Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Physiol Biochem ; 64(4): 349-56, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19391461

RESUMO

Although it is well established that the presence of nutrients in the gut lumen can bring about changes in GI function, the mechanisms and pathways by which these changes occur has not been fully elucidated. It has been known for many years that luminal nutrients stimulate the release of hormones and regulatory peptides from gut endocrine cells and that luminal nutrients activate intrinsic and extrinsic neural pathways innervating the gut. Activation of gut endocrine cells and neural pathways by nutrients in the gut lumen is key in coordination of postprandial GI function and also in the regulation of food intake. Recent evidence suggests that these pathways can be modified by long term changes in diet or by inflammatory processes in the gut wall. Thus it is important to determine the cellular and molecular mechanisms underlying these processes not only to increase our understanding of as part of basic physiology but also to understand changes in these pathways that occur in the presence of pathophysiology and disease. This review summarizes some of the latest data that we have obtained, together with information from the other laboratories, which have elucidated some of the mechanisms involved in nutrient detection in the gut wall. The focus is on monosaccharides and protein hydrolysates as there is some evidence for a role for nutrient transporters in detection of these nutrients.


Assuntos
Fenômenos Fisiológicos do Sistema Digestório , Células Enteroendócrinas/fisiologia , Alimentos , Trato Gastrointestinal/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Animais , Células Enteroendócrinas/metabolismo , Hormônios Gastrointestinais/metabolismo , Trato Gastrointestinal/inervação , Trato Gastrointestinal/metabolismo , Glucose/metabolismo , Humanos , Proteínas/metabolismo , Nervo Vago/fisiologia
2.
Endocrinology ; 148(10): 4695-703, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17641001

RESUMO

Peptide YY (PYY)(3-36), released by intestinal lipid elicits functional effects that comprise the intestinal feedback response to luminal nutrients, but the pathway of action is not fully characterized. The aim of the present study was to determine the role of the apolipoprotein (apo) A-IV-cholecystokinin (CCK)(1) receptor (CCK(1)R) pathway in exogenous PYY(3-36)-induced activation of the gut-brain axis and inhibition of gastric emptying and food intake. PYY(3-36) (5 microg/100 g ip) significantly inhibited gastric emptying of a chow meal in wild-type but not A-IV(-/-) mice andCCK(1)R receptor blockade with devazepide (10 microg/100 g), abolished PYY(3-36)-induced inhibition of gastric emptying. PYY(3-36)-induced inhibition of food intake in both ad libitum-fed and 16-h fasted mice was unaltered in A-IV(-/-) mice, compared with wild-type controls, or by CCK(1)R receptor blockade with devazepide. PYY(3-36) activated neurons in the midregion of the nucleus of the solitary tract (bregma -7.32 to -7.76 mm) in A-IV(+/+) mice; this was measured by immunohistochemical localization of Fos protein. PYY(3-36)-induced Fos expression was significantly reduced by 65% in A-IV(+/+) mice pretreated systemically with the sensory neurotoxin capsaicin (5 mg/100 g), 78% by the CCK(1)R antagonist, devazepide (10 microg/100 g), and 39% by the Y2R antagonist, BIIE0246 (200 and 600 microg/100 g) and decreased by 67% in apo A-IV(-/-) mice, compared with A-IV(+/+) controls. The data suggest a role for apo A-IV and the CCK(1)R in PYY(3-36)-induced activation of the vagal afferent pathway and inhibition of gastric emptying, but this is likely not the pathway mediating the effects of PYY(3-36) on food intake.


Assuntos
Apolipoproteínas A/fisiologia , Retroalimentação Fisiológica/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Intestinos/fisiologia , Peptídeo YY/farmacologia , Receptor de Colecistocinina A/fisiologia , Ração Animal , Animais , Apolipoproteínas A/deficiência , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Jejum , Retroalimentação Fisiológica/fisiologia , Esvaziamento Gástrico/efeitos dos fármacos , Esvaziamento Gástrico/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Fragmentos de Peptídeos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Núcleo Solitário/citologia , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/metabolismo , Núcleo Solitário/fisiologia
3.
Int J Obes Suppl ; 6(Suppl 1): S8-S14, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28685024

