF1 signaling may have relevance as peripheral effector of the acute stress response. There is evidence that stressors that stimulate colonic motor function such as endotoxin or early maternal separation upregulates CRF-CRF1 signaling in the colon.118,119 In addition peripherally injected peptide antagonists α-helical CRF9–41, astressin or astressin-B blocked or blunted the stimulation of distal colonic transit and high amplitude high frequency contraction and fecal pellet output induced by acute restraint or WAS in rats or mice.72,76,110,113,115

Corticotropin-releasing Factor Receptor 1 in the Colon: Mechanisms to Stimulate Propulsive Motor Function

Recent studies establishing the expression of both CRF1 receptor and ligands in the colon at the gene and protein levels in various cells including neuronal (ENS), endocrine (enterochromaffin cells) and immune cells (mast cells, eosinophiles, T-helper lymphocytes in the lamina propria) in rodents.54,116,118,120122 This provides a neuroanatomical substrata pointing toward a local role CRF signaling pathways as recently reviewed.123

Consistent with a local action, intraperitoneal injection of CRF-induced colonic stimulation is not affected by ganglion blockade, in contrast with the evidence on the role of the autonomic nervous system in mediating central CRF-induced stimulation of colonic propulsive motor function.73,96 Further support for direct action within the colon came from in vitro studies in isolated colonic rat preparations or isolated rat colonic muscle strips where CRF increased basal myoelectrical peristaltic activity and phasic contractions and electric field stimulation off-contractions.110,122,124

Convergent evidence substantiates that the peripheral stimulatory action of CRF or Ucn1 involved the increase ENS neuronal activity in cholinergic, nitrergic and serotonergic neurotransmission promoting peristaltic through the activation of CRF1 receptors on ENS in rats or guinea pigs.54,107,116,122,125 First, the use of laser captured combined with reverse transcriptase polymerase chain reaction or immunohistochemical detection in myenteric whole mount preparation of the colon revealed that CRF1 receptors are primarily expressed at the gene and protein levels on myenteric neurons compared with other layers of the rat or guinea colon under basal conditions.54,116,122,125 Second, the neuronal blocker, tetrodotoxin abolished Ucn1-evoked phasic contractions in rat colonic smooth muscle strips,122 indicative of an ENS mediated component. Third, when injected intraperitoneally, CRF in rats or partial restraint in mice induces Fos expression, a marker of neuronal activation,126 in CRF1 receptor expressing cholinergic and nitrergic myenteric neurons in the proximal colon while Ucn2 under the same conditions did not induce Fos.54,116,117,127,128 It is to note that atropine, a muscarinic blocker, does not affect intraperitoneal CRF-induced neuronal activation in colonic myenteric ganglia, indicating that the Fos response is not secondary to the activation of muscarinic receptors either on the myenteric ganglia (which possess both nicotinic and muscarinic receptors) or on colonic smooth muscle cells but rather to a direct effect on enteric neurons.127 Additional support came from electrophysiological recording showing that direct administration of CRF or Ucn1 onto colonic myenteric and submucosal plexus preparations of guinea pig excites both myenteric and submucosal neurons through CRF1 receptor.125,129,130 Other studies in rat submucosal neuron preparation showed that CRF induces a tetradotoxin sensitive and CRF1 receptor mediated neuronal activation monitored by the increase in intracellular calcium.131

Other peripheral molecular pathways of the potent stimulation of colonic secretory motor function by peripheral injection of CRF may include the crosstalk between the enteric neuroendocrine and immune systems. CRF added in vitro to rat colonic section up-regulates IL-6 in the colonic tissues and potentiates IL-6 induced submucosal neuron activity of the colon through CRF1 receptor.131

Corticotropin-releasing Factor Receptor 2 in the Colon: Modulation of Stress-related Stimulation of Propulsive Motor Function

The CRF2 ligands, Ucns and CRF2 receptors are also expressed in rodent colon.54,55,123,132135 The CRF2 receptors are localized at the gene and protein levels in cells of the rodent colonic ENS54 although less prominently than that CRF1 receptor130,131 and human colonic lamina propria mononuclear cells,135 subepithelial mast cells136 and epithelial cells55 supporting a local action to influence neuronal and immune responses.

Recent studies indicate that intraperitoneal pretreatment with Ucn2 acting through CRF2 receptors inhibits peripheral CRF-or Ucn1 induced stimulation of colonic motor function while pre-treatment with astressin2-B or genetic deletion of CRF2 in mice exacerbated the restraint stress or intraperitoneal CRF and Ucn1 induced stimulatory colonic responses.54,137 These data are indicative that acute stress-related colonic stimulation engages not only the colonic CRF1-mediated enteric stimulatory pathway but also CRF2 to dampen the colonic response to stress. This interaction is occurring in myenteric neurons as evidenced by the localization of CRF2 receptors on CRF1 expressing myenteric neurons and the reduction of CRF-induced ERK1/2 signaling in the rat colon myenteric primary neurons by CRF2 receptors activation.54 The differential effects of CRF1 and CRF2 receptors are also reported to exist in stress-induced intestinal mucosal barrier function alterations. In pigs exposed to early-weaning stress, intestinal barrier dysfunction and hypersecretion is mediated through CRF1 activation whereas activation of peripheral CRF2 plays a protective role in the alterations of intestinal barrier function in response to early life stress.138

Role of Brain Corticotropin-releasing Factor Receptors in Stress-related Visceral Hyperalgesia

Corticotropin-releasing Factor Receptor 1 Signaling Is Involved in the Stress-related Hyperalgesia

Gué et al139 provided the first evidence that CRF injected icv induced visceral hyperalgesia to colorectal distention (CRD) in rats, mimicking the response induced by partial restraint stress. Furthermore, the icv injection of the CRF1/CRF2 antagonists, α-helical CRF9–41 prevented the effects of restraint pointing to the involvement of central CRF signaling in acute stress-related development of visceral hyperalgesia.139 Subsequent studies using selective CRF1 antagonists,59 namely NBI-35965, NBI-27914, NBI-30775, CP-154,526, CP-376395, antalarmin, JTC-017, DMP-696, NGD 98-2, or NGD 9002 supported the involvement of CRF1 subtype in the hyperalgesic response to CRD induced by icv CRF injection80,140 or in a variety of experimental models inducing visceral hyperalgesia including acute or repeated exposure to WAS alone or combined with neonatal maternal separation, consecutive sets of nociceptive CRD, repeated daily CRD 6 weeks after the development of colitis, intracolonic infusion of 0.5% acetic acid, or performing the CRD in a high-anxiety rat strain, the Wistar Kyoto (WKO).80,90,140152 Likewise in CRF1 receptor knockout mice, the visceral motor response to phasic CRD is reduced.149 These consistent pre-clinical reports indicate that CRF1 signaling plays a critical role in the visceral hyperalgesia occurring in different contexts: early life adverse events, repeated psychological stress in adulthood, chronic high anxiety as well as peripherally initiated mechanisms associated with previous colonic inflammation or repeated nociceptive CRD.

By contrast, the central effects of CRF2 receptor activation on visceral pain are scant. In a recent study Lee et al152 indicates that perfusion of selective CRF2 receptor antagonist, astressin2-B into the anterolateral BNST reduced visceral motor response to low CRD pressure in rats. Further studies are needed to assess the contribution of brain CRF2 receptors in the modulation of visceral pain.

Mechanisms Involved in Corticotropin-releasing Factor Receptor 1 Mediated Prevention of Visceral Hyperalgesia

Brain sites responsive to CRF and CRF1 antagonist to influence stress-related visceral hyperalgesia encompass the hippocampus, anterior lateral bed nucleus of the stria terminalis (BNST) and the central amygdala (CeA). One study showed that the CRF1/CRF2 antagonists, α-helical CRF9–41 microinjected into the hippocampus or peripheral injection of the CRF1 antagonist, JTC-017 decreased abdominal contraction frequency evoked by nociceptive tonic CRD along with the anxiety response to CRD in rats.90 There is also recent evidence that the CRF1 antagonist, CP-376395 microinjected into the anterolateral BNST after repeated WAS reduced the visceral motor response to CRD.152 Converging studies also point CeA as a responsive site to the CRF1 antagonist, CP-376395 to reduced visceral hyperalgesia to CRD in the WKO rat strain.153 Conversely CRF microinjected into the CeA increased sensitivity to CRD performed at 20 to 80 mmHg in Wistar rats through CRF1 receptor as shown by the blockade of CRF action by the intra-CeA injection of CP-154,526.154 Neuroanatomical evidence that CRF is densely expressed in cell bodies and axon terminals of cortical amygdala projecting to widespread regions of the basal forebrain and pontine/brainstem including the locus coeruleus (LC) support the notion that endogenous CRF acts as a ligand to act on CRF1 receptor in the CeA.155,156 Moreover CRF-like immunoreactivity and gene expression in CeA are increased in response to CRD157 and various conditions inducing visceral pain or hyperalgesia.158161

The underlying mechanisms through which blockade of brain CRF1 receptor activation influences the development of stress or mechanosensitization-related visceral hyperalgesia are still not fully understood. One component may involve the dampening of pontine noradrenergic output from the visceral to the forebrain. Recent electrophysiologic reports in anesthetized rats showed that the peptide antagonist D-Phe12CRF12–41, or astressin injected into the cerebrospinal fluid or directly into the LC and the selective CRF1 antagonist, NBI-35965, injected iv prevented the activation of the LC noradrenergic neurons responsive to both central injection of CRF and CRD.162164 Consistent with electrophysiological demonstration of LC noradrenergic silencing by CRF receptor antagonists, the CRF1 antagonist, JTC-017 reduces noradrenaline release in the hippocampus and CeA induced by CRD.90,154 The release of noradrenaline in the cortical and limbic rostral efferent projections from the LC or CeA165 is known to induce arousal and anxiogenic responses along with hypervigilence to visceral input which is a commonly found feature in IBS patients.166,167

Role of Colonic Corticotropin-releasing Factor Receptors in Stress-related Hyperalgesia

