Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 299
Filter
Add more filters










Publication year range
1.
Neurogastroenterol Motil ; 27(1): 1-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25557223

ABSTRACT

Corticotropin-releasing factor (CRF)-CRF1 receptor in the brain plays a key role in stress-related alterations of behavior including anxiety/depression, and autonomic and visceral functions. In particular, CRF1 signaling mediates hypersensitivity to colorectal distension (CRD) in various models (early life adverse events, repeated psychological stress, chronic high anxiety, postcolonic inflammation, or repeated nociceptive CRD). So far, knowledge of brain sites involved is limited. A recent article demonstrates in rats that CRF microinjected into the central amygdala (CeA) induces a hyperalgesic response to CRD and enhances the noradrenaline and dopamine levels at this site. The visceral and noradrenaline, unlike dopamine, responses were blocked by a CRF1 antagonist injected into the CeA. Here, we review the emerging role that CRF-CRF1 signaling plays in the CeA to induce visceral hypersensitivity. In the somatic pain field, CRF in the CeA was shown to induce pain sensitization. This is mediated by the activation of postsynaptic CRF1 receptors and protein kinase A signaling that increases N-methyl-d-aspartate receptor neurotransmission. In addition, the activation of tetraethylamonium-sensitive ion channels such as Kv3 accelerates repolarization and firing rate. Whether facilitation of pain transmission underlies CRF action in the CeA-induced visceral hypersensitivity will need to be delineated. CRF1 signaling in the CeA is also an important component of the neuronal circuitry inducing anxiety-like behavior and positioned at the interphase of the reciprocal relationship between pain and affective state. The hyperactivity of this system may represent the neuroanatomical and biochemical substrate contributing to the coexpression of hypersensitivity to CRD and mood disorders in subsets of irritable bowel syndrome patients.


Subject(s)
Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Hyperalgesia/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Anxiety/metabolism , Colon/metabolism , Dilatation, Pathologic , Humans , Mice , Neurons/metabolism , Pain , Rats , Rectum/metabolism , Signal Transduction , Stress, Psychological/metabolism
2.
Neurogastroenterol Motil ; 26(12): 1771-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25327342

ABSTRACT

BACKGROUND: Constipation and L-dopa-induced gastric dysmotility are common gastrointestinal (GI) symptoms in Parkinson's disease (PD). We investigated the novel ghrelin agonist, HM01 influence on GI motor dysfunctions in 6-hydroxydopamine (6-OHDA) rats. METHODS: HM01 pharmacological profiles were determined in vitro and in vivo in rats. We assessed changes in fecal output and water content, and gastric emptying (GE) in 6-OHDA rats treated with orogastric (og) HM01 and L-dopa/carbidopa (LD/CD, 20/2 mg/kg). Fos immunoreactivity (ir) cells in specific brain and lumbosacral spinal cord were quantified. KEY RESULTS: HM01 displayed a high binding affinity to ghrelin receptor (Ki: 1.42 ± 0.36 nM), 4.3 ± 1.0 h half-life and high brain/plasma ratio. 6-OHDA rats had reduced daily fecal output (22%) and water intake (23%) compared to controls. HM01 (3 and 10 mg/kg) similarly reversed the decreased 4-h fecal weight and water content in 6-OHDA rats. Basal GE was not modified in 6-OHDA rats, however, LD/CD (once or daily for 8 days) delayed GE in 6-OHDA and control rats that was prevented by HM01 (3 mg/kg acute or daily before LD/CD). HM01 increased Fos-ir cell number in the area postrema, arcuate nucleus, nucleus tractus solitarius, and lumbosacral intermediolateral column of 6-OHDA rats where 6-OHDA had a lowering effect compared to controls. CONCLUSIONS & INFERENCES: 6-OHDA rats display constipation- and adipsia-like features of PD and L-dopa-inhibited GE. The new orally active ghrelin agonist, HM01 crosses the blood-brain barrier and alleviates these alterations suggesting a potential benefit for PD with GI disorders.


Subject(s)
Brain/drug effects , Constipation/prevention & control , Gastrointestinal Transit/drug effects , Ghrelin/agonists , Parkinsonian Disorders/complications , Animals , Antiparkinson Agents/adverse effects , Constipation/etiology , Immunohistochemistry , Levodopa/adverse effects , Male , Oxidopamine/toxicity , Rats , Rats, Sprague-Dawley
3.
Horm Metab Res ; 45(13): 975-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24048879