RESUMO

The microbiota-gut-brain axis is currently being explored in many types of rodent models, including models of behavioral, neurodegenerative and metabolic disorders. Our laboratory is interested in determining the mechanisms and consequences of activation of vagal afferent neurons that lead to activation of parasympathetic reflexes and changes in feeding behavior in the context of obesity. Obesity is associated with microbial dysbiosis, decreased intestinal barrier function, gut inflammation, metabolic endotoxemia, chronic low-grade systemic inflammation and desensitization of vagal afferent nerves. This review will present the evidence that altered gut microbiota together with decreased gut barrier function allows the passage of bacterial components or metabolites in obese individuals, leading to the disruption of vagal afferent signaling and consequently resulting in an increase in body weight. We first review the most recent descriptions of gut microbial dysbiosis due to a high fat diet and describe changes in the gut barrier and the evidence of increased intestinal permeability in obesity. We then will review the evidence to show how manipulating the gut microbiota via pre and probiotics can restore gut barrier function and prevent weight gain. Lastly, we present possible mechanisms by which the microbe-gut-brain axis may have a role in obesity. The studies mentioned in this review have provided new targets to treat and prevent obesity and have highlighted how the microbiota-gut-brain axis is involved.

4.
Neuroscience ; 20(1): 201-8, 1987 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3550518

RESUMO

The cellular origin of peptides derived from preprovasoactive intestinal peptide has been studied in rat stomach and ileum. Antisera specific for the C-terminal regions of the N-terminal flanking peptide (preprovasoactive intestinal peptide 22-80), bridging peptide (preprovasoactive intestinal peptide 111-124), C-terminal flanking peptide (preprovasoactive intestinal peptide 156-170) and vasoactive intestinal peptide were used in immunohistochemical studies on sections and whole mounts. All four antisera stained nerve fibres and cell bodies in the stomach and intestine. However, there were distinct differences in the pattern of colocalization of peptides derived from provasoactive intestinal peptide. In the sub-mucous plexus of the ileum virtually 100% of neurons reacting with vasoactive intestinal peptide antibodies also reacted with antibodies to the other three peptides. In contrast, in the stomach, while all vasoactive intestinal peptide-immunoreactive neurons of the myenteric plexus contained C-terminal flanking peptide- and bridging peptide-like immunoreactivity, only 50% of these cells reacted with the antiserum to N-terminal flanking peptide. The data indicate that in a population of neurons in the myenteric plexus of the rat stomach, preprovasoactive intestinal peptide is processed in such a way that the antigenic determinant of the N-terminal flanking peptide is not produced. In a second population of enteric neurons in the stomach, and in the intestine, it appears that processing of preprovasoactive intestinal peptide results in the production of peptides reacting with antibodies to vasoactive intestinal peptide, the flanking and bridging peptides.


Assuntos
Mucosa Gástrica/metabolismo , Íleo/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Especificidade de Anticorpos , Feminino , Imunofluorescência , Masculino , Plexo Mientérico/metabolismo , Ratos , Ratos Endogâmicos , Plexo Submucoso/metabolismo
5.
Neuroscience ; 123(1): 101-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14667445

RESUMO

Immunohistochemistry and confocal microscopy were used to investigate mu opioid receptor (muOR) internalization in enteric neurons of the guinea-pig ileum following abdominal surgery. The following surgical procedures were performed under halothane or isofluorane anesthesia: a) midline abdominal skin incision, b) laparotomy or c) laparotomy with intestinal manipulation. Gastrointestinal transit was evaluated by using a non-absorbable marker and measuring fecal pellet output. In neurons from normal and control (anesthesia alone) animals, muOR was predominantly at the cell surface. muOR endocytosis following skin incision was not significantly different from controls (21.2+/-3.5% vs. 13.7+/-2.1%, mean+/-S.E.M.), whereas it was significantly increased by laparotomy (46.5+/-6.1%; P<0.01 vs. controls) or laparotomy plus intestinal manipulation (40.5+/-6.1%; P<0.01 vs. controls) 30 min following surgery compared with controls. muOR endocytosis remained elevated at 4 h (38.6+/-1.2%; P<0.01 vs. controls), whereas it was similar to controls at 6 and 12 h (17.5+/-5.8% and 11.2+/-3.0%). muOR endocytosis occurred in cholinergic and nitrergic neurons. Gastrointestinal transit was significantly delayed by laparotomy or laparotomy plus intestinal manipulation (12.8+/-1.2 and 13.8+/-0.6 h vs. 7.0+/-0.5 in controls; P<0.01), but was not significantly changed by skin incision (8.2+/-0.6 h). The findings of the present study support the concept that the noxious stimulation caused by abdominal surgery induces release of endogenous opioids thus resulting in muOR endocytosis in neurochemically distinct enteric neurons. muOR internalization can serve as indirect evidence of opioid release and as a means to visualize neuronal pathways activated by opioids.