Corticotropin-releasing Factor Receptor 1

Recent reports suggest that enhanced CRF/CRF1 receptor signaling in the colon, in addition to central activation, bears relevance as part of the peripheral efferent components responsible for the induction of visceral hyperalgesia. There is evidence that peripheral injection of CRF induces visceral hypersensitivity to CRD, an effect reproduced by the intraperitoneal administration of the selective CRF1 agonist, cortagine in rats and mice.117 Conversely in rats repeatedly exposed to WAS for 10 days, peripheral injection of peptide CRF antagonist astressin before each stress session prevented the development of visceral hyperalgesia supporting the participation of a peripheral component to the development of visceral hyperalgesia.144 This is further supported by the demonstration that the visceral hyperalgesia induced by cortagine injected peripherally is abolished by peripheral, but not central, injection of astressin at an equivalent dose.117

Several mechanisms are involved in the visceral hyperalgesia linked with the activation of CRF1 receptors in the colon. Peripheral CRF induces colonic mast cells degranulation.168170 which can in turn lead to the release of several preformed or newly synthesized mediators eg, histamine, tryptase, prostaglandin E2, and nerve growth factor known to activate or sensitize sensory afferents.171,172 The involvement of colonic mast cell is further supported by the use of mast cell stabilizer, doxantrazole, that prevented acute stress-induced colonic hypersensitivity to a second set of CRD.139,169 Additionally, peripheral CRF signaling activation disrupts the intestinal epithelial barrier and CRF1 receptor antagonists block stress-induced increases in intestinal permeability.123,168,173 Such CRF receptor mediated alteration of intestinal permeability increases the penetration of soluble factors (antigens) into the lamina propria, which can result in nociceptors sensitization.174 Increased intestinal permeability is indeed a phenomenon that appears as a prerequisite for the development of visceral hyperalgesia in rodents.175

Corticotropin-releasing Factor Receptor 2

In contrast, activation of CRF2 by intraperitoneal injection of the selective CRF2 peptide agonist, Ucn2, as well as sauvagine, a non selective CRF1/ CRF2 agonist with higher affinity to CRF2, blunt repeated CRD- induced visceral sensitization.146,176 This indicates opposite effects of CRF1 and CRF2 signaling in the mechanosensitization of the colon. Similarly, CRF2 activation by intraperitoneally injected Ucn2 suppressed CRD- induced spinal phosphorylation of ERK 1/2 and visceral pain, although the exact site of CRF2 activation to inhibit spinal ERK 1/2 activity is not known.176

Corticotropin-releasing Factor Signaling in Healthy and Irritable Bowel Syndrome Subjects

Colonic Responses to Peripheral Corticotropin-releasing Factor Signaling

The actions of peripheral CRF on the colon reported in experimental studies have been largely reproduced in healthy subjects or IBS patients along with preliminary evidence of the beneficial effect of peripheral injections of peptide CRF antagonist (Tables 1 and 2). With regards to the intestinal motility, one clinical report showed that systemic administration of CRF increases the motility index in the descending colon in both healthy and IBS groups with a significant higher response in the IBS groups.177

Table 1.

Clinical Studies Showing Mimicry Between Activation of the Corticotropin-releasing Factor Signaling System and Irritable Bowel Syndrome-related Symptoms in Humans

CRF agonists stimulate/activate/increase in healthy humans or IBS patients References
  HPA axis 21
  Colonic motility 21
  Permeability/bacterial translocation 184
  Viscerosensitivity 178,179
  Mast cells 184
  Anxiety/depression 208
  Inflammation 183
CRF antagonists reverse IBS-associated increase of
  Anxiety +/− 194
  Motility +/− 21
  Permeability 184
  Viscerosensitivity 21

CRF, corticotrophin-releasing factor; IBS, irritable bowel syndrome; HPA, hypothalamic-pituitary-adrenal.

Table 2.

Preclinical Studies Highlighting the Relevance of Corticotropin-releasing Factor Receptor 1 Blockade to Reduce Irritable Bowel Syndrome-like Symptoms (see reviews21,123,209,210)

Clinical features of IBS-diarrhea predomient patients CRF1 antagonists in experimental animals block stress-related
Co-morbidity with anxiety and depression Anxiety/depression
Hypervigilance CRD-induced locus coeruleus activation-induced hypervigilance
Changes in autonomic functions (↓ vagal, ↑ sympathetic) Autonomic responses (↓ vagal, ↑ sympathetic)
Increased bowel movements/diarrhea Increased colonic motility/defecation/diarrhea
Ion transport dysfunction Colonic mucosal barrier dysfunction
Change in mast cells (number, and activation) Activation of mast cells
Increase barrier permeability Increase barrier permeability/antigen translocation
Lower pain threshold to colorectal distention Hypersensitivity to colorectal distention

IBS, irritable bowel syndrome; CRF1, corticotrophin-releasing factor receptor 1; CRD, colorectal distention.

Reports assessing changes in visceral pain established that systemic injection of the preferential CRF1 agonist, ovine CRF lowered pain thresholds to repetitive rectal distensions and enhanced the intensity of discomfort sensation to CRD in healthy humans.177179 Beneficial effects of systemic injection of α-helical CRF9–41 was reported in a setting of electrical rectal stimulation which evoked abdominal pain and anxiety in IBS patients compared with healthy subjects.180 There is also clinical evidence that systemic administration of the peptide antagonist normalizes the altered EEG activities in IBS patients under basal and in response to CRD.181

Similar to rodent data, CRF receptors have been identified in human colonic biopsies and enteric nervous system.136,182,183 CRF administered intravenously activates subepithelial mast cells, and stimulates transcellular uptake of protein antigens in colonic biopsies of healthy subjects.184 Since increased uptake of antigen-sized macromolecules is associated with inflammation, and there is increasing evidence that IBS patients display a low graded colonic inflammation, intraepithelial lymphocytes, mast cell degranulation and increased permeability, these changes may be consistent with enhanced colonic CRF signaling.185,186 Additionally, recent evidence showed the expression of CRF1 receptors and variants on human enterochromaffin-like cells, the BON subclone 1 cells. In these cells, CRF induces a CRF1 receptor mediated stimulation of 5-HT release and up regulated the expression of 5-HT synthesizing enzyme, tryptophan hydroxylase.36 Considering the important role of 5-HT in the pathophysiology of IBS,187 this may represents an additional site of action of CRF to increase colonic motility and visceral pain in human subjects.

Does Targeting Corticotropin-releasing Factor Receptor Have Therapeutic Values?

The vast amount of preclinical studies showing efficacy of CRF1 antagonists in various experimental IBS models raised high expectation regarding potential translational applications of CRF1 antagonists for therapeutic benefit in stress-sensitive human disorders. However, while a large number of small molecules CRF1 antagonists have been disclosed and used in a pre-clinical setting,59 only a few compounds have progressed to clinical trials.188 Indeed, many of these small molecule CRF1 antagonists encountered several set backs related to unattractive pharmacokinetics, prominent tissue accumulation due to lipophilicity, long elimination half-life, high protein binding, or reactive metabolite formation59,189 that precluded their clinical testing. Efforts are ongoing to overcome these issues as shown by recently disclosure of new compounds that are less lipophilic with improved pharmacokinetic properties due to the substitution of the propyl groups at the C7 position of the pyrazolol[1,5α]pyrimidine core with heterocycles.33,189192 The CRF1 antagonists which are or have reached clinical trials include CP-316311 (Pfizer), pexacerfont (Bristol-Myers Squibb), GSK-876008 and GS-561679 (GlaxoSmithKline), R317573 (Johnson & Johnson), and few others, see review.58,59 CRF1 antagonists such as NBI-30545 and NBI-35965 have the distinct advantage to be orally active and water soluble.58,59,80,192

Several clinical studies provided proof-of-concept that oral administration of CRF1 antagonists dampened stress-related biological responses in healthy human subjects. NBI-34041/SB-723620 given at 100 mg/day for 14 days reduced the Trier Social Stress Test and mental arithmetic test-induced rise in plasma levels of ACTH and cortisol in healthy male volunteers while not influencing basal circadian release of ACTH.193 Further clinical testing, however, was not continued due to preclinical toxicology studies. In a clinical model of generalized anxiety induced by 20-min inhalation of 7.5% CO2, preliminary studies in 12 healthy subjects showed that R317573 (40 mg once daily) given for 7 days significantly reduced panic symptom score and generalized anxiety compared to placebo.194 Positron emission tomography (PET) studies indicated that the acute administration of R317573 at 30 and 200 mg results in dose-related changes in regional glucose metabolism in regions relevant to mood and anxiety disorders.195 A recent clinical study using functional magnetic resonance imaging showed that an acute oral administration of CRF1 antagonist, GW876008 (single dose of 20 or 200 mg orally) dampened the amygdala activation produced by the anticipation of visceral pain compared to placebo drug in IBS female patients.196 Of note, CRF-CRF1 signaling in the CeA is also well established to be a key component of the neuronal circuitry contributing to anxiety-like behavior197 and therefore well positioned to drive the reciprocal relationship between visceral pain and affective mood.198 This may provide the neuroanatomical and biochemical substrata to speculate that overactivity of the CRF-CRF1 in the CeA may underlie the comorbidity of subset of IBS patients who display hypersensitivity to CRD199 and mood disorders.200

However, whether the existing or newly developed CRF1 antagonists59 will progress to show therapeutic benefits in subsets of IBS is still to be established. An early clinical trial in IBS-diarrhea predominant patients did not show beneficial effect of oral administration of the CRF1 antagonist paxacerfont (BMS-562086) on IBS symptoms although a dose-related trend to reduce visceral pain was observed at 25 mg and 100 mg for 2 weeks.201 Several aspects may have been suboptimal including the dosing regimen as the CRF1 mediated ACTH levels were not monitored in this clinical trials.201 The lack of information on the central and peripheral occupancy of CRF1 antagonists in humans in the absence of PET ligand as well as the potential role of the more than 10 splice variants of the CRF1 receptor (some of which are functional)36,40 are additional factors. Efforts are undergoing to test radioligand for the CRF1 receptor to allow rational dose selection relying on large occupancy in the brain and colon. Importantly CRF1 antagonists may exert beneficial effects only under conditions of elevated brain/colonic CRF/CRF1 signaling pathways that cannot be identified in the heterogeneous populations tested so far.