ABSTRACT

Hunger and satiety are regulated in a complex fashion by a few food intake stimulatory (orexigenic) and a multitude of inhibitory (anorexigenic) factors produced in the periphery (mainly in the gastrointestinal tract) or directly in the brain. Within the brain, the hypothalamus plays a pivotal role as a production site of food intake regulatory factors. Importantly, this site integrates peripheral and central signaling factors to orchestrate food intake and in the long term body weight. Our knowledge on these regulatory pathways is not static but rather rapidly changing as new factors as well as up- and downstream signaling pathways of already known transmitters are uncovered. Hypothalamic nucleobindin2 (NUCB2), the precursor of nesfatin-1, was first described in 2006 and nesfatin-1 found to be a novel anorexigenic modulator of food intake and body weight. The initial report stimulated several groups to investigate the biological actions of nesfatin-1 and subsequent studies delineated the underlying brain mechanisms involved in its food reducing effect. Of interest was the demonstration that NUCB2 also exerts its anorexigenic action in the paraventricular nucleus of the hypothalamus and is regulated at this site by changes in metabolic status with a diurnal rhythm inversely related to that of feeding in rats. The present review describes the current state-of-knowledge on central nesfatin-1's effects on food intake and body weight and highlights important missing links regarding cellular signaling mechanisms involved in nesfatin-1's action.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Energy Metabolism/physiology , Homeostasis/physiology , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Animals , Body Weight/physiology , Eating/physiology , Humans , Nucleobindins , Rats
4.
Obes Rev ; 14(11): 859-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23980879

ABSTRACT

Nesfatin-1 was discovered in 2006 and introduced as a potential novel anorexigenic modulator of food intake and body weight. The past years have witnessed increasing evidence establishing nesfatin-1 as a potent physiological inhibitor of food intake and body weight and unravelled nesfatin-1's interaction with other brain transmitters to exert its food consumption inhibitory effect. As observed for other anorexigenic brain neuropeptides, nesfatin-1 is also likely to exert additional, if not pleiotropic, actions in the brain and periphery. Recent studies established the prominent expression of the nesfatin-1 precursor, nucleobindin2 (NUCB2), in the stomach and pancreas, where nesfatin-1 influences endocrine secretion. This review will highlight the current experimental state-of-knowledge on the effects of NUCB2/nesfatin-1 on food intake, body weight and glucose homeostasis. Potential implications in human obesity will be discussed in relation to the evidence of changes in circulating levels of NUCB2/nesfatin-1 in disease states, the occurrence of genetic NUCB2 polymorphisms and--in contrast to several other hormones--the independence of leptin signalling known to be blunted under conditions of chronically increased body weight.


Subject(s)
Appetite Regulation , Body Weight , Calcium-Binding Proteins/physiology , DNA-Binding Proteins/physiology , Nerve Tissue Proteins/physiology , Obesity/metabolism , Animals , Appetite Regulation/genetics , Body Weight/genetics , Calcium-Binding Proteins/genetics , DNA-Binding Proteins/genetics , Forecasting/methods , Humans , Leptin/genetics , Leptin/physiology , Nerve Tissue Proteins/genetics , Nucleobindins , Obesity/etiology , Obesity/genetics , Signal Transduction/genetics
5.
Neurogastroenterol Motil ; 24(9): e425-36, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22779732

ABSTRACT

BACKGROUND: Prevalent non-motor symptoms of Parkinson's disease (PD) include gastrointestinal motor impairments and advanced stage PD displays pathological aggregates of α-synuclein in colonic enteric neurons. We previously showed that 12 months old mice overexpressing human wild type (WT) α-synuclein under the Thy1 promoter (Thy1-aSyn) displayed colonic motor dysfunction. We investigated functional gut alterations at earlier ages and histological correlates. METHODS: Defecation, gastric emptying (GE), and immunostaining for α-synuclein, peripheral choline acetyltransferase (pChAT), tyrosine hydroxylase (TH), neuronal nitric oxide synthase (nNOS), and vasoactive intestinal peptide (VIP) in distal colon myenteric plexuses were assessed in male Thy1-aSyn compared to littermate WT mice. KEY RESULTS: Thy1-aSyn mice aged 2.5-3 or 7-8 months old had 81% and 55% reduction in fecal pellet output, respectively, in the first 15 min of exposure to a novel environment. The reduction remained significant in the older group for 2-h, and subsequent refeeding resulted also in a 60% and 69% reduction of defecation in the first hour, respectively. Thy1-aSyn mice (8-10 months) displayed increased α-synuclein in the myenteric plexuses with abundant varicose terminals surrounding pChAT-immunoreactive (ir) neurons, and only a few, nNOS-ir neurons. There were no conspicuous changes in pChAT- and nNOS-ir neurons, or TH- and VIP-ir nerve fibers. Thy1-aSyn mice aged 4-18 months had normal GE. CONCLUSIONS & INFERENCES: The occurrence of over-production of pre-synaptic α-synuclein in colonic myenteric ganglia several months before the loss of striatal dopamine may provide an anatomical basis for interference with cholinergic neuronal activation, causing an early impairment in defecation to stimuli.