Assuntos
Endocitose/fisiologia , Sistema Nervoso Entérico/metabolismo , Íleo/metabolismo , Íleo/cirurgia , Receptores Opioides mu/metabolismo , Abdome/fisiologia , Abdome/cirurgia , Animais , Sistema Nervoso Entérico/química , Cobaias , Masculino , Neurônios/química , Neurônios/metabolismo , Receptores Opioides mu/análise
6.
Ann N Y Acad Sci ; 713: 143-56, 1994 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-8185155

RESUMO

Cholecystokinin (CCK) stimulates vagal afferent fiber discharge, both gastric and intestinal, which seems to result in reflex decrease in gastric motility, gastric acid secretion, and stimulation of pancreatic protein secretion. Endogenous release of CCK by fat or soybean trypsin inhibitor also alters function by way of a capsaicin-sensitive pathway. We suggest that CCK is released locally from the intestine and acts locally or systemically to stimulate vagal afferent fiber discharge to alter proximal gastrointestinal function (Fig. 14). In this way, in addition to its effect on food intake, CCK and the neural pathway integrate function in the proximal gastrointestinal tract, regulating the entry of food into the duodenum to ensure effective digestion and absorption.


Assuntos
Colecistocinina/farmacologia , Colecistocinina/fisiologia , Ingestão de Alimentos/fisiologia , Estômago/fisiologia , Nervo Vago/fisiologia , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/fisiologia , Animais , Benzodiazepinonas/farmacologia , Devazepida , Ingestão de Alimentos/efeitos dos fármacos , Esvaziamento Gástrico/efeitos dos fármacos , Mecanorreceptores/efeitos dos fármacos , Mecanorreceptores/fisiologia , Terminações Nervosas/efeitos dos fármacos , Terminações Nervosas/fisiologia , Ratos , Receptores da Colecistocinina/antagonistas & inibidores , Receptores da Colecistocinina/fisiologia , Sincalida/farmacologia , Estômago/efeitos dos fármacos , Estômago/inervação , Nervo Vago/efeitos dos fármacos
7.
Ann N Y Acad Sci ; 632: 272-82, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1952629

RESUMO

The present findings have revealed a new aspect of how mechanisms of gastric mucosal resistance to injury are called into effect and are coordinated by the nervous system. Capsaicin-sensitive sensory neurons in the stomach play a physiological role in monitoring acid influx into the superficial mucosa. Once activated, they strengthen gastric mucosal defense against deep injury, with a key process in this respect being an increase in blood flow through the gastric mucosa. This concept opens up completely new perspectives in the physiology and pathophysiology of the gastric mucosa if we consider that the long-term integrity of the gastric mucosa may be under the subtle control of acid-sensitive sensory neurons and that, vice versa, improper functioning of these neutral control mechanisms may predispose to gastric ulcer disease. The present observations also indicate that some of the peptides contained in gastric sensory nerve endings might fulfill a transmitter or mediator role in controlling gastric mucosal blood flow and integrity. Whereas substance P and neurokinin A are unlikely to play a role in the regulation of gastric mucosal blood flow, there is severalfold evidence that CGRP is very important in this respect. This peptide, which in the rat gastric mucosa originates exclusively from spinal sensory neurons, is released upon stimulation of sensory nerve endings and is extremely potent in facilitating gastric mucosal blood flow and in protecting the mucosa from injurious factors. Selective ablation of spinal sensory neurons containing CGRP weakens the resistance of the gastric mucosa against acid injury, which is most likely due to inhibition of protective vasodilator reflexes. We now aim at providing direct pharmacological evidence that antagonism of endogenously released CGRP results in similar pathophysiological consequences as ablation of capsaicin-sensitive sensory neurons.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Mucosa Gástrica/fisiologia , Neurônios Aferentes/fisiologia , Neuropeptídeos/fisiologia , Animais , Capsaicina/farmacologia , Mucosa Gástrica/irrigação sanguínea , Mucosa Gástrica/inervação , Neurônios Aferentes/efeitos dos fármacos , Fluxo Sanguíneo Regional
8.
Surgery ; 123(5): 518-27, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9591004