Conclusion

Both experimental and clinical studies in healthy subjects support that the activation of CRF1 pathways recaptures cardinal features of IBS-diarrhea symptoms (anxiogenic/hypervigilance behavior, colonic mast cell activation and serotonin release, stimulation of colonic propulsive motor function and watery diarrhea, and visceral hyperalgesia) (Table 1). Another key preclinical evidence arises from pharmacological interventions whereby blockade of brain or peripheral CRF1 receptors prevent/blunt the development of the above CRF or stress-related functional or cellular alterations.123,202 Selective CRF1 antagonists or CRF1/CRF2 antagonists abolished or reduced exogenous CRF and stress-induced anxiogenic/depressive behavior, stimulation of colonic motility, mucus secretion, mast cell activation, defecation, diarrhea, and pain related to colonic hyperalgesia (Table 2). Therefore, the conceptual framework supports that sustained activation of the CRF1 system at central and/or peripheral sites may be one component underlying IBS symptoms. Targeting these mechanisms by CRF1 antagonists provided a relevant novel therapeutic venue. However so far these promising preclinical data have not translated yet into therapeutic use of CRF1 antagonists. Therefore, whether the existing or newly developed CRF1 antagonists will progress to therapeutic benefits for stress-sensitive diseases including IBS for a subset of patients is still a work in progress. Of note is also the inhibitory effects of CRF2 receptor activation on stress-related gut motility and sensitization, as a gut stress coping system and the possible CRF1-CRF2 cross talk in the colon54,176,203 and the brain204 that need to be taken into consideration in targeting CRF signaling for gut centric intestinal secretomotor alterations and visceral pain therapeutic purposes. CRF being pivotal in the body’s response to stressful stimuli and the extensive preclinical data indicating that dysregulation of CRF-CRF1 system is implicated in the etiology and maintenance of several stress-sensitive disorders as recently reviewed,20,205207 a better understanding of CRF signaling is likely to provide novel insights in the otherwise challenging pathophysiology of functional gut diseases such as IBS.

Footnotes

Financial support: This work was supported by the NIHDDK grants R01 DK-33061 (YT), R01 DK-57236 (YT), R01 DK-078676 (MM), DK-41301 (Animal Models Core, YT, MM), P50 AR-049550 (YT, MM), and VA Merit and Senior Scientist Awards (YT).

Conflicts of interest: None.