Subject(s)
Colon/physiology , Defecation/physiology , Ganglia, Autonomic/metabolism , Myenteric Plexus/metabolism , alpha-Synuclein/metabolism , Animals , Choline O-Acetyltransferase/metabolism , Colon/innervation , Colon/physiopathology , Ganglia, Autonomic/physiopathology , Gastric Emptying/physiology , Male , Mice , Mice, Transgenic , Myenteric Plexus/physiopathology , Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Tyrosine 3-Monooxygenase/metabolism
6.
Neurogastroenterol Motil ; 24(11): 1031-e547, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22776034

ABSTRACT

BACKGROUND: Chronic psychological stress-induced alterations in visceral sensitivity have been predominantly assessed in male rodents. We investigated the effect of acute and repeated water avoidance stress (WAS) on the visceromotor response (VMR) to colorectal distension (CRD) and the role of opioids in male and cycling female Wistar rats using a novel non-invasive manometric technique. METHODS: After a baseline VMR (1st CRD, day 0), rats were exposed to WAS (1 h day(-1) ) either once or for four consecutive days, without injection or with naloxone (1 mg kg(-1) ) or saline injected subcutaneously before each WAS session. KEY RESULTS: The VMR to CRD recorded on day 1 or 4 immediately after the last WAS was reduced in both females and males. The visceral analgesia was mainly naloxone-dependent in females, but naloxone-independent in males. In non-injected animals, on days 2 and 5, VMR was not significantly different from baseline in males whereas females exhibited a significant VMR increase at 60 mmHg on day 5. Basal CRD and CRD on days 1, 2, and 5 in both sexes without WAS induced similar VMR. CONCLUSIONS & INFERENCES: When monitored non-invasively, psychological stress induces an immediate poststress visceral analgesia mediated by an opiate signaling system in females while naloxone-independent in males, and hyperalgesia at 24 h after repeated stress only in females. These data highlight the importance of sex-specific interventions to modulate visceral pain response to stress.


Subject(s)
Hyperalgesia/psychology , Receptors, Opioid/physiology , Stress, Psychological/complications , Visceral Pain/psychology , Analgesics, Opioid/metabolism , Animals , Colon/drug effects , Colon/metabolism , Colon/physiopathology , Female , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Manometry , Myoelectric Complex, Migrating/physiology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Rats , Rats, Wistar , Rectum/drug effects , Rectum/metabolism , Rectum/physiopathology , Sex Characteristics , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Visceral Pain/metabolism , Visceral Pain/physiopathology
7.
Neurogastroenterol Motil ; 24(5): e235-45, 2012 May.
Article in English | MEDLINE | ID: mdl-22443313

ABSTRACT

BACKGROUND: Levodopa (L-dopa) is the most commonly used treatment for alleviating symptoms of Parkinson's disease. However, L-dopa delays gastric emptying, which dampens its absorption. We investigated whether ghrelin prevents L-dopa action on gastric emptying and enhances circulating L-dopa in rats. METHODS: Gastric emptying of non-nutrient methylcellulose/phenol red viscous solution was determined in fasted rats treated with orogastric or intraperitoneal (i.p.) L-dopa, or intravenous (i.v.) ghrelin 10 min before orogastric L-dopa. Plasma L-dopa and dopamine levels were determined by high pressure liquid chromatography. Plasma acyl ghrelin levels were assessed by radioimmunoassay. Fos expression in the brain was immunostained after i.v. ghrelin (30 µg kg(-1)) 10 min before i.p. L-dopa. KEY RESULTS: Levodopa (5 and 15 mg kg(-1)) decreased significantly gastric emptying by 32% and 62%, respectively, when administered orally, and by 91% and 83% when injected i.p. Ghrelin (30 or 100 µg kg(-1), i.v.) completely prevented L-dopa's (15 mg kg(-1), orogastrically) inhibitory action on gastric emptying and enhanced plasma L-dopa and dopamine levels compared with vehicle 15 min after orogastric L-dopa. Levodopa (5 mg kg(-1)) did not modify plasma acyl ghrelin levels at 30 min, 1, and 2 h after i.v. injection. Levodopa (15 mg kg(-1), i.p.) induced Fos in brain autonomic centers, which was not modified by i.v. ghrelin. CONCLUSIONS & INFERENCES: Ghrelin counteracts L-dopa-induced delayed gastric emptying but not Fos induction in the brain and enhances circulating L-dopa levels. Potential therapeutic benefits of ghrelin agonists in Parkinson's disease patients treated with L-dopa remain to be investigated.


Subject(s)
Dopamine Agents/pharmacology , Gastric Emptying/drug effects , Ghrelin/pharmacology , Levodopa/antagonists & inhibitors , Levodopa/pharmacology , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/metabolism , Catheterization , Dopamine/blood , Dopamine Agents/blood , Fasting/metabolism , Gene Expression/drug effects , Genes, fos , Immunohistochemistry , Injections, Intraperitoneal , Intubation, Gastrointestinal , Levodopa/blood , Male , Radioimmunoassay , Rats , Rats, Sprague-Dawley
8.
Curr Med Chem ; 19(1): 35-42, 2012.
Article in English | MEDLINE | ID: mdl-22300074