RESUMO

BACKGROUND: Calcitonin gene-related peptide (CGRP) is a widely distributed neuropeptide contained in intrinsic and extrinsic neurons of the gastrointestinal wall that has been shown to be released by noxious stimulation, to be involved in nociception, to inhibit gastrointestinal motility, and to partly mediate postoperative gastric ileus. We hypothesized that abdominal surgery-induced release of CGRP might inhibit postoperative colonic motility and food intake. METHODS: Colonic transit, stool pellet number, stool pellet weight, and food intake were measured for 48 hours after induction of postoperative ileus in rats. CGRP was immunoneutralized by preoperative injection of CGRP monoclonal antibody, or visceral afferent nerve fibers containing CGRP were functionally ablated by topical capsaicin treatment of the vagus nerves or of the celiac/superior mesenteric ganglia before abdominal surgery. RESULTS: Abdominal surgery increased colonic transit time and decreased 24-hour cumulative stool pellet number, stool pellet weight, and food intake. CGRP immunoneutralization reversed postoperative inhibition of colonic transit, 24-hour cumulative stool pellet number, stool pellet weight, and food intake by 77%, 82%, 80%, and 52%, respectively. Whereas ablation of vagal afferent nerve fibers had no effect, spinal afferent nerve fiber ablation reversed postoperative inhibition of 24-hour cumulative stool pellet number, stool pellet weight, and food intake by 41%, 38%, and 19%, respectively. CONCLUSIONS: CGRP and spinal afferent nerve fibers partly mediate postoperative colonic ileus and inhibition of food intake in the rat. By the magnitude of reversal of postoperative ileus, CGRP seems to be an important mediator of postoperative colonic ileus. Our results for the first time show involvement of a neuropeptide and spinal afferents in the mediation of postoperative colonic ileus and postoperative inhibition of food intake in rats.


Assuntos
Abdome/cirurgia , Vias Aferentes/fisiopatologia , Peptídeo Relacionado com Gene de Calcitonina/análise , Doenças do Colo/fisiopatologia , Obstrução Intestinal/fisiopatologia , Complicações Pós-Operatórias/fisiopatologia , Medula Espinal/fisiopatologia , Nervo Vago/fisiopatologia , Vias Aferentes/química , Animais , Anticorpos Monoclonais , Capsaicina/farmacologia , Doenças do Colo/etiologia , Defecação , Comportamento Alimentar , Gânglios Simpáticos/efeitos dos fármacos , Gânglios Simpáticos/fisiopatologia , Trânsito Gastrointestinal , Obstrução Intestinal/etiologia , Masculino , Ratos , Ratos Sprague-Dawley , Medula Espinal/química , Circulação Esplâncnica , Nervo Vago/efeitos dos fármacos
9.
Neuroreport ; 8(3): 733-7, 1997 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-9106757

RESUMO

The mechanism(s) by which intestinal smooth muscle tension is signaled to extrinsic primary afferent neurons is poorly understood. In order to characterize myocyte-neuron communication, we developed a coculture system using rat dorsal root ganglion (DRG) neurons and myocytes obtained from the circular muscle layer of the rat distal colon. Both cell types maintained their phenotype in culture, as demonstrated by positive immunocytochemical staining for neuron-specific enolase and smooth muscle actin. Myocytes showed mechanosensitivity in the form of increases in [Ca2+]i in response to light mechanical touch of the plasma membrane. This increase in [Ca2+]i was independent of extracellular Ca2+ and passed as a propagated wave from muscle cells into adjacent DRG neurites. The inhibitory effect of octanol on this intercellular propagation suggests propagation of [Ca2+]i gradients via heterologous gap junctions. This preparation may serve a useful model system for the study of the interaction of visceral afferents and their target cells.


Assuntos
Vias Aferentes/fisiologia , Colo/fisiologia , Gânglios Espinais/fisiologia , Músculo Liso/fisiologia , Neurônios/fisiologia , 1-Octanol , Animais , Transporte Axonal , Cálcio/metabolismo , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Colo/citologia , Técnica Indireta de Fluorescência para Anticorpo , Gânglios Espinais/citologia , Imuno-Histoquímica , Masculino , Músculo Liso/citologia , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neurônios/citologia , Octanóis/farmacologia , Ratos , Ratos Sprague-Dawley
10.
Peptides ; 12(6): 1279-83, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1815214