References

  • 1.Cabanis PJG. In: Introduction: On the relations between the physical and moral aspects of man. Mora G, Saidi MD, editors. Baltimore: Johns Hopkins University Press; 1981. p. 650. [Google Scholar]
  • 2.Beaumont W. In: Experiments and observations on the gastric juice and the physiology of digestion. Osler W, editor. New York: Dover Publications Inc; 1833. [Google Scholar]
  • 3.Fukudo S, Kanazawa M. Gene, environment, and brain-gut interactions in irritable bowel syndrome. J Gastroenterol Hepatol. 2011;26(suppl 3):110–115. doi: 10.1111/j.1440-1746.2011.06631.x. [DOI] [PubMed] [Google Scholar]
  • 4.Jones MP, Dilley JB, Drossman D, Crowell MD. Brain-gut connections in functional GI disorders: anatomic and physiologic relationships. Neurogastroenterol Motil. 2006;18:91–103. doi: 10.1111/j.1365-2982.2005.00730.x. [DOI] [PubMed] [Google Scholar]
  • 5.Taché Y. Central nervous system action of neuropeptides to induce or prevent experimental gastroduodenal ulcerations. Perspect Behav Med. 1988;5:101–112. [Google Scholar]
  • 6.Aziz Q, Thompson DG. Brain-gut axis in health and disease. Gastroenterology. 1998;114:559–578. doi: 10.1016/S0016-5085(98)70540-2. [DOI] [PubMed] [Google Scholar]
  • 7.Al Omran Y, Aziz Q. The brain-gut axis in health and disease. Adv Exp Med Biol. 2014;817:135–153. doi: 10.1007/978-1-4939-0897-4_6. [DOI] [PubMed] [Google Scholar]
  • 8.Konturek SJ, Konturek JW, Pawlik T, Brzozowski T. Brain-gut axis and its role in the control of food intake. J Physiol Pharmacol. 2004;55(1 Pt 2):137–154. [PubMed] [Google Scholar]
  • 9.Stasi C, Orlandelli E. Role of the brain-gut axis in the pathophysiology of Crohn’s disease. Dig Dis. 2008;26:156–166. doi: 10.1159/000116774. [DOI] [PubMed] [Google Scholar]
  • 10.Koloski NA, Jones M, Kalantar J, Weltman M, Zaguirre J, Talley NJ. The brain–gut pathway in functional gastrointestinal disorders is bidirectional: a 12-year prospective population-based study. Gut. 2012;61:1284–1290. doi: 10.1136/gutjnl-2011-300474. [DOI] [PubMed] [Google Scholar]
  • 11.Coss-Adame E, Rao SS. Brain and gut interactions in irritable bowel syndrome: new paradigms and new understandings. Curr Gastroenterol Rep. 2014;16:379. doi: 10.1007/s11894-014-0379-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Taché Y, Bernstein CN. Evidence for the role of the brain-gut axis in inflammatory bowel disease: depression as cause and effect? Gastroenterology. 2009;136:2058–2061. doi: 10.1053/j.gastro.2009.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Slyepchenko A, Carvalho AF, Cha DS, Kasper S, McIntyre RS. Gut emotions - mechanisms of action of probiotics as novel therapeutic targets for depression and anxiety disorders. CNS Neurol Disord Drug. doi: 10.2174/1871527313666141130205242. Targets Published Online First: 30 Nov 2014. [DOI] [PubMed] [Google Scholar]
  • 14.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. 464–503. [Google Scholar]
  • 15.Lomax AE, Sharkey KA, Furness JB. The participation of the sympathetic innervation of the gastrointestinal tract in disease states. Neurogastroenterol Motil. 2010;22:7–18. doi: 10.1111/j.1365-2982.2009.01381.x. [DOI] [PubMed] [Google Scholar]
  • 16.Grundy D. Neuroanatomy of visceral nociception: vagal and splanchnic afferent. Gut. 2002;51(suppl 1):i2–i5. doi: 10.1136/gut.51.suppl_1.i2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Filaretova L. The hypothalamic-pituitary-adrenocortical system: hormonal brain-gut interaction and gastroprotection. Auton Neurosci. 2006;125:86–93. doi: 10.1016/j.autneu.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • 18.Holzer P, Farzi A. Neuropeptides and the microbiota-gut-brain axis. Adv Exp Med Biol. 2014;817:195–219. doi: 10.1007/978-1-4939-0897-4_9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Stengel A, Taché Y. Corticotropin-releasing factor signaling and visceral response to stress. Exp Biol Med (Maywood) 2010;235:1168–1178. doi: 10.1258/ebm.2010.009347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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 N Y Acad Sci. 2008;1148:29–41. doi: 10.1196/annals.1410.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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]
  • 22.Vale W, Vaughan J, Perrin M. Corticotropin-releasing factor (CRF) family of ligands and their receptors. The Endocrinologist. 1997;7:S3–S9. doi: 10.1097/00019616-199707011-00002. [DOI] [Google Scholar]
  • 23.Vale W, Spiess J, Rivier C, Rivier J. Characterization of a 41- residue ovine hypothalamic peptide that stimulates secretion of corticotropin and b-endorphin. Science. 1981;213:1394–1397. doi: 10.1126/science.6267699. [DOI] [PubMed] [Google Scholar]
  • 24.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]
  • 25.Vaughan J, Donaldson C, Bittencourt J, et al. Urocortin, a mammalian neuropeptide related to fish urotensin I and to corticotropin-releasing factor. Nature. 1995;378:287–292. doi: 10.1038/378287a0. [DOI] [PubMed] [Google Scholar]
  • 26.Hsu SY, Hsueh AJ. Human stresscopin and stresscopin-related peptide are selective ligands for the type 2 corticotropin-releasing hormone receptor. Nat Med. 2001;7:605–611. doi: 10.1038/87936. [DOI] [PubMed] [Google Scholar]
  • 27.Lewis K, Li C, Perrin MH, et al. 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;98:7570–7575. doi: 10.1073/pnas.121165198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.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;55:21–26. doi: 10.1124/pr.55.1.3. [DOI] [PubMed] [Google Scholar]
  • 29.Reyes TM, Lewis K, Perrin MH, et al. 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;98:2843–2848. doi: 10.1073/pnas.051626398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chalmers DT, Lovenberg TW, Grigoriadis DE, Behan DP, De Souza EB. Corticotropin-releasing factor receptors: from molecular biology to drug design. Trends Pharmacol Sci. 1996;17:166–172. doi: 10.1016/0165-6147(96)81594-X. [DOI] [PubMed] [Google Scholar]
  • 31.Perrin MH, Vale WW. Corticotropin releasing factor receptors and their ligand family. Ann N Y Acad Sci. 1999;885:312–328. doi: 10.1111/j.1749-6632.1999.tb08687.x. [DOI] [PubMed] [Google Scholar]
  • 32.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;15:6340–6350. doi: 10.1523/JNEUROSCI.15-10-06340.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Liapakis G, Venihaki M, Margioris A, Grigoriadis D, Gkountelias K. Members of CRF family and their receptors: from past to future. Curr Med Chem. 2011;18:2583–2600. doi: 10.2174/092986711795933704. [DOI] [PubMed] [Google Scholar]
  • 34.Hillhouse EW, Grammatopoulos DK. The molecular mechanisms underlying the regulation of the biological activity of corticotropin-releasing hormone receptors: implications for physiology and pathophysiology. Endocr Rev. 2006;27:260–286. doi: 10.1210/er.2005-0034. [DOI] [PubMed] [Google Scholar]
  • 35.Karteris E, Markovic D, Chen J, Hillhouse EW, Grammatopoulos DK. Identification of a novel corticotropin-releasing hormone type 1β-like teceptor variant lacking exon 13 in human pregnant myometrium regulated by estradiol-17β and progesterone. Endocrinology. 2010;151:4959–4968. doi: 10.1210/en.2010-0622. [DOI] [PubMed] [Google Scholar]
  • 36.Wu SV, Yuan PQ, Lai J, et al. Activation of Type 1 CRH receptor isoforms induces serotonin release from human carcinoid BON-1N cells: an enterochromaffin cell model. Endocrinology. 2011;152:126–137. doi: 10.1210/en.2010-0997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.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]
  • 38.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;148:1675–1687. doi: 10.1210/en.2006-0565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yuan PQ, Wu SV, Taché Y. Urocortins and CRF type 2 receptor isoforms expression in the rat stomach are regulated by endotoxin: role in the modulation of delayed gastric emptying. Am J Physiol Gastrointest Liver Physiol. 2012;303:G20–G31. doi: 10.1152/ajpgi.00547.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zmijewski MA, Slominski AT. CRF1 receptor splicing in epidermal keratinocytes: potential biological role and environmental regulations. J Cell Physiol. 2009;218:593–602. doi: 10.1002/jcp.21632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chen AM, Perrin MH, Digruccio MR, et al. A soluble mouse brain splice variant of type 2α corticotropin-releasing factor (CRF) receptor binds ligands and modulates their activity. Proc Natl Acad Sci USA. 2005;102:2620–2625. doi: 10.1073/pnas.0409583102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Markovic D, Vatish M, Gu M, et al. The onset of labor alters corticotropin-releasing hormone type 1 receptor variant expression in human myometrium: putative role of interleukin-1beta. Endocrinology. 2007;148:3205–3213. doi: 10.1210/en.2007-0095. [DOI] [PubMed] [Google Scholar]
  • 43.Dautzenberg FM, Hauger RL. The CRF peptide family and their receptors: yet more partners discovered. Trends Pharmacol Sci. 2002;23:71–77. doi: 10.1016/S0165-6147(02)01946-6. [DOI] [PubMed] [Google Scholar]
  • 44.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;101:9468–9473. doi: 10.1073/pnas.0403159101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rivier J, Gulyas J, Kunitake K, et al. Stressin1-A, a potent corticotropin releasing factor receptor 1 (CRF1)-selective peptide agonist. J Med Chem. 2007;50:1668–1674. doi: 10.1021/jm0613875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Grammatopoulos DK. Insights into mechanisms of corticotropin-releasing hormone receptor signal transduction. Br J Pharmacol. 2012;166:85–97. doi: 10.1111/j.1476-5381.2011.01631.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pal K, Swaminathan K, Xu HE, Pioszak AA. Structural basis for hormone recognition by the Human CRFR2α G protein-coupled receptor. J Biol Chem. 2010;285:40351–40361. doi: 10.1074/jbc.M110.186072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ardati A, Goetschy V, Gottowick J, et al. Human CRF2 a and b splice variants: pharmacological characterization using radioligand binding and a luciferase gene expression assay. Neuropharmacology. 1999;38:441–448. doi: 10.1016/S0028-3908(98)00201-9. [DOI] [PubMed] [Google Scholar]
  • 49.Kostich WA, Chen A, Sperle K, Largent BL. Molecular identification and analysis of a novel human corticotropin-releasing factor (CRF) receptor: the CRF2ÿ receptor. Mol Endocrinol. 1998;12:1077–1085. doi: 10.1210/mend.12.8.0145. [DOI] [PubMed] [Google Scholar]