ABSTRACT

Earlier experimental studies indicated that the integrity of vagal pathway was required to confer gastric protection against damaging agents. Several peptides located in the brainstem initially identified to influence vagal outflow to the stomach, as assessed by electrophysiological approach or by vagal dependent alterations of gastric secretory and motor function, were investigated for their influence in the vagal regulation of the resistance of the gastric mucosa to injury. Thyrotropin releasing hormone (TRH), or its stable TRH analog, RX-77368, injected at low doses into the cisterna magna or the dorsal motor nucleus (DMN) was the first peptide reported to protect the gastric mucosa against ethanol injury through stimulation of vagal cholinergic pathways, inducing the release of gastric prostaglandins/nitric oxide (NO) and the recruitment of efferent function of capsaicin sensitive afferent fibers containing calcitonin-gene related peptide (CGRP). Activation of endogenous TRH-TRH1 receptor signaling located in the brainstem plays a role in adaptive gastric protection against damaging agents. Since then, an expanding number of peptides, namely peptide YY, CGRP, adrenomedullin, amylin, glugacon-like peptide, opioid peptides acting on µ, δ1 or δ2 receptors, nocicpetin, nocistatin, ghrelin, leptin and TLQP-21, a peptide derived from VGF prohormone, have been reported to act in the brainstem to afford gastric protection against ethanol injury largely through similar peripheral effectors mechanisms than TRH. Therefore gastric prostaglandins and CGRP/NO pathways represent a common final mechanism through which brain peptides confer vagally mediated gastroprotection against injury. A better understanding of brain circuitries through which these peptides are released will provide new strategies to recruit integrated and multifaceted gastroprotective mechanisms.


Subject(s)
Gastric Mucosa/physiology , Neuropeptides/physiology , Stomach Diseases/prevention & control , Vagus Nerve/physiology , Animals , Brain Stem/physiology , Capsaicin , Gastric Mucosa/innervation , Neurons, Afferent/physiology , Nitric Oxide/physiology , Prostaglandins/physiology
9.
Neurogastroenterol Motil ; 23(7): e294-308, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21569179

ABSTRACT

BACKGROUND: Activation of brain somatostatin receptors (sst(1-5) ) with the stable pan-sst(1-5) somatostatin agonist, ODT8-SST blocks acute stress and central corticotropin-releasing factor (CRF)-mediated activation of endocrine and adrenal sympathetic responses. Brain CRF signaling is involved in delaying gastric emptying (GE) immediately post surgery. We investigated whether activation of brain sst signaling pathways modulates surgical stress-induced inhibition of gastric emptying and food intake. METHODS: Fasted rats were injected intracisternally (i.c.) with somatostatin agonists and underwent laparotomy and 1-min cecal palpation. Gastric emptying of a non-nutrient solution and circulating acyl and desacyl ghrelin levels were assessed 50min post surgery. Food intake was monitored for 24 h. KEY RESULTS: The abdominal surgery-induced inhibition of GE (65%), food intake (73% at 2h) and plasma acyl ghrelin levels (67%) was completely prevented by ODT8-SST (1µg per rat, i.c.). The selective sst(5) agonist, BIM-23052 prevented surgery-induced delayed GE, whereas selective sst(1) , sst(2) , or sst(4) agonists had no effect. However, the selective sst(2) agonist, S-346-011 (1µg per rat, i.c.) counteracted the abdominal surgery-induced inhibition of acyl ghrelin and food intake but not the delayed GE. The ghrelin receptor antagonist, [D-Lys(3) ]-GHRP-6 (0.93mg kg(-1) , intraperitoneal, i.p.) blocked i.p. ghrelin-induced increased GE, while not influencing i.c. ODT8-SST-induced prevention of delayed GE and reduced food intake after surgery. CONCLUSIONS & INFERENCES: ODT8-SST acts in the brain to prevent surgery-induced delayed GE likely via activating sst(5) . ODT8-SST and the sst(2) agonist prevent the abdominal surgery-induced decrease in food intake and plasma acyl ghrelin indicating dissociation between brain somatostatin signaling involved in preventing surgery-induced suppression of GE and feeding response.


Subject(s)
Abdomen/surgery , Eating/drug effects , Gastric Emptying/drug effects , Ghrelin/blood , Peptide Fragments/pharmacology , Somatostatin/analogs & derivatives , Somatostatin/agonists , Animals , Brain/drug effects , Brain/physiology , Corticotropin-Releasing Hormone/physiology , Eating/physiology , Gastric Emptying/physiology , Injections, Intraventricular , Male , Models, Animal , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Somatostatin/administration & dosage , Somatostatin/pharmacology , Somatostatin-28/administration & dosage , Somatostatin-28/pharmacology , Stress, Psychological/physiopathology
10.
Neurogastroenterol Motil ; 23(6): e223-36, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21564422