RESUMO

The role of vagal afferent pathways and cholecystokinin (CCK) in mediating changes in gastric motor function after a meal was investigated in urethane-anesthetized rats. Proximal gastric motor function was measured manometrically, and nutrients were infused into an isolated segment of duodenum. Inhibition of gastric motility in response to duodenal infusion of protein (peptone or casein), but not carbohydrate (glucose), was significantly attenuated by administration of the CCK antagonist, L364,718. Selective ablation of vagal afferents by perineural treatment with the sensory neurotoxin, capsaicin, significantly reduced responses to both duodenal protein and glucose. These results suggest that protein in the duodenum decreases proximal gastric motor function via release of CCK and a vagal capsaicin-sensitive afferent pathway. In contrast, glucose acts via a capsaicin-sensitive vagal pathway not involving CCK. Thus separate neural and hormonal mechanisms mediate the effects of different nutrients in the duodenal feedback regulation of gastric motor function.


Assuntos
Capsaicina/farmacologia , Colecistocinina/fisiologia , Esvaziamento Gástrico/efeitos dos fármacos , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/fisiologia , Animais , Benzodiazepinonas/farmacologia , Colecistocinina/antagonistas & inibidores , Devazepida , Duodeno/inervação , Duodeno/fisiologia , Retroalimentação , Esvaziamento Gástrico/fisiologia , Masculino , Fenômenos Fisiológicos da Nutrição , Ratos , Ratos Endogâmicos , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia
11.
Peptides ; 17(8): 1307-11, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8971924

RESUMO

CCK-58 has been shown to be the major circulating form of the hormone in the dog and human. To date, there have been no reports on its biological activity in vivo. We report here that CCK-8 and CCK-58 were equipotent in decreasing gastric motor function after bolus doses and in stimulating protein secretion after continuous infusion in urethane-anesthetized rats. The present results are the first on the in vivo activity of CCK-58, and indicate that because CCK-58 is equipotent to CCK-8, and because it is a major released and circulating form, it may be considered as a major contributor to the expression of cholecystokinin bioactivity.


Assuntos
Colecistocinina/farmacologia , Colecistocinina/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Sincalida/farmacologia , Sincalida/fisiologia , Aminoácidos/análise , Animais , Colecistocinina/química , Cães , Motilidade Gastrointestinal/fisiologia , Humanos , Indicadores e Reagentes , Masculino , Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Sincalida/química
12.
Peptides ; 9(4): 735-7, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-3265786

RESUMO

The central nervous system action of rat alpha-calcitonin gene-related peptide (alpha-CGRP) on gastric emptying of a liquid, noncaloric, methylcellulose solution was assessed in 24-hr fasted, conscious rats using phenol red method as a marker. Intracisternal injection of alpha-CGRP (0.75-250 pmol) dose-dependently inhibited gastric emptying by 27-94% as measured 20 min after oral administration of the solution. The ED50 was 6.2 pmol. alpha-CGRP injected intravenously at 250 pmol delayed gastric emptying by 71% whereas a lower dose (75 pmol) was inactive. Intracisternal alpha-CGRP-induced inhibition of gastric emptying was completely abolished by bilateral adrenalectomy and partially suppressed by subdiaphragmatic vagotomy or coeliac/superior mesenteric ganglionectomy. Adrenalectomy or vagotomy in saline-treated animals did not significantly modify the rate of gastric emptying whereas coeliac/superior mesenteric ganglionectomy caused a significant 29% inhibition as compared to the nonoperated group. These results demonstrate that alpha-CGRP is a potent centrally acting inhibitor of gastric emptying of a nonnutrient liquid. The inhibitory effect of intracisternal injection of CGRP appears to be mediated by the adrenal gland and in part by the sympathetic and parasympathetic nervous system.


Assuntos
Calcitonina/farmacologia , Cisterna Magna/fisiologia , Esvaziamento Gástrico/efeitos dos fármacos , Neuropeptídeos/farmacologia , Adrenalectomia , Animais , Peptídeo Relacionado com Gene de Calcitonina , Cisterna Magna/efeitos dos fármacos , Relação Dose-Resposta a Droga , Injeções , Injeções Intravenosas , Masculino , Neuropeptídeos/administração & dosagem , Ratos , Ratos Endogâmicos , Valores de Referência , Vagotomia
13.
Peptides ; 14(4): 743-7, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8234019

RESUMO

The influence of corticotropin releasing factor (CRF) microinjected into the paraventricular nucleus of the hypothalamus (PVN) on colonic motility was investigated in conscious, fasted rats. Rats were chronically implanted with a bilateral guide cannula into the PVN and a catheter into the proximal colon to record motor activity manometrically. Microinjection of CRF (0.6 nmol/rat) into the PVN increased both phasic and tonic motor activity in the proximal colon. Atropine sulfate (1 mg/kg, IP) completely abolished the colonic motor response to CRF. Microinjection of CRF (0.6 nmol/rat) into sites outside of the PVN did not modify colonic motor activity. These data show that CRF acts in the PVN to stimulate tonic and phasic motor activity in the proximal colon. Corticotropin releasing factor action is site specific and mediated through cholinergic pathways.