  • 50.Suman-Chauhan N, Carnell P, Franks R, et al. Expression and characterisation of human and rat CRF2alpha receptors. Eur J Pharmacol. 1999;379:219–227. doi: 10.1016/S0014-2999(99)00505-1. [DOI] [PubMed] [Google Scholar]
  • 51.Miyata I, Shiota C, Ikeda Y, et al. Cloning and characterization of a short variant of the corticotropin-releasing factor receptor subtype from rat amygdala. Biochem Biophys Res Commun. 1999;256:692–696. doi: 10.1006/bbrc.1999.0392. [DOI] [PubMed] [Google Scholar]
  • 52.Grigoriadis DE, Hoare SR, Lechner SM, Slee DH, Williams JA. Drugability of extracellular targets: discovery of small molecule drugs targeting allosteric, functional, and subunit-selective sites on GPCRs and ion channels. Neuropsychopharmacology. 2009;34:106–125. doi: 10.1038/npp.2008.149. [DOI] [PubMed] [Google Scholar]
  • 53.Berger H, Heinrich N, Wietfeld D, Bienert M, Beyermann M. Evidence that corticotropin-releasing factor receptor type 1 couples to Gs- and Gi-proteins through different conformations of its J-domain. Br J Pharmacol. 2006;149:942–947. doi: 10.1038/sj.bjp.0706926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gourcerol G, Wu SV, Yuan PQ, et al. Activation of corticotropin-releasing factor receptor 2 mediates the colonic motor coping response to acute stress in rodents. Gastroenterology. 2011;140:1586–1596. e6. doi: 10.1053/j.gastro.2011.01.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Moss AC, Anton P, Savidge T, Newman P, Cheifetz AS, Gay J, et al. Urocortin II mediates pro-inflammatory effects in human colonocytes via corticotropin-releasing hormone receptor 2alpha. Gut. 2007;56:1210–1217. doi: 10.1136/gut.2006.110668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Rivier JE, Rivier CL. Corticotropin-releasing factor peptide antagonists: design, characterization and potential clinical relevance. Front Neuroendocrinol. 2014;35:161–170. doi: 10.1016/j.yfrne.2013.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yamada Y, Mizutani K, Mizusawa Y, Hantani Y, Tanaka M, Tanaka Y, et al. New class of corticotropin-releasing factor (CRF) antagonists: small peptides having high binding affinity for CRF receptor. J Med Chem. 2004;47:1075–1078. doi: 10.1021/jm034180+. [DOI] [PubMed] [Google Scholar]
  • 58.Kehne JH, Cain CK. Therapeutic utility of non-peptidic CRF1 receptor antagonists in anxiety, depression, and stress-related disorders: evidence from animal models. Pharmacol Ther. 2010;128:460–487. doi: 10.1016/j.pharmthera.2010.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zorrilla EP, Koob GF. Progress in corticotropin-releasing factor-1 antagonist development. Drug Discov Today. 2010;15:371–383. doi: 10.1016/j.drudis.2010.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Turnbull AV, Rivier C. Corticotropin-releasing factor (CRF) and endocrine response 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.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]
  • 62.Gilligan PJ. Corticotropin-releasing factor receptor antagonists. Expert Opin Ther Pat. 2006;16:913–924. doi: 10.1517/13543776.16.7.913. [DOI] [PubMed] [Google Scholar]
  • 63.Kastin AJ, Akerstrom V. Differential interactions of urocortin/corticotropin-releasing hormone peptides with the blood-brain barrier. Neuroendocrinology. 2002;75:367–374. doi: 10.1159/000059433. [DOI] [PubMed] [Google Scholar]
  • 64.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;290:629–634. [PubMed] [Google Scholar]
  • 65.Drüge G1, Raedler A, Greten H, Lenz HJ. Pathways mediating CRF-induced inhibition of gastric acid secretion in rats. Am J Physiol. 1989;256(1 Pt 1):G214–G219. doi: 10.1152/ajpgi.1989.256.1.G214. [DOI] [PubMed] [Google Scholar]
  • 66.Zorrilla EP, Koob GF. The therapeutic potential of CRF1 antagonists for anxiety. Expert Opin Investig Drugs. 2004;13:799–828. doi: 10.1517/13543784.13.7.799. [DOI] [PubMed] [Google Scholar]
  • 67.Charlton BG, Ferrier IN, Perry RH. Distribution of corticotropin-releasing factor-like immunoreactivity in human brain. Neuropeptides. 1987;10:329–334. doi: 10.1016/S0143-4179(87)90083-7. [DOI] [PubMed] [Google Scholar]
  • 68.Wang L, Goebel-Stengel M, Stengel A, Wu SV, Ohning G, Taché Y. Comparison of CRF-immunoreactive neurons distribution in mouse and rat brains and selective induction of Fos in rat hypothalamic CRF neurons by abdominal surgery. Brain Res. 2011;1415:34–46. doi: 10.1016/j.brainres.2011.07.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Justice NJ, Yuan ZF, Sawchenko PE, Vale W. Type 1 corticotropin-releasing factor receptor expression reported in BAC transgenic mice: implications for reconciling ligand-receptor mismatch in the central corticotropin-releasing factor system. J Comp Neurol. 2008;511:479–496. doi: 10.1002/cne.21848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.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(5 Pt 2):R1438–R1449. doi: 10.1152/ajpregu.1998.275.5.R1438. [DOI] [PubMed] [Google Scholar]
  • 71.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]
  • 72.Williams CL, Peterson JM, Villar RG, Burks TF. Corticotropin-releasing factor directly mediates colonic responses to stress. Am J Physiol. 1987;253(4 Pt 1):G582–G586. doi: 10.1152/ajpgi.1987.253.4.G582. [DOI] [PubMed] [Google Scholar]
  • 73.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]
  • 74.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(Pt 1):221–234. doi: 10.1113/jphysiol.2003.059659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Martinez V, Rivier J, Wang L, Taché Y. Central injection of a new corticotropin-releasing factor (CRF) antagonist, astressin, blocks CRF- and stress-related alterations of gastric and colonic motor function. J Pharmacol Exp Ther. 1997;280:754–760. [PubMed] [Google Scholar]
  • 76.Miyata K, Ito H, Fukudo S. Involvement of the 5-HT3 receptor in CRH-induce defecation in rats. Am J Physiol. 1998;274(5 Pt 1):G827–G831. doi: 10.1152/ajpgi.1998.274.5.G827. [DOI] [PubMed] [Google Scholar]
  • 77.Jiménez M, Buéno L. Inhibitory effects of neuropeptide Y (NPY) on CRF and stress-induced cecal motor response in rats. Life Sci. 1990;47:205–211. doi: 10.1016/0024-3205(90)90321-H. [DOI] [PubMed] [Google Scholar]
  • 78.Hirata T, Keto Y, Nakata M, et al. Effects of serotonin 5-HT3 receptor antagonists on CRF-induced abnormal colonic water transport and defecation in rats. Eur J Pharmacol. 2008;587:281–284. doi: 10.1016/j.ejphar.2008.03.040. [DOI] [PubMed] [Google Scholar]
  • 79.Park AJ, Collins J, Blennerhassett PA, et al. Altered colonic function and microbiota profile in a mouse model of chronic depression. Neurogastroenterol Motil. 2013;25:733–e575. doi: 10.1111/nmo.12153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Million M, Zhao JF, Luckey A, et al. The newly developed CRF1-receptor antagonists, NGD 98-2 and NGD 9002, suppress acute stress-induced stimulation of colonic motor function and visceral hypersensitivity in rats. PLoS One. 2013;8:e73749. doi: 10.1371/journal.pone.0073749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.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;641:21–28. doi: 10.1016/0006-8993(94)91810-4. [DOI] [PubMed] [Google Scholar]
  • 82.Gué M, Junien JL, Buéno L. Conditioned emotional response in rats enhances colonic motility through the central release of corticotropin-releasing factor. Gastroenterology. 1991;100:964–970. doi: 10.1016/0016-5085(91)90270-u. [DOI] [PubMed] [Google Scholar]
  • 83.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;95:1510–1517. doi: 10.1016/s0016-5085(88)80070-2. [DOI] [PubMed] [Google Scholar]
  • 84.Martinez V, Taché Y. Role of CRF receptor 1 in central CRF-induced stimulation of colonic propulsion in rats. Brain Res. 2001;893:29–35. doi: 10.1016/S0006-8993(00)03277-7. [DOI] [PubMed] [Google Scholar]
  • 85.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;877:345–353. doi: 10.1016/S0006-8993(00)02719-0. [DOI] [PubMed] [Google Scholar]
  • 86.Funatsu T, Takeuchi A, Hirata T, Keto Y, Akuzawa S, Sasamata M. Effect of ramosetron on conditioned emotional stress-induced colonic dysfunction as a model of irritable bowel syndrome in rats. Eur J Pharmacol. 2007;573:190–195. doi: 10.1016/j.ejphar.2007.06.041. [DOI] [PubMed] [Google Scholar]
  • 87.Gabry KE, Chrousos GP, Rice KC, et al. Marked suppression of gastric ulcerogenesis and intestinal responses to stress by a novel class of drugs. Mol Psychiatry. 2002;7:474–483. doi: 10.1038/sj.mp.4001031. [DOI] [PubMed] [Google Scholar]
  • 88.Ataka K, Kuge T, Fujino K, Takahashi T, Fujimiya M. Wood creosote prevents CRF-induced motility via 5-HT3 receptors in proximal and 5-HT4 receptors in distal colon in rats. Auton Neurosci. 2007;133:136–145. doi: 10.1016/j.autneu.2006.11.002. [DOI] [PubMed] [Google Scholar]
  • 89.Yoshimoto S, Cerjak D, Babygirija R, Bulbul M, Ludwig K, Takahashi T. Hypothalamic circuit regulating colonic transit following chronic stress in rats. Stress. 2012;15:227–236. doi: 10.3109/10253890.2011.614297. [DOI] [PubMed] [Google Scholar]
  • 90.Saito K, Kasai T, Nagura Y, Ito H, Kanazawa M, Fukudo S. Corticotropin-releasing hormone receptor 1 antagonist blocks brain-gut activation induced by colonic distention in rats. Gastroenterology. 2005;129:1533–1543. doi: 10.1053/j.gastro.2005.07.053. [DOI] [PubMed] [Google Scholar]
  • 91.Bale TL, Picetti R, Contarino A, Koob GF, Vale WW, Lee KF. Mice deficient for both corticotropin-releasing factor receptor 1 (CRFR1) and CRFR2 have an impaired stress response and display sexually dichotomous anxiety-like behavior. J Neurosci. 2002;22:193–199. doi: 10.1523/JNEUROSCI.22-01-00193.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Taché Y, Martinez V, Wang L, Million M. CRF1 receptor signaling pathways are involved in stress-related alterations of colonic function and viscerosensitivity: implications for irritable bowel syndrome. Br J Pharmacol. 2004;141:1321–1330. doi: 10.1038/sj.bjp.0705760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Mönnikes H, Schmidt BG, Tebbe J, Bauer C, Taché Y. Microinfusion of corticotropin releasing factor into the locus coeruleus/subcoeruleus stimulates colonic motor function in rats. Brain Res. 1994;644:101–108. doi: 10.1016/0006-8993(94)90352-2. [DOI] [PubMed] [Google Scholar]