ABSTRACT

BACKGROUND: Corticotropin-releasing factor (CRF) signaling induced by stress is well established to delay gastric emptying (GE) and stimulate colonic functions. The somatostatin receptor (sst(1-5) ) agonist, ODT8-SST acts in the brain to inhibit stress-induced adrenocorticotropic hormone and epinephrine secretion. We investigated whether ODT8-SST acts in the brain to influence stress-related alterations of gastric and colonic motor function and sst receptor subtype(s) involved. METHODS: Peptides were injected intracerebroventricularly (i.c.v.) under short isoflurane anesthesia and GE, fecal pellet output (FPO) and distal colonic motility monitored in conscious mice. KEY RESULTS: The stress of acute anesthesia/vehicle i.c.v. injection reduced GE by 67% and increased defecation by 99% compared to non-injected controls. Both responses were abolished by ODT8-SST (1µg= 0.75nmol) or sst(1) agonist (0.65-1.95nmol). The sst(1) agonist (1.95nmol) also prevented the abdominal surgery-induced delayed GE. Octreotide (sst(2) >sst(5) > sst(3) ) and the sst(2) or sst(4) agonists (1µg=0.78 or 0.70nmol, respectively) injected i.c.v. did not influence FPO while i.c.v. somatostatin-28 mimicked ODT8-SST's effect. The ODT8-SST-induced increased food intake was inhibited by i.c.v. sst(2) antagonist while the reduced FPO was unchanged. ODT8-SST i.c.v. reduced distal colonic motility in semi-restrained mice compared with vehicle and blocked water avoidance- and i.c.v. CRF (0.5µg=0.09nmol)-induced stimulated FPO while a similar colonic secretomotor response to i.p. 5-hydroxytryptophane (10mgkg(-1) =36.4µmol kg(-1) ) was unaltered. Conclusions & Inferences ODT8-SST counteracts stress/i.c.v. CRF-related stimulation of colonic motor function and delayed GE which can be reproduced mainly by activation of sst(1) receptors. These data opens new insight to brain somatostatinergic signaling pathways interfering with brain circuitries involved in gut motor responses to acute stress.


Subject(s)
Colon/physiology , Gastrointestinal Motility/physiology , Receptors, Somatostatin/metabolism , Stress, Physiological , Animals , Brain/drug effects , Brain/physiology , Corticotropin-Releasing Hormone/metabolism , Defecation/drug effects , Gastric Emptying/physiology , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Peptide Fragments/pharmacology , Receptors, Somatostatin/agonists , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
11.
Obes Rev ; 12(4): 261-71, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20546141

ABSTRACT

The protein nucleobindin 2 (NUCB2) or NEFA (DNA binding/EF-hand/acidic amino acid rich region) was identified over a decade ago and implicated in intracellular processes. New developments came with the report that post-translational processing of hypothalamic NUCB2 may result in nesfatin-1, nesfatin-2 and nesfatin-3 and convergent studies showing that nesfatin-1 and full length NUCB2 injected in the brain potently inhibit the dark phase food intake in rodents including leptin receptor deficient Zucker rats. Nesfatin-1 also reduces body weight gain, suggesting a role as a new anorexigenic factor and modulator of energy balance. In light of the obesity epidemic and its associated diseases, underlying new mechanisms regulating food intake may be promising targets in the drug treatment of obese patients particularly as the vast majority of them display reduced leptin sensitivity or leptin resistance while nesfatin-1's mechanism of action is leptin independent. Although much progress on the localization of NUCB2/nesfatin-1 in the brain and periphery as well as on the understanding of nesfatin-1's anorexic effect have been achieved during the past three years, several important mechanisms have yet to be unraveled such as the identification of the nesfatin-1 receptor and the regulation of NUCB2 processing and nesfatin-1 release.


Subject(s)
Appetite Regulation/drug effects , Energy Intake/drug effects , Hypothalamus/drug effects , Peptide Hormones/physiology , Satiety Response/drug effects , Animals , Appetite Regulation/physiology , Body Weight/drug effects , Body Weight/physiology , Calcium-Binding Proteins , DNA-Binding Proteins , Disease Models, Animal , Eating , Energy Intake/physiology , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , Leptin/metabolism , Nerve Tissue Proteins , Nucleobindins , Obesity/drug therapy , Obesity/metabolism , Peptide Hormones/metabolism , Rats , Satiety Response/physiology , Signal Transduction
12.
J Physiol Pharmacol ; 61(4): 399-407, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20814067

ABSTRACT

The consequences of selective activation of brain somatostatin receptor-2 (sst2) were assessed using the sst2 agonist, des-AA(1,4-6,11-13)-[DPhe(2),Aph7(Cbm),DTrp(8)]-Cbm-SST-Thr-NH2. Food intake (FI) was monitored in ad libitum fed rats chronically implanted with an intracerebroventricular (i.c.v.) cannula. The sst(2) agonist injected i.c.v. at 0.1 and 1 microg/rat dose-dependently increased light phase FI from 2 to 6 hours post injection (2.3+/-0.5 and 7.5+/-1.2 respectively vs. vehicle: 0.2+/-0.2 g/300 g bw, P<0.001). Peptide action was reversed by i.c.v. injection of the sst2 antagonist, des-AA(1,4-6,11-13)-[pNO(2)-Phe(2),DCys(3),Tyr(7),DAph(Cbm)8]-SST-2Nal-NH(2) and not reproduced by intraperitoneal injection (30 microg/rat). The sst(2) antagonist alone i.c.v. significantly decreased the cumulative 14-hours dark phase FI by 29.5%. Other behaviors, namely grooming, drinking and locomotor activity were also increased by the sst(2) agonist (1 microg/rat, i.c.v.) as monitored during the 2(nd) hour post injection while gastric emptying of solid food was unaltered. Rectal temperature rose 1 hour after the sst(2) agonist (1 microg/rat, i.c.v.) with a maximal response maintained from 1 to 4 hours post injection. These data show that selective activation of the brain sst(2) receptor induces a feeding response in the light phase not associated with changes in gastric emptying. The food intake reduction following sst(2) receptor blockade suggests a role of this receptor in the orexigenic drive during the dark phase.