Assuntos
Colo/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Jejum , Motilidade Gastrointestinal/fisiologia , Núcleo Hipotalâmico Paraventricular/química , Animais , Atropina/farmacologia , Masculino , Microinjeções , Ratos , Ratos Sprague-Dawley
14.
Peptides ; 13(2): 249-54, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1409004

RESUMO

The gastric mucosa, in particular submucosal blood vessels, are innervated by afferent neurons containing neuropeptides such as calcitonin gene-related peptide. Stimulation of sensory neurons innervating the gastric mucosa increases submucosal blood flow. Since sensory neurons supplying the stomach are of dual origin from nodose and dorsal root ganglia, we examined the effect of selective ablation of either the vagal or spinal sensory innervation to the upper gastrointestinal tract on the increase in gastric mucosal blood flow in response to acid back diffusion into the gastric mucosa. Perineural application of capsaicin to the celiac/superior mesenteric ganglia, but not to the vagus nerves, significantly inhibited by 53% the hyperemic response to acid back diffusion. Tissue levels of immunoreactive calcitonin gene-related peptide in the gastric corpus were significantly reduced (by 73%) by periceliac capsaicin treatment, but unaffected by perivagal capsaicin treatment. These data suggest that spinal capsaicin-sensitive afferents containing calcitonin gene-related peptide immunoreactivity are involved in mediating increases in gastric mucosal blood flow. This increase in gastric mucosal blood flow mediated by sensory neurons may act as a protective mechanism against mucosal injury, similar to responses seen in other tissues such as skin.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Ácido Gástrico/metabolismo , Mucosa Gástrica/inervação , Hiperemia/fisiopatologia , Neurônios Aferentes/fisiologia , Nervos Espinhais/fisiologia , Animais , Bradicardia/fisiopatologia , Denervação , Difusão , Mucosa Gástrica/irrigação sanguínea , Mucosa Gástrica/metabolismo , Masculino , Ratos , Ratos Endogâmicos , Distribuição Tecidual , Nervo Vago/fisiologia
15.
Peptides ; 11(4): 789-95, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2122423

RESUMO

Central injection of TRH or its stable analog, RX77368, produces a vagal cholinergic stimulation of gastric acid secretion, mucosal blood flow and motor function. In the present study, we have investigated the contribution of capsaicin-sensitive vagal afferent fibers to the gastric responses to intracisternal injection of RX77368. Gastric acid secretion, measured in acute gastric fistula rats anesthetized with urethane, in response to intracisternal injection of RX77368 (3-30 ng) was reduced by 21-65% by perineural pretreatment of the vagus nerves with capsaicin 10-20 days before experiments. The increase in gastric mucosal blood flow measured by hydrogen gas clearance induced by intracisternal injection of RX77368 (30 ng) was also reduced by 65% in capsaicin-pretreated rats. In contrast, increases in gastric motor function measured manometrically or release of gastric luminal serotonin in response to intracisternal injection of RX77368 (3-30 ng) were unaltered by capsaicin pretreatment. The mechanism by which vagal afferent fibers contribute to the secretory and blood flow responses to the stable TRH analog is unclear at present, but it is possible that the decrease in gastric mucosal blood flow by lesion of capsaicin-sensitive vagal afferents limits the secretory response.