  • 94.Mönnikes H, Raybould HE, Schmidt B, Taché Y. CRF in the paraventricular nucleus of the hypothalamus stimulates colonic motor activity in fasted rats. Peptides. 1993;14:743–747. doi: 10.1016/0196-9781(93)90107-R. [DOI] [PubMed] [Google Scholar]
  • 95.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(1 Pt 1):G137–G143. doi: 10.1152/ajpgi.1992.262.1.G137. [DOI] [PubMed] [Google Scholar]
  • 96.Mönnikes H, Schmidt BG, Taché Y. Psychological stress-induced accelerated colonic transit in rats involves hypothalamic corticotropin-releasing factor. Gastroenterology. 1993;104:716–723. doi: 10.1016/0016-5085(93)91006-4. [DOI] [PubMed] [Google Scholar]
  • 97.Mönnikes H, Heymann-Monnikes I, Taché Y. CRF in the paraventricular nucleus of the hypothalamus induces dose-related behavioral profile in rats. Brain Res. 1992;574:70–76. doi: 10.1016/0006-8993(92)90801-F. [DOI] [PubMed] [Google Scholar]
  • 98.Weiss JM, Stout JC, Aaron MF, et al. Depression and anxiety: role of the locus coeruleus and corticotropin-releasing factor. Brain Res Bull. 1994;35:561–572. doi: 10.1016/0361-9230(94)90170-8. [DOI] [PubMed] [Google Scholar]
  • 99.Rouzade-Dominguez ML, Pernar L, Beck S, Valentino RJ. Convergent responses of Barrington’s nucleus neurons to pelvic visceral stimuli in the rat: a juxtacellular labelling study. Eur J Neurosci. 2003;18:3325–3334. doi: 10.1111/j.1460-9568.2003.03072.x. [DOI] [PubMed] [Google Scholar]
  • 100.Sawchenko PE, Swanson LW. Immunohistochemical identification of neurons in the paraventricular nucleus of the hypothalamus that project to the medulla or the spinal cord in the rat. J Comp Neurol. 1982;205:260–272. doi: 10.1002/cne.902050306. [DOI] [PubMed] [Google Scholar]
  • 101.Tebbe JJ, Mronga S, Tebbe CG, Ortmann E, Arnold R, Schäfer MK. Ghrelin-induced stimulation of colonic propulsion is dependent on hypothalamic neuropeptide Y1- and corticotrophin-releasing factor 1 receptor activation. J Neuroendocrinol. 2005;17:570–576. doi: 10.1111/j.1365-2826.2005.01340.x. [DOI] [PubMed] [Google Scholar]
  • 102.Mönnikes H, Tebbe J, Bauer C, Grote C, Arnold R. Neuropeptide Y in the paraventricular nucleus of the hypothalamus stimulates colonic transit by peripheral cholinergic and central CRF pathways. Neurogastroenterol Motil. 2000;12:343–352. doi: 10.1046/j.1365-2982.2000.00212.x. [DOI] [PubMed] [Google Scholar]
  • 103.Tebbe JJ, Pasat IR, Mönnikes H, Ritter M, Kobelt P, Schäfer MK. Excitatory stimulation of neurons in the arcuate nucleus intitiates central CRF-dependent stimulation of colonic propulsion in rats. Brain Res. 2005;1036:130–138. doi: 10.1016/j.brainres.2004.12.034. [DOI] [PubMed] [Google Scholar]
  • 104.Tebbe JJ, Ortmann E, Schumacher K, et al. Cocaine- and amphetamine-regulated transcript stimulates colonic motility via central CRF receptor activation and peripheral cholinergic pathways in fed, conscious rats. Neurogastroenterol Motil. 2004;16:489–496. doi: 10.1111/j.1365-2982.2004.00561.x. [DOI] [PubMed] [Google Scholar]
  • 105.Junien JL, Gue M, Bueno L. Neuropeptide Y and sigma ligand (JO 1784) act through a Gi protein to block the psychological stress and corticotropin-releasing factor-induced colonic motor activation in rats. Neuropharmacology. 1991;30:1119–1124. doi: 10.1016/0028-3908(91)90142-X. [DOI] [PubMed] [Google Scholar]
  • 106.Nakade Y, Tsukamoto K, Iwa M, Pappas TN, Takahashi T. Glucagon like peptide-1 accelerates colonic transit via central CRF and peripheral vagal pathways in conscious rats. Auton Neurosci. 2007;131:50–56. doi: 10.1016/j.autneu.2006.06.007. [DOI] [PubMed] [Google Scholar]
  • 107.Nakade Y, Fukuda H, Iwa M, et al. Restraint stress stimulates colonic motility via central corticotropin-releasing factor and peripheral 5-HT3 receptors in conscious rats. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1037–G1044. doi: 10.1152/ajpgi.00419.2006. [DOI] [PubMed] [Google Scholar]
  • 108.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;301:611–617. doi: 10.1124/jpet.301.2.611. [DOI] [PubMed] [Google Scholar]
  • 109.Tsukamoto K, Nakade Y, Mantyh C, Ludwig K, Pappas TN, Takahashi T. Peripherally administered CRF stimulates colonic motility via central CRF receptors and vagal pathways in conscious rats. Am J Physiol Regul Integr Comp Physiol. 2006;290:R1537–R1541. doi: 10.1152/ajpregu.00713.2005. [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;119:1569–1579. doi: 10.1053/gast.2000.20251. [DOI] [PubMed] [Google Scholar]
  • 111.Million M, Maillot C, Saunders PR, Rivier J, Vale W, Taché Y. Human urocortin II, a new CRF-related peptide, displays selective CRF2-mediated action on gastric transit in rats. Am J Physiol Gastrointest Liver Physiol. 2002;282:G34–G40. doi: 10.1152/ajpgi.00283.2001. [DOI] [PubMed] [Google Scholar]
  • 112.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;435:231–235. doi: 10.1016/S0014-2999(01)01574-6. [DOI] [PubMed] [Google Scholar]
  • 113.Castagliuolo I, Lamont JT, Qiu B, et al. Acute stress causes mucin release from rat colon: role of corticotropin releasing factor and mast cells. Am J Physiol. 1996;271(5 Pt 1):G884–G892. doi: 10.1152/ajpgi.1996.271.5.G884. [DOI] [PubMed] [Google Scholar]
  • 114.Maillot C, Wang L, Million M, Taché Y. Intraperitoneal corticotropin-releasing factor and urocortin induce Fos expression in brain and spinal autonomic nuclei and long lasting stimulation of colonic motility in rats. Brain Res. 2003;974:70–81. doi: 10.1016/S0006-8993(03)02553-8. [DOI] [PubMed] [Google Scholar]
  • 115.Gourcerol G, Wang L, Adelson DW, Larauche M, Taché Y, Million M. Cholinergic giant migrating contractions in conscious mouse colon assessed by using a novel noninvasive solid-state manometry method: modulation by stressors. Am J Physiol Gastrointest Liver Physiol. 2009;296:G992–G1002. doi: 10.1152/ajpgi.90436.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.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;19:923–936. doi: 10.1111/j.1365-2982.2007.00978.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Larauche M, Gourcerol G, Wang L, et al. Cortagine, a CRF1 agonist, induces stresslike alterations of colonic function and visceral hypersensitivity in rodents primarily through peripheral pathways. Am J Physiol Gastrointest Liver Physiol. 2009;297:G215–G227. doi: 10.1152/ajpgi.00072.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Yuan PQ, Wu SV, Wang L, Taché Y. Corticotropin releasing factor in the rat colon: expression, localization and upregulation by endotoxin. Peptides. 2010;31:322–331. doi: 10.1016/j.peptides.2009.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.o’malley D, Dinan TG, Cryan JF. Alterations in colonic corticotropin-releasing factor receptors in the maternally separated rat model of irritable bowel syndrome: differential effects of acute psychological and physical stressors. Peptides. 2010;31:662–670. doi: 10.1016/j.peptides.2010.01.004. [DOI] [PubMed] [Google Scholar]
  • 120.Chatzaki E, Crowe PD, Wang L, Million M, Taché Y, Grigoriadis D. CRF receptor type 1 and 2 expression and anatomical distribution in the rat colon. J Neurochem. 2004;90:309–316. doi: 10.1111/j.1471-4159.2004.02490.x. [DOI] [PubMed] [Google Scholar]
  • 121.O’malley D, Julio-Pieper M, Gibney SM, Gosselin RD, Dinan TG, Cryan JF. Differential stress-induced alterations of colonic corticotropin-releasing factor receptors in the Wistar Kyoto rat. Neurogastroenterol Motil. 2010;22:301–311. doi: 10.1111/j.1365-2982.2009.01412.x. [DOI] [PubMed] [Google Scholar]
  • 122.Kimura T, Amano T, Uehara H, et al. 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–G910. doi: 10.1152/ajpgi.00066.2007. [DOI] [PubMed] [Google Scholar]
  • 123.Larauche M, Kiank C, Taché Y. Corticotropin releasing factor signaling in colon and ileum: regulation by stress and pathophysiological implications. J Physiol Pharmacol. 2009;60(suppl 7):33–46. [PMC free article] [PubMed] [Google Scholar]
  • 124.Mancinelli R, Azzena GB, Diana M, Forgione A, Fratta W. In vitro excitatory actions of corticotropin-releasing factor on rat colonic motility. J Auton Pharmacol. 1998;18:319–324. doi: 10.1046/j.1365-2680.1998.1860319.x. [DOI] [PubMed] [Google Scholar]
  • 125.Liu S, Gao X, Gao N, et al. Expression of type 1 corticotropin-releasing factor receptor in the guinea pig enteric nervous system. J Comp Neurol. 2005;481:284–298. doi: 10.1002/cne.20370. [DOI] [PubMed] [Google Scholar]
  • 126.Dragunow M, Faull R. The use of c-fos as a metabolic marker in neuronal pathways tracing. J Neurosci Methods. 1989;29:261–265. doi: 10.1016/0165-0270(89)90150-7. [DOI] [PubMed] [Google Scholar]
  • 127.Miampamba M, Maillot C, Million M, Taché Y. Peripheral CRF activates myenteric neurons in the proximal colon through CRF1 receptor in conscious rats. Am J Physiol Gastrointest Liver Physiol. 2002;282:G857–G865. doi: 10.1152/ajpgi.00434.2001. [DOI] [PubMed] [Google Scholar]
  • 128.Miampamba M, Million M, Yuan PQ, Larouche M, Taché Y. Water avoidance stress activates colonic myenteric neurons in female rats. Neuroreport. 2007;18:679–682. doi: 10.1097/WNR.0b013e3280bef7f8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Hanani M, Wood JD. Corticotropin-releasing hormone excites myenteric neurons in the guinea-pig small intestin. Eur J Pharmacol. 1992;211:23–27. doi: 10.1016/0014-2999(92)90256-4. [DOI] [PubMed] [Google Scholar]
  • 130.Liu S, Ren W, Qu MH, et al. Differential actions of urocortins on neurons of the myenteric division of the enteric nervous system in guinea pig distal colon. Br J Pharmacol. 2010;159:222–236. doi: 10.1111/j.1476-5381.2009.00516.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.O’Malleya D, Cryana JF, Dinana TG. Crosstalk between interleukin-6 and corticotropin-releasing factor modulate submucosal plexus activity and colonic secretion. Brain Behav Immun. 2013;30:115–124. doi: 10.1016/j.bbi.2013.01.078. [DOI] [PubMed] [Google Scholar]
  • 132.Chang J, Hoy JJ, Idumalla PS, Clifton MS, Pecoraro NC, Bhargava A. Urocortin 2 expression in the rat gastrointestinal tract under basal conditions and in chemical colitis. Peptides. 2007;28:1453–1460. doi: 10.1016/j.peptides.2007.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chatzaki E, Anton PA, Million M, et al. Corticotropin-releasing factor receptor subtype 2 in human colonic mucosa: down-regulation in ulcerative colitis. World J Gastroenterol. 2013;19:1416–1423. doi: 10.3748/wjg.v19.i9.1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Saruta M, Takahashi K, Suzuki T, Fukuda T, Torii A, Sasano H. Urocortin 3/stresscopin in human colon: possible modulators of gastrointestinal function during stressful conditions. Peptides. 2005;26:1196–1206. doi: 10.1016/j.peptides.2005.01.014. [DOI] [PubMed] [Google Scholar]