Subject(s)
Body Temperature/physiology , Brain/metabolism , Eating/physiology , Grooming/physiology , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Animals , Body Temperature/drug effects , Brain/drug effects , Eating/drug effects , Gastric Emptying/drug effects , Gastric Emptying/physiology , Grooming/drug effects , Male , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
13.
Gut ; 59(6): 744-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20551459

ABSTRACT

OBJECTIVE: Chronic psychological stress is associated with development of intestinal barrier dysfunction and impairs host defence mechanisms. The intestinal epithelium, consisting of enterocytes, endocrine cells, goblet cells and Paneth cells, is an important component of this barrier. In the present study, the impact of maternal deprivation (MD) on secretory lineages of duodenal epithelium and the involvement of the peripheral corticotropin-releasing factor (CRF) pathway were investigated. METHODS: Rat pups were deprived of their dam for 3 h/day (days 5-20). Non-deprived pups served as controls. On days 8, 13, 20, 24, 34, 44 and 84, duodenal tissues were collected for quantitative real-time PCR and immunohistochemistry studies. RESULTS: MD induced a sustained decrease in the number of Paneth and goblet cells but hyperplasia of endocrine cells. These alterations were associated with a duodenal increase of CRF, urocortin 2 and CRF receptor subtype 2 (CRFR(2)) mRNA, whereas CRFR(1) expression was decreased. The effects of MD on intestinal epithelium were inhibited by the CRFR(1)/R(2) antagonist astressin injected daily before MD. Studies using specific receptor antagonists in rats subjected to MD revealed that CRFR(1) was involved in the hyperplasia of endocrine cells and CRFR(2) in the depletion of Paneth cells. Conversely, daily injection of CRF and of the CRFR(2) agonist urocortin 2 in control rats resulted in changes in epithelial differentiation similar to MD. CONCLUSIONS: The activation of CRFR(1) and CRFR(2) induced by MD markedly altered the quantitative distribution of secretory cells of the intestinal epithelium. These alterations, in particular the depletion of Paneth and goblet cells, may create conditions leading to the development of an epithelial barrier defect.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Duodenum/pathology , Intestinal Mucosa/pathology , Maternal Deprivation , Stress, Psychological/pathology , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/physiology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/pharmacology , Duodenum/drug effects , Duodenum/metabolism , Duodenum/physiopathology , Enteroendocrine Cells/pathology , Goblet Cells/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiopathology , Paneth Cells/pathology , Peptide Fragments/pharmacology , Rats , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Transcription Factors/metabolism , Urocortins/pharmacology
14.
Peptides ; 31(2): 322-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19944726

ABSTRACT

Little is known about CRF expression and regulation in the rat colon compared to the brain. We investigated CRF gene expression, cellular location, and regulation by endotoxin and corticosterone in the male rat colon at 6h after intraperitoneal (ip) injection. CRF mRNA level, detected by reverse transcription-polymerase chain reaction (RT-PCR) was 1.3-fold higher in the distal than proximal colon and 3.4-fold higher in the proximal colonic submucosa plus muscle layers than in mucosa. CRF immunoreactivity was located in the epithelia, lamina propria and crypts, and co-localized with tryptophan hydroxylase, a marker for enterochromaffin (EC) cells, and in enteric neurons. Lipopolysaccharide (LPS, 100 microg/kg, ip) increased defecation by 2.9-fold and upregulated CRF mRNA by 2.5-fold in the proximal and 1.1-fold in the distal colon while there was no change induced by corticosterone as monitored by quantitative PCR. LPS-induced increased CRF mRNA expression occurred in the submucosa plus muscle layers (1.5-fold) and the mucosa of proximal colon (0.9-fold). LPS increased significantly CRF immunoreactivity in the submucosal and myenteric plexuses of proximal and distal colon compared to saline groups. These results indicate that in rats, CRF is expressed in both proximal and distal colon and more prominently in enteric neurons of the submucosa plus muscle layers and subject to upregulation at the gene and protein levels by LPS through corticosteroid independent pathways. These data suggests that colonic CRF may be part of the local effector limb of the CRF(1) receptor mediated colonic alterations induced by acute stress.