Assuntos
Capsaicina/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Hormônio Liberador de Tireotropina/análogos & derivados , Nervo Vago/efeitos dos fármacos , Vias Aferentes/efeitos dos fármacos , Animais , Vasos Sanguíneos/efeitos dos fármacos , Cisterna Magna , Ácido Gástrico/metabolismo , Mucosa Gástrica/irrigação sanguínea , Mucosa Gástrica/metabolismo , Motilidade Gastrointestinal/efeitos dos fármacos , Injeções , Masculino , Ácido Pirrolidonocarboxílico/análogos & derivados , Ratos , Ratos Endogâmicos , Fluxo Sanguíneo Regional , Serotonina/metabolismo , Hormônio Liberador de Tireotropina/farmacologia
16.
Peptides ; 14(6): 1225-9, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-7510881

RESUMO

The role of capsaicin-sensitive pathways and CGRP in postoperative gastric ileus was investigated. Abdominal surgery was performed under enflurane anesthesia, and 5 min later, the 20-min rate of gastric emptying was measured by the phenol red method in conscious rats. Surgery inhibited gastric emptying by 76-83% compared with rats receiving anesthesia alone. Capsaicin on the celiac/mesenteric ganglia (10-21 days before) reduced gastric ileus by 33 +/- 8%, whereas perivagal capsaicin had no effect. The IV CGRP-induced inhibition of gastric emptying was completely reversed by the CGRP antagonist, CGRP(8-37) (30 micrograms, IV); CGRP(8-37) (15, 30, or 60 micrograms) or CGRP monoclonal antibody #4901 (2 mg protein) decreased the inhibition of gastric emptying by 11 +/- 7%, 51 +/- 13%, 47 +/- 3%, and 45 +/- 17%, respectively. These results indicate that CGRP and splanchnic capsaicin-sensitive afferents are involved in mediating part of the gastric ileus observed immediately after abdominal surgery.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Capsaicina/farmacologia , Gânglios Simpáticos/efeitos dos fármacos , Gastropatias/prevenção & controle , Vias Aferentes/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/antagonistas & inibidores , Peptídeo Relacionado com Gene de Calcitonina/imunologia , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Masculino , Fragmentos de Peptídeos/farmacologia , Complicações Pós-Operatórias/prevenção & controle , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Substância P/antagonistas & inibidores , Nervo Vago/efeitos dos fármacos
17.
Regul Pept ; 79(2-3): 125-30, 1999 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-10100925

RESUMO

UNLABELLED: PYY is released from the distal ileum by fat and may be involved in mediating lipid-induced inhibition of gastric acid secretion and intestinal motility. The role of PYY in intestinal lipid-induced inhibition of gastric emptying in awake rats was investigated using a specific polyclonal antibody raised against PYY. METHODS: Gastric emptying of liquids was measured in awake rats fitted with a Thomas gastric cannula. Intralipid (total dose 50 or 100 mg) was perfused for 10 min (0.05 ml/min) into a duodenal (n = 11) or mid-intestinal cannula (60 cm from Ligament of Treitz; n = 8), and gastric emptying was measured over the 5-10 min period. Gastric emptying was measured 15 min after IP injection of PYY (1 nmol/rat). PYY antibody (20 mg) or a control antibody (anti-KLH; keyhole limpet hemocyanin) was injected ip 8-12 h before experiments. RESULTS: Exogenous PYY (1 nmol) inhibited gastric emptying and administration of PYY antibody blocked this response. Perfusion of lipid (50 and 100 mg) into the proximal intestine produced a 46% and 66% inhibition of gastric emptying respectively. Inhibition of gastric emptying in response to 50 mg lipid in the proximal small intestine was unaffected by administration of PYY antibody but was abolished by administration of the CCK A receptor antagonist devazepide (0.1 mg/kg ip). Perfusion of lipid into the distal intestine (50 and 100 mg) inhibited gastric emptying by 10% and 32% respectively. Inhibition of gastric emptying in response to 100 mg lipid in the distal intestine was unaffected by PYY antibody. CONCLUSIONS: Lipid perfused into either the proximal or distal intestine inhibits gastric emptying via a PYY-independent mechanism. CCK is involved in proximal lipid induced inhibition of gastric emptying.


Assuntos
Esvaziamento Gástrico/fisiologia , Metabolismo dos Lipídeos , Peptídeo YY/metabolismo , Animais , Devazepida/farmacologia , Antagonistas de Hormônios/farmacologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/fisiologia , Lipídeos/farmacologia , Testes de Neutralização , Ratos , Ratos Sprague-Dawley
18.
Neurogastroenterol Motil ; 7(1): 9-14, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7627868

RESUMO

Carbohydrates are a component of chyme that initiate feedback control of gastric emptying. The aim of the study was to investigate the mechanism by which sensors in the intestine are activated by carbohydrate to initiate intestinal feedback of gastric motor function. Intestinal perfusion with D-glucose inhibited gastric motility in awake rats. This response was reproduced by 3-O-methyl glucose, a non-metabolizable analogue of glucose that is absorbed by the Na(+)-glucose co-transporter, but not by 2-deoxy-D-glucose, and was attenuated by perfusion of the intestine with phloridzin, a competitive blocker of the Na(+)-glucose co-transporter. Feeding a high carbohydrate diet to increase the number of co-transporters reduced the response to intestinal glucose. It was concluded that activation of sensors to initiate feedback inhibition of gastric motility may be dependent either on rapid accumulation of glucose within epithelial cells or on activation of the Na(+)-glucose co-transporter.