  • 135.Muramatsu Y, Fukushima K, Iino K, et al. Urocortin and corticotropin-releasing factor receptor expression in the human colonic mucosa. Peptides. 2000;21:1799–1809. doi: 10.1016/S0196-9781(00)00335-1. [DOI] [PubMed] [Google Scholar]
  • 136.Wallon C, Söderholm JD. Corticotropin-releasing hormone and mast cells in the regulation of mucosal barrier function in the human colon. Ann N Y Acad Sci. 2009;1165:206–210. doi: 10.1111/j.1749-6632.2009.04030.x. [DOI] [PubMed] [Google Scholar]
  • 137.Million M, Wang L, Stenzel-Poore MP, et al. Enhanced pelvic responses to stressors in female CRF-overexpressing mice. Am J Physiol Regul Integr Comp Physiol. 2006;292:R1429–R1438. doi: 10.1152/ajpregu.00626.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Smith F, Clark JE, Overman BL, et al. Early weaning stress impairs development of mucosal barrier function in the porcine intestine. Am J Physiol Gastrointest Liver Physiol. 2010;298:G352–G363. doi: 10.1152/ajpgi.00081.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Gué M, Del Rio-Lacheze C, Eutamene H, Théodorou V, Fioramonti J, Buéno L. Stress-induced visceral hypersensitivity to rectal distension in rats: role of CRF and mast cells. Neurogastroenterol Motil. 1997;9:271–279. doi: 10.1046/j.1365-2982.1997.d01-63.x. [DOI] [PubMed] [Google Scholar]
  • 140.Greenwood-Van Meerveld B, Johnson AC, Cochrane S, Schulkin J, Myers DA. Corticotropin-releasing factor 1 receptor-mediated mechanisms inhibit colonic hypersensitivity in rats. Neurogastroenterol Motil. 2005;17:415–422. doi: 10.1111/j.1365-2982.2005.00648.x. [DOI] [PubMed] [Google Scholar]
  • 141.Million M, Grigoriadis DE, Sullivan S, et al. A novel water-soluble selective CRF1 receptor antagonist, NBI 35965, blunts stress-induced visceral hyperalgesia and colonic motor function in rats. Brain Res. 2003;985:32–42. doi: 10.1016/S0006-8993(03)03027-0. [DOI] [PubMed] [Google Scholar]
  • 142.Schwetz I, Bradesi S, McRoberts JA, et al. Delayed stress-induced colonic hypoersensitivity in male Wistar rats: role of neurokinin-1 and corticotropin releasing factor-1 receptors. Am J Physiol Gastrointest Liver Physiol. 2004;286:G683–G691. doi: 10.1152/ajpgi.00358.2003. [DOI] [PubMed] [Google Scholar]
  • 143.Schwetz I, McRoberts JA, Coutinho SV, et al. Corticotropin-releasing factor receptor 1 mediates acute and delayed stress-induced visceral hyperalgesia in maternally separated Long-Evans rats. Am J Physiol Gastrointest Liver Physiol. 2005;289:G704–G712. doi: 10.1152/ajpgi.00498.2004. [DOI] [PubMed] [Google Scholar]
  • 144.Larauche M, Bradesi S, Million M, et al. Corticotropin-releasing factor type 1 receptors mediate the visceral hyperalgesia induced by repeated psychological stress in rats. Am J Physiol Gastrointest Liver Physiol. 2008;294:G1033–G1040. doi: 10.1152/ajpgi.00507.2007. [DOI] [PubMed] [Google Scholar]
  • 145.Saito-Nakaya K, Hasegawa R, Nagura Y, Ito H, Fukudo S. Corticotropin-releasing hormone receptor 1 antagonist blocks colonic hypersensitivity induced by a combination of inflammation and repetitive colorectal distension. Neurogastroenterol Motil. 2008;20:1147–1156. doi: 10.1111/j.1365-2982.2008.01151.x. [DOI] [PubMed] [Google Scholar]
  • 146.Million M, Maillot C, Adelson DW, et al. Peripheral injection of sauvagine prevents repeated colorectal distention-induced visceral pain in female rats. Peptides. 2005;26:1188–1195. doi: 10.1016/j.peptides.2005.02.004. [DOI] [PubMed] [Google Scholar]
  • 147.Buckley MM, O’Halloran KD, Rae MG, Dinan TG, o’malley D. Modulation of enteric neurons by interleukin-6 and corticotropin-releasing factor contributes to visceral hypersensitivity and altered colonic motility in a rat model of irritable bowel syndrome. J Physiol. 2014;592(Pt 23):5235–5250. doi: 10.1113/jphysiol.2014.279968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Smith C, Nordstrom E, Sengupta JN, Miranda A. Neonatal gastric suctioning results in chronic visceral and somatic hyperalgesia: role of corticotropin releasing factor. Neurogastroenterol Motil. 2007;19:692–699. doi: 10.1111/j.1365-2982.2007.00949.x. [DOI] [PubMed] [Google Scholar]
  • 149.Trimble N, Johnson AC, Foster A, Greenwood-Van Meerveld B. Corticotropin-releasing factor receptor 1-deficient mice show decreased anxiety and colonic sensitivity. Neurogastroenterol Motil. 2007;19:754–760. doi: 10.1111/j.1365-2982.2007.00951.x. [DOI] [PubMed] [Google Scholar]
  • 150.Bradesi S, Martinez V, Lao L, Larsson H, Mayer EA. Involvement of vasopressin 3 receptors in chronic psychological stress-induced visceral hyperalgesia in rats. Am J Physiol Gastrointest Liver Physiol. 2009;296:G302–G309. doi: 10.1152/ajpgi.90557.2008. [DOI] [PubMed] [Google Scholar]
  • 151.Jia FY, Li XL, Li TN, Wu J, Xie BY, Lin L. Role of nesfatin-1 in a rat model of visceral hypersensitivity. World J Gastroenterol. 2013;19:3487–3493. doi: 10.3748/wjg.v19.i22.3487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Tran L, Schulkin J, Greenwood-Van Meerveld B. Importance of CRF receptor-mediated mechanisms of the bed nucleus of the stria terminalis in the processing of anxiety and pain. Neuropsychopharmacology. 2014;39:2633–2645. doi: 10.1038/npp.2014.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Johnson AC, Tran L, Schulkin J, Greenwood-Van Meerveld B. Importance of stress receptor-mediated mechanisms in the amygdala on visceral pain perception in an intrinsically anxious rat. Neurogastroenterol Motil. 2012;24:479–86. e219. doi: 10.1111/j.1365-2982.2012.01899.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Su J, Tanaka Y, Muratsubaki T, Kano M, Kanazawa M, Fukudo S. Injection of corticotropin-releasing hormone into the amygdala aggravates visceral nociception and induces noradrenaline release in rats. Neurogastroenterol Motil. doi: 10.1111/nmo.12462.. Published Online First: 31 Oct 2014. [DOI] [PubMed] [Google Scholar]
  • 155.Van Bockstaele EJ, Colago EE, Valentino RJ. Amygdaloid corticotropin-releasing factor targets locus coeruleus dendrites: substrate for the co-ordination of emotional and cognitive limbs of the stress response. J Neuroendocrinol. 1998;10:743–757. doi: 10.1046/j.1365-2826.1998.00254.x. [DOI] [PubMed] [Google Scholar]
  • 156.Gray TS. Amygdaloid CRF pathways. Role in autonomic, neuroendocrine, and behavioral responses to stress. Ann N Y Acad Sci. 1993;697:53–60. doi: 10.1111/j.1749-6632.1993.tb49922.x. [DOI] [PubMed] [Google Scholar]
  • 157.Kim SH, Han JE, Hwang S, Oh DH. The expression of corticotropin-releasing factor in the central nucleus of the amygdala, induced by colorectal distension, is attenuated by general anesthesia. J Korean Med Sci. 2010;25:1646–1651. doi: 10.3346/jkms.2010.25.11.1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Ji G, Fu Y, Ruppert KA, Neugebauer V. Pain-related anxiety-like behavior requires CRF1 receptors in the amygdala. Mol Pain. 2007;3:13. doi: 10.1186/1744-8069-3-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Rouwette T, Klemann K, Gaszner B, et al. Differential responses of corticotropin-releasing factor and urocortin 1 to acute pain stress in the rat brain. Neuroscience. 2011;183:15–24. doi: 10.1016/j.neuroscience.2011.03.054. [DOI] [PubMed] [Google Scholar]
  • 160.Greenwood-Van Meerveld B, Johnson AC, Schulkin J, Myers DA. Long-term expression of corticotropin-releasing factor (CRF) in the paraventricular nucleus of the hypothalamus in response to an acute colonic inflammation. Brain Res. 2006;1071:91–96. doi: 10.1016/j.brainres.2005.11.071. [DOI] [PubMed] [Google Scholar]
  • 161.Nishii H, Nomura M, Aono H, Fujimoto N, Matsumoto T. Up-regulation of galanin and corticotropin-releasing hormone mRNAs in the key hypothalamic and amygdaloid nuclei in a mouse model of visceral pain. Regul Pept. 2007;141:105–112. doi: 10.1016/j.regpep.2006.12.022. [DOI] [PubMed] [Google Scholar]
  • 162.Kosoyan HP, Grigoriadis DE, Taché Y. The CRF1 antagonist, NBI-35965 abolished the activation of locus coeruleus by colorectal distention and intracisternal CRF in rats. Brain Res. 2005;1056:85–96. doi: 10.1016/j.brainres.2005.07.010. [DOI] [PubMed] [Google Scholar]
  • 163.Rouzade-Dominguez ML, Curtis AL, Valentino RJ. Role of Barrington’s nucleus in the activation of rat locus coeruleus neurons by colonic distension. Brain Res. 2001;917:206–218. doi: 10.1016/S0006-8993(01)02917-1. [DOI] [PubMed] [Google Scholar]
  • 164.Lechner SM, Curtis AL, Brons R, Valentino RJ. Locus coeruleus activation by colon distention: role of corticotropin-releasing factor and excitatory amino acids. Brain Res. 1997;756:114–124. doi: 10.1016/S0006-8993(97)00116-9. [DOI] [PubMed] [Google Scholar]
  • 165.Kravets JL, Reyes BA, Unterwald EM, Van Bockstaele EJ. Direct targeting of peptidergic amygdalar neurons by noradrenergic afferents: linking stress-integrative circuitry. Brain Struct Funct. doi: 10.1007/s00429-013-0674-8.. Published Online First: 23 Nov 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Naliboff BD, Munakata J, Fullerton S, et al. Evidence for two distinct perceptual alterations in irritable bowel syndrome. Gut. 1997;41:505–512. doi: 10.1136/gut.41.4.505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Berridge CW. Noradrenergic modulation of arousal. Brain Res Rev. 2008;58:1–17. doi: 10.1016/j.brainresrev.2007.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Barreau F, Cartier C, Leveque M, et al. Pathways involved in gut mucosal barrier dysfunction induced in adult rats by maternal deprivation: corticotrophin-releasing factor and nerve growth factor interplay. J Physiol. 2007;580:347–356. doi: 10.1113/jphysiol.2006.120907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Eutamene H, Theodorou V, Fioramonti J, Bueno L. Acute stress modulates the histamine content of mast cells in the gastrointestinal tract through interleukin-1 and corticotropin-releasing factor release in rats. J Physiol. 2003;553(Pt 3):959–966. doi: 10.1113/jphysiol.2003.052274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Larauche M. Novel insights in the role of peripheral corticotropin-releasing factor and mast cells in stress-induced visceral hypersensitivity. Neurogastroenterol Motil. 2012;24:201–205. doi: 10.1111/j.1365-2982.2011.01867.x. [DOI] [PubMed] [Google Scholar]
  • 171.Sengupta JN. Visceral pain: the neurophysiological mechanism. Handb Exp Pharmacol. 2009:31–74. doi: 10.1007/978-3-540-79090-7_2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Cervero F, Laird JM. Understanding the signaling and transmission of visceral nociceptive events. J Neurobiol. 2004;61:45–54. doi: 10.1002/neu.20084. [DOI] [PubMed] [Google Scholar]
  • 173.Teitelbaum AA, Gareau MG, Jury J, Yang PC, Perdue MH. Chronic peripheral administration of corticotropin-releasing factor causes colonic barrier dysfunction similar to psychological stress. Am J Physiol Gastrointest Liver Physiol. 2008;295:G452–G459. doi: 10.1152/ajpgi.90210.2008. [DOI] [PubMed] [Google Scholar]
  • 174.Soderholm JD, Perdue MH. II. Stress and intestinal barrier function. Am J Physiol Gastrointest Liver Physiol. 2001;280:G7–G13. doi: 10.1152/ajpgi.2001.280.1.G7. [DOI] [PubMed] [Google Scholar]
  • 175.Ait-Belgnaoui A, Bradesi S, Fioramonti J, Theodorou V, Bueno L. Acute stress-induced hypersensitivity to colonic distension depends upon increase in paracellular permeability: role of myosin light chain kinase. Pain. 2005;113:141–147. doi: 10.1016/j.pain.2004.10.002. [DOI] [PubMed] [Google Scholar]
  • 176.Million M, Wang L, Wang Y, et al. CRF2 receptor activation prevents colorectal distension-induced visceral pain and spinal ERK1/2 phosphorylation in rats. Gut. 2006;55:172–181. doi: 10.1136/gut.2004.051391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.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;42:845–849. doi: 10.1136/gut.42.6.845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Lembo T, Plourde V, Shui Z, et al. 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]
  • 179.Nozu T, Kudaira M. Corticotropin-releasing factor induces rectal hypersensitivity after repetitive painful rectal distention in healthy humans. J Gastroenterol. 2006;41:740–744. doi: 10.1007/s00535-006-1848-4. [DOI] [PubMed] [Google Scholar]
  • 180.Sagami Y, Shimada Y, Tayama J, et al. Effect of a corticotropin releasing hormone receptor antagonist on colonic sensory and motor function in patients with irritable bowel syndrome. Gut. 2004;53:958–964. doi: 10.1136/gut.2003.018911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.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;19:471–483. doi: 10.1111/j.1365-2982.2007.00903.x. [DOI] [PubMed] [Google Scholar]
  • 182.Yuan PQ, Wu SV, Elliott J, et al. Expression of corticotropin releasing factor receptor type 1 (CRF1) in the human gastrointestinal tract and upregulation in the colonic mucosa in patients with ulcerative colitis. Peptides. 2012;38:62–69. doi: 10.1016/j.peptides.2012.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Wallon C, Persborn M, Jönsson M, et al. Eosinophils express muscarinic receptors and corticotropin-releasing factor to disrupt the mucosal barrier in ulcerative colitis. Gastroenterology. 2011;140:1597–1607. doi: 10.1053/j.gastro.2011.01.042. [DOI] [PubMed] [Google Scholar]
  • 184.Wallon C, Yang PC, Keita AV, et al. Corticotropin-releasing hormone (CRH) regulates macromolecular permeability via mast cells in normal human colonic biopsies in vitro. Gut. 2008;57:50–58. doi: 10.1136/gut.2006.117549. [DOI] [PubMed] [Google Scholar]
  • 185.Chadwick VS, Chen W, Shu D, et al. Activation of the mucosal immune system in irritable bowel syndrome. Gastroenterology. 2002;122:1778–1783. doi: 10.1053/gast.2002.33579. [DOI] [PubMed] [Google Scholar]
  • 186.Dinan TG, Quigley EM, Ahmed SM, et al. Hypothalamic-pituitary-gut axis dysregulation in irritable bowel syndrome: plasma cytokines as a potential biomarker? Gastroenterology. 2006;130:304–311. doi: 10.1053/j.gastro.2005.11.033. [DOI] [PubMed] [Google Scholar]
  • 187.Stasi C, Bellini M, Bassotti G, Blandizzi C, Milani S. Serotonin receptors and their role in the pathophysiology and therapy of irritable bowel syndrome. Tech Coloproctol. 2014;18:613–621. doi: 10.1007/s10151-013-1106-8. [DOI] [PubMed] [Google Scholar]
  • 188.Williams JP. Corticotropin-releasing factor 1 receptor antagonists: a patent review. Expert Opin Ther Pat. 2013;23:1057–1068. doi: 10.1517/13543776.2013.795545. [DOI] [PubMed] [Google Scholar]
  • 189.Chen C. Recent advances in small molecule antagonists of the corticotropin-releasing factor type-1 receptor-focus on pharmacology and pharmacokinetics. Curr Med Chem. 2006;13:1261–1282. doi: 10.2174/092986706776873014. [DOI] [PubMed] [Google Scholar]
  • 190.Tellew JE, Lanier M, Moorjani M, et al. Discovery of NBI-77860/GSK561679, a potent corticotropin- releasing factor (CRF1) receptor antagonist with improved pharmacokinetic properties. Bioorg Med Chem Lett. 2010;20:7259–7264. doi: 10.1016/j.bmcl.2010.10.095. [DOI] [PubMed] [Google Scholar]
  • 191.Zhuo X, Hartz RA, Bronson JJ, et al. Comparative biotransformation of pyrazinone-containing corticotropin-releasing factor receptor-1 antagonists: minimizing the reactive metabolite formation. Drug Metab Dispos. 2010;38:5–15. doi: 10.1124/dmd.109.028910. [DOI] [PubMed] [Google Scholar]
  • 192.Hodgetts KJ, Ge P, Yoon T, et al. Discovery of N-(1-ethylpropyl)[3-methoxy-5-(2-methoxy-4-trifluoromethoxyphenyl)-6-methylpyrazin-2-yl]amine 59 (NGD 98-2): an orally active corticotropin releasing factor-1 (CRF-1) receptor antagonist. J Med Chem. 2011;54:4187–4206. doi: 10.1021/jm200365y. [DOI] [PubMed] [Google Scholar]
  • 193.Ising M, Zimmermann US, Kunzel HE, et al. High-affinity CRF1 receptor antagonist NBI-34041: preclinical and clinical data suggest safety and efficacy in attenuating elevated stress response. Neuropsychopharmacology. 2007;32:1941–1949. doi: 10.1038/sj.npp.1301328. [DOI] [PubMed] [Google Scholar]
  • 194.Bailey JE, Papadopoulos A, Diaper A, et al. Preliminary evidence of anxiolytic effects of the CRF1 receptor antagonist R317573 in the 7.5% CO2 proof-of-concept experimental model of human anxiety. J Psychopharmacol. 2011 doi: 10.1177/0269881111400650. [DOI] [PubMed] [Google Scholar]
  • 195.Schmidt ME, Andrews RD, van der Ark P, et al. Dose-dependent effects of the CRF1 receptor antagonist R317573 on regional brain activity in healthy male subjects. Psychopharmacology (Berl) 2010;208:109–119. doi: 10.1007/s00213-009-1714-1. [DOI] [PubMed] [Google Scholar]
  • 196.Hubbard CS, Labus JS, Bueller J, et al. Corticotropin-releasing factor receptor 1 antagonist alters regional activation and effective connectivity in an emotional-arousal circuit during expectation of abdominal pain. J Neurosci. 2011;31:12491–12500. doi: 10.1523/JNEUROSCI.1860-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Ji G, Neugebauer V. Differential effects of CRF1 and CRF2 receptor antagonists on pain-related sensitization of neurons in the central nucleus of the amygdala. J Neurophysiol. 2007;97:3893–3904. doi: 10.1152/jn.00135.2007. [DOI] [PubMed] [Google Scholar]
  • 198.Neugebauer V, Li W, Bird GC, Han JS. The amygdala and persistent pain. Neuroscientist. 2004;10:221–234. doi: 10.1177/1073858403261077. [DOI] [PubMed] [Google Scholar]
  • 199.Bouin M, Plourde V, Boivin M, et al. Rectal distention testing in patients with irritable bowel syndrome: sensitivity, specificity, and predictive values of pain sensory thresholds. Gastroenterology. 2002;122:1771–1777. doi: 10.1053/gast.2002.33601. [DOI] [PubMed] [Google Scholar]
  • 200.North CS, Hong BA, Alpers DH. Relationship of functional gastrointestinal disorders and psychiatric disorders: implications for treatment. World J Gastroenterol. 2007;13:2020–2027. doi: 10.3748/wjg.v13.i14.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Sweetser S, Camilleri M, Linker Nord SJ, et al. Do corticotropin releasing factor-1 receptors influence colonic transit and bowel function in women with irritable bowel syndrome? Am J Physiol Gastrointest Liver Physiol. 2009;296:G1299–G1306. doi: 10.1152/ajpgi.00011.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Martinez V, Taché Y. CRF1 receptors as a therapeutic target for irritable bowel syndrome. Curr Pharm Des. 2006;12:4071–4088. doi: 10.2174/138161206778743637. [DOI] [PubMed] [Google Scholar]
  • 203.Mahajan S, Liao M, Barkan P, Takahashi K, Bhargava A. Urocortin 3 expression at baseline and during inflammation in the colon: corticotropin releasing factor receptors cross-talk. Peptides. 2014;54:58–66. doi: 10.1016/j.peptides.2014.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Fu Y, Neugebauer V. Differential mechanisms of CRF1 and CRF2 receptor functions in the amygdala in pain-related synaptic facilitation and behavior. J Neurosci. 2008;28:3861–3876. doi: 10.1523/JNEUROSCI.0227-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Zorrilla EP, Logrip ML, Koob GF. Corticotropin releasing factor: a key role in the neurobiology of addiction. Front Neuroendocrinol. 2014;35:234–244. doi: 10.1016/j.yfrne.2014.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Gravanis A, Margioris AN. The corticotropin-releasing factor (CRF) family of neuropeptides in inflammation: potential therapeutic applications. Curr Med Chem. 2005;12:1503–1512. doi: 10.2174/0929867054039008. [DOI] [PubMed] [Google Scholar]
  • 207.Holsboer F, Ising M. Central CRH system in depression and anxiety--evidence from clinical studies with CRH1 receptor antagonists. Eur J Pharmacol. 2008;583:350–357. doi: 10.1016/j.ejphar.2007.12.032. [DOI] [PubMed] [Google Scholar]
  • 208.Paez-Pereda M, Hausch F, Holsboer F. Corticotropin releasing factor receptor antagonists for major depressive disorder. Expert Opin Investig Drugs. 2011;20:519–535. doi: 10.1517/13543784.2011.565330. [DOI] [PubMed] [Google Scholar]
  • 209.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;8:258–273. doi: 10.2174/156652408784533788. [DOI] [PubMed] [Google Scholar]
  • 210.Stengel A, Taché Y. Neuroendocrine control of the gut during stress: corticotropin-releasing factor signaling pathways in the spotlight. Annu Rev Physiol. 2009;71:219–239. doi: 10.1146/annurev.physiol.010908.163221. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Neurogastroenterology and Motility are provided here courtesy of The Korean Society of Neurogastroenterology and Motility

RESOURCES