Subject(s)
Colon/metabolism , Corticotropin-Releasing Hormone/metabolism , Endotoxins/pharmacology , Up-Regulation/drug effects , Animals , Colon/drug effects , Corticotropin-Releasing Hormone/genetics , Defecation/drug effects , Enterochromaffin Cells/cytology , Enterochromaffin Cells/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipopolysaccharides/pharmacology , Male , Mucous Membrane/cytology , Mucous Membrane/metabolism , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myenteric Plexus/cytology , Myenteric Plexus/drug effects , Myenteric Plexus/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/genetics
15.
Am J Physiol Gastrointest Liver Physiol ; 296(5): G992-G1002, 2009 May.
Article in English | MEDLINE | ID: mdl-19299579

ABSTRACT

There is a glaring lack of knowledge on mouse colonic motility in vivo, primarily due to unavailability of adequate recording methods. Using a noninvasive miniature catheter pressure transducer inserted into the distal colon, we assessed changes in colonic motility in conscious mice induced by various acute or chronic stressors and determined the neurotransmitters mediating these changes. Mice exposed to restraint stress (RS) for 60 min displayed distal colonic phasic contractions including high-amplitude giant migrating contractions (GMCs), which had peak amplitudes >25 mmHg and occurred at a rate of 15-25 h(-1) of which over 50% were aborally propagative. Responses during the first 20-min of RS were characterized by high-frequency and high-amplitude contractions that were correlated with defecation. RS-induced GMCs and fecal pellet output were blocked by atropine (0.5 mg/kg ip) or the corticotrophin releasing factor (CRF) receptor antagonist astressin-B (100 microg/kg ip). RS activated colonic myenteric neurons as shown by Fos immunoreactivity. In mice previously exposed to repeated RS (60 min/day, 14 days), or in transgenic mice that overexpress CRF, the duration of stimulation of phasic colonic contractions was significantly shorter (10 vs. 20 min). In contrast to RS, abdominal surgery abolished colonic contractions including GMCs. These findings provide the first evidence for the presence of frequent cholinergic-dependent GMCs in the distal colon of conscious mice and their modulation by acute and chronic stressors. Noninvasive colonic manometry opens new venues to investigate colonic motor function in genetically modified mice relevant to diseases that involve colonic motility alterations.


Subject(s)
Cholinergic Fibers/metabolism , Colon/innervation , Gastrointestinal Motility , Manometry/methods , Myenteric Plexus/physiopathology , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/physiopathology , Acute Disease , Animals , Atropine/pharmacology , Cholinergic Fibers/drug effects , Colon/drug effects , Colon/surgery , Colonic Diseases/etiology , Colonic Diseases/physiopathology , Corticotropin-Releasing Hormone/pharmacology , Defecation , Digestive System Surgical Procedures/adverse effects , Disease Models, Animal , Equipment Design , Gastrointestinal Motility/drug effects , Ileus/etiology , Ileus/physiopathology , Male , Manometry/instrumentation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Miniaturization , Muscarinic Antagonists/pharmacology , Myenteric Plexus/drug effects , Myenteric Plexus/metabolism , Nitrergic Neurons/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/genetics , Restraint, Physical/adverse effects , Stress, Psychological/etiology , Stress, Psychological/metabolism , Time Factors , Transducers, Pressure , Up-Regulation
16.
J Physiol Pharmacol ; 60 Suppl 7: 33-46, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20388944

ABSTRACT

It is well established that central corticotropin releasing factor (CRF) signaling mediates the gastrointestinal responses to stress. However, as shown in the brain, both CRF receptors and ligands are also widely expressed in the colon and the ileum of humans and rodents, and stress modulates their expression. Several functional studies documented that peripheral injection of CRF or urocortin stimulates colonic transit, motility, Fos expression in myenteric neurons, and defecation through activation of CRF(1) receptors, whereas it decreases ileal contractility via CRF(2) receptors. Additionally, intraperitoneal administration of CRF induces colonic mast cells degranulation via both CRF(1) and CRF(2) receptors and increases ion secretion and mucosal permeability to macromolecules, which can in turn promote intestinal inflammation and alter visceral sensitivity. Most peripheral CRF-induced alterations of colonic and ileal functions mimic effects which are observed after stress exposure, and CRF receptor antagonists given peripherally prevent stress-induced GI dysfunction. Furthermore, CRF peptides can reproduce secretomotor and mucosal alterations in vitro. Therefore, accumulated clinical and preclinical evidence supports in addition to the brain, a role for peripheral CRF signaling in mediating stress-induced effects on gastrointestinal sensorimotor, mucosal and immune functions, that may be components of underlying mechanisms involved in stress-related impact on inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS).


Subject(s)
Colon/physiology , Colon/physiopathology , Corticotropin-Releasing Hormone/physiology , Ileum/physiology , Ileum/physiopathology , Receptors, Corticotropin-Releasing Hormone/physiology , Stress, Physiological , Animals , Gene Expression Regulation , Humans , Intestinal Diseases/physiopathology , Ligands , Signal Transduction
17.
Neurogastroenterol Motil ; 20(4): 269-84, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18371009

ABSTRACT

Cyclic vomiting syndrome (CVS) was initially described in children but can occur in all age groups. Cyclic vomiting syndrome is increasingly recognized in adults. However, the lack of awareness of CVS in adults has led to small numbers of diagnosed patients and a paucity of published data on the causes, diagnosis and management of CVS in adults. This article is a state-of-knowledge overview on CVS in adults and is intended to provide a framework for management and further investigations into CVS in adults.


Subject(s)
Vomiting/diagnosis , Vomiting/physiopathology , Vomiting/therapy , Adult , Child , Humans , Syndrome
18.
Neurogastroenterol Motil ; 19(11): 923-36, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17973638

ABSTRACT

Intraperitoneal (i.p.) corticotropin releasing factor (CRF) induced a CRF(1) receptor-dependent stimulation of myenteric neurons and motility in the rat proximal colon. We characterize the colonic enteric nervous system response to CRF in conscious rats. Laser capture microdissection combined with reverse transcriptase polymerase chain reaction (RT-PCR) and immunohistochemistry in longitudinal muscle myenteric plexus whole-mount colonic preparations revealed CRF(1) receptor expression in myenteric neurons. CRF (i.p., 10 microg kg(-1)) induced Fos immunoreactivity (IR) (cells per ganglion) selectively in myenteric plexus of proximal (18.3 +/- 2.4 vs vehicle: 0.0 +/- 0.0) and distal colon (16.8 +/- 1.2 vs vehicle: 0.0 +/- 0.0), but not in that of gastric corpus, antrum, duodenum, jejunum and ileum. The selective CRF(1) agonist, stressin(1)-A (i.p., 10 microg kg(-1)) also induced Fos IR in myenteric but not in submucosal plexus of the proximal and distal colon. Fos IR induced by CRF was located in 55 +/- 1.9% and 53 +/- 5.1% of CRF(1) receptor-IR myenteric neurons and in 44 +/- 2.8% and 40 +/- 3.9% of cholinergic neurons with Dogiel type I morphology, and in 20 +/- 1.6% and 80 +/- 3.3% of nitrergic neurons in proximal and distal colon respectively. CRF and stressin(1)-A elicit defecation and diarrhoea. These data support that one mechanism through which peripherally injected CRF ligands stimulate colonic function involves a direct action on colonic cholinergic and nitrergic myenteric neurons expressing CRF(1) receptor.


Subject(s)
Cholinergic Fibers/metabolism , Colon/innervation , Corticotropin-Releasing Hormone/analogs & derivatives , Corticotropin-Releasing Hormone/pharmacology , Myenteric Plexus/metabolism , Nitrergic Neurons/metabolism , Peptides, Cyclic/pharmacology , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Cholinergic Fibers/pathology , Colon/pathology , Gastrointestinal Motility/drug effects , Male , Myenteric Plexus/pathology , Nitrergic Neurons/pathology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/agonists
19.
Endocrinology ; 148(12): 6115-23, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17932219

ABSTRACT

Interactions between gastrointestinal signals are a part of integrated systems regulating food intake (FI). We investigated whether cholecystokinin (CCK)-8 and urocortin systems potentiate each other to inhibit FI and gastric emptying (GE) in fasted mice. Urocortin 1 and urocortin 2 (1 microg/kg) were injected ip alone or with CCK (3 microg/kg) in lean, diet-induced obese (DIO) or corticotropin-releasing factor receptor-2 (CRF(2))-deficient mice. Gastric vagal afferent activity was recorded from a rat stomach-vagus in vitro preparation. When injected separately, urocortin 1, urocortin 2, or CCK did not modify the 4-h cumulative FI in lean mice. However, CCK plus urocortin 1 or CCK plus urocortin 2 decreased significantly the 4-h FI by 39 and 27%, respectively, compared with the vehicle + vehicle group in lean mice but not in DIO mice. Likewise, CCK-urocortin-1 delayed GE in lean but not DIO mice, whereas either peptide injected alone at the same dose had no effect. CCK-urocortin 2 suppression of FI was observed in wild-type but not CRF(2)-deficient mice. Gastric vagal afferent activity was increased by intragastric artery injection of urocortin 2 after CCK at a subthreshold dose, and the response was reversed by devazepide. These data establish a peripheral synergistic interaction between CCK and urocortin 1 or urocortin 2 to suppress FI and GE through CRF(2) receptor in lean mice that may involve CCK modulation of gastric vagal afferent responsiveness to urocortin 2. Such synergy is lost in DIO mice, suggesting a resistance to the satiety signaling that may contribute to maintain obesity.


Subject(s)
Cholecystokinin/pharmacology , Eating/drug effects , Peptide Fragments/pharmacology , Receptors, Corticotropin-Releasing Hormone/physiology , Satiation/drug effects , Urocortins/pharmacology , Animals , Cholecystokinin/administration & dosage , Drug Synergism , Gastric Emptying/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Models, Biological , Obesity/physiopathology , Peptide Fragments/administration & dosage , Rats , Receptors, Corticotropin-Releasing Hormone/genetics , Signal Transduction/drug effects , Stomach/drug effects , Stomach/innervation , Stomach/physiology , Urocortins/administration & dosage , Vagus Nerve/drug effects , Vagus Nerve/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...