Assuntos
Proteínas de Transporte/metabolismo , Motilidade Gastrointestinal/fisiologia , Glucose/farmacologia , Intestinos/fisiologia , Glicoproteínas de Membrana , Proteínas de Membrana/metabolismo , Proteínas de Transporte de Monossacarídeos , Animais , Depressão Química , Carboidratos da Dieta/farmacologia , Retroalimentação/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Glucose/análogos & derivados , Absorção Intestinal/efeitos dos fármacos , Masculino , Manitol/farmacologia , Concentração Osmolar , Florizina/farmacologia , Ratos , Ratos Sprague-Dawley , Transportador 1 de Glucose-Sódio
19.
Neurogastroenterol Motil ; 16 Suppl 1: 60-3, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15066007

RESUMO

The enteric nervous system in combination with inputs from parasympathetic and sympathetic nerves regulate the contractile, secretory and vasomotor activity of the gastrointestinal track via neural reflexes. Sensory elements which may be present in specialized neurones, enteroendocrine cells or mast cells detect changes in force, chemical composition or even foreign antigens. Sensory elements signal the enteric nervous system to correct these changes by altering contractile activity, secretion and blood flow. Advances have been made in understanding the sensory mechanisms that are involved in 5-hydroxytryptamine (5-HT) release from enterochromaffin cells (EC) or a model for EC cells. These advances relate to roles for ATP and its metabolites ADP and adenosine in mechanotransduction and a role for a sodium glucose cotransporter, a SGLT-like protein, in chemotransduction.


Assuntos
Sistema Digestório/inervação , Sistema Nervoso Entérico/fisiologia , Células Enterocromafins/fisiologia , Mecanotransdução Celular/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Sistema Nervoso Entérico/citologia , Motilidade Gastrointestinal/fisiologia , Humanos , Reflexo/fisiologia
20.
Neurogastroenterol Motil ; 14(4): 403-8, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12213108

RESUMO

Abstract In a number of different experimental paradigms of somatic pain, there is evidence for a vagally mediated antinociceptive system. This pathway probably involves opioid mechanisms. However, whether this pathway is activated in visceral pain or if it involves subdiaphragmatic vagal afferents is unclear. The aim of the present study was to determine whether subdiaphragmatic vagal afferents mediate antinociception in response to a visceral stimulus and whether this involves an opioid pathway. Colorectal distension was performed in fasted, conscious male Sprague-Dawley rats using a balloon catheter connected to an electronic distension device. The number of abdominal contractions (visceromotor response) in response to a tonic colorectal distension (60 mmHg for 10 min) was recorded. Experiments were performed in sham or subdiaphragmatically vagotomized, perineural vehicle- or capsaicin-treated rats (to functionally denervate vagal afferents) before and after administration of naloxone (25 mg kg(-1) bodyweight intraperitoneally). Vagotomy, capsaicin and naloxone pretreatments all significantly enhanced the visceromotor response to colorectal distension. The effect of naloxone in capsaicin-treated rats did not appear to be additive. These results suggest that activation of subdiaphragmatic afferents, which can be blocked by capsaicin, may play a role in opioid-dependent antinociceptive pathways activated by a noxious visceral stimulus.


Assuntos
Colo/fisiologia , Diafragma/inervação , Peptídeos Opioides/fisiologia , Medição da Dor/métodos , Reto/fisiologia , Nervo Vago/fisiologia , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/fisiologia , Animais , Colo/efeitos dos fármacos , Colo/inervação , Diafragma/efeitos dos fármacos , Diafragma/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Masculino , Naloxona/farmacologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/estatística & dados numéricos , Ratos , Ratos Sprague-Dawley , Reto/efeitos dos fármacos , Reto/inervação , Vagotomia/métodos , Vagotomia/estatística & dados numéricos , Nervo Vago/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA