Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Regul Pept ; 119(3): 209-12, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15120482

RESUMO

Orexin A (OXA) is found in the central nervous system (CNS) and in the gut. Peripheral administration of OXA to rats results in an inhibition of fasting motility. Plasma OXA increases during fasting and central administration of OXA increases food intake. The aim of the present study was to assess the pharmacokinetic profile of OXA and the effect of intravenously (i.v.) administered OXA on plasma concentrations of insulin and glucagon concentrations. Rats were given OXA i.v. (100 pmol kg(-1) min(-1)) for time periods of 0, 10, 20, 30 min and for 10, 20, 30 min after ceasing a 30-min infusion. After each time period, rats were then sacrificed and blood obtained. OXA was also administered at increasing doses (0, 100, 300 and 500 pmol kg(-1) min(-1)) for 30 min and blood was obtained. Plasma OXA, insulin and glucagon levels were measured using commercially available radioimmunoassay (RIA) kits. The plasma half-life of OXA was 27.1+/-9.5 min. Stepwise increasing infusion rates of OXA confirmed a linear concentration-time curve and thus first-order kinetics. Its volume of distribution indicated no binding to peripheral tissues. Plasma glucagon decreased during infusion of OXA, while insulin was unaffected. Plasma OXA was raised fourfold after food intake. Thus, OXA has a longer plasma half-life than many other peptides found in the gut. This needs to be taken into account when assessing effects of OXA on biological parameters after peripheral administration.


Assuntos
Glucagon/sangue , Insulina/sangue , Peptídeos e Proteínas de Sinalização Intracelular/farmacocinética , Neuropeptídeos/farmacocinética , Animais , Sistema Nervoso Central/metabolismo , Jejum/metabolismo , Meia-Vida , Infusões Intravenosas , Mucosa Intestinal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Neuropeptídeos/administração & dosagem , Neuropeptídeos/metabolismo , Orexinas , Ratos , Ratos Sprague-Dawley
2.
Am J Physiol Gastrointest Liver Physiol ; 285(4): G688-95, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12816759

RESUMO

Orexin A (OXA)-positive neurons are found in the lateral hypothalamic area and the enteric nervous system. The aim of this study was to investigate the mechanism of OXA action on small bowel motility. Electrodes were implanted in the serosa of the rat small intestine for recordings of myoelectric activity during infusion of saline or OXA in naive rats, vagotomized rats, rats pretreated with guanethidine (3 mg/kg) or N(omega)-nitro-L-arginine (L-NNA; 1 mg/kg). Naive rats were given a bolus of the orexin receptor-1 (OX1R) antagonist (SB-334867-A; 10 mg/kg), and the effect of both OXA and SB-334867-A on fasting motility was studied. Double-label immunocytochemistry with primary antibodies against OXA, neuronal nitric oxide synthase (nNOS), and OX1R was performed. OXA induced a dose-dependent prolongation of the cycle length of the migrating myoelectric complex (MMC) and, in the higher doses, replaced the activity fronts with an irregular spiking pattern. Vagotomy or pretreatment with guanethidine failed to prevent the response to OXA. The OXA-induced effect on the MMC cycle length was completely inhibited by pretreatment with L-NNA (P < 0.05), as did SB-334867-A. The OX1R antagonist shortened the MMC cycle length from 14.1 (12.0-23.5) to 11.0 (9.5-14.7) min (P < 0.05) during control and treatment periods, respectively. Colocalization of OXA and nNOS was observed in myenteric neurons of the duodenum and nerve fibers in the circular muscle. Our results indicate that OXA inhibition of the MMC involves the OX1R and that activation of a L-arginine/NO pathway possibly originating from OX1R/nNOS-containing neurons in the myenteric plexus may mediate this effect. Endogenous OXA may have a physiological role in regulating the MMC.


Assuntos
Proteínas de Transporte/farmacologia , Intestino Delgado/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Complexo Mioelétrico Migratório/efeitos dos fármacos , Neuropeptídeos/farmacologia , Óxido Nítrico/farmacologia , Receptores de Neuropeptídeos/fisiologia , Ureia/análogos & derivados , Animais , Benzoxazóis/farmacologia , Proteínas de Transporte/análise , Duodeno/química , Eletromiografia , Guanetidina/farmacologia , Homeostase , Imuno-Histoquímica , Intestino Delgado/fisiologia , Masculino , Complexo Mioelétrico Migratório/fisiologia , Naftiridinas , Neuropeptídeos/análise , Neurotransmissores/farmacologia , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase Tipo I , Nitroarginina/farmacologia , Receptores de Orexina , Orexinas , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G , Receptores de Neuropeptídeos/análise , Receptores de Neuropeptídeos/antagonistas & inibidores , Ureia/farmacologia , Vagotomia
3.
Am J Physiol Gastrointest Liver Physiol ; 282(3): G470-9, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11841997

RESUMO

The orexins [orexin A (OXA) and orexin B (OXB)] are novel neuropeptides that increase food intake in rodents. The aim of this study was to determine the distribution of orexin and orexin receptors (OX1R and OX2R) in the rat duodenum and examine the effects of intravenous orexin on fasting gut motility. OXA-like immunoreactivity was found in varicose nerve fibers in myenteric and submucosal ganglia, the circular muscle, the mucosa, submucosal and myenteric neurons, and numerous endocrine cells of the mucosa. OXA neurons displayed choline acetyltransferase immunoreactivity, and a subset contained vasoactive intestinal peptide. OXA-containing endocrine cells were identified as enterochromaffin (EC) cells based on the presence of 5-hydroxytryptamine immunoreactivity. OX1R was expressed by neural elements of the gut, and EC cells expressed OX2R. OXA at 100 and 500 pmol x kg(-1) x min(-1) significantly increased the myoelectric motor complex (MMC) cycle length compared with saline. Similarly, OXB increased the MMC cycle length at 100 pmol x kg(-1) x min(-1), but there was no further effect at 500 pmol x kg(-1) x min(-1). We postulate that orexins may affect the MMC through actions on enteric neurotransmission after being released from EC cells and/or enteric neurons.


Assuntos
Proteínas de Transporte/análise , Proteínas de Transporte/farmacologia , Duodeno/fisiologia , Jejum , Motilidade Gastrointestinal/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Neuropeptídeos/análise , Neuropeptídeos/farmacologia , Animais , Duodeno/química , Duodeno/efeitos dos fármacos , Eletromiografia , Imunofluorescência , Imuno-Histoquímica , Masculino , Neurônios/química , Receptores de Orexina , Orexinas , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G , Receptores de Neuropeptídeos/análise
4.
Neuroreport ; 12(18): 3929-34, 2001 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-11742213

RESUMO

Previous reports have indicated that vesicular glutamate transporters (VGLUTs) are found only in central neurons. We show that neurons in the gut, which also contain glutamate and markers of intrinsic primary afferent neurons, display VGLUT2 immunoreactivity in several species, including humans. Glutamatergic (VGLUT2-immunoreactive) varicosities, which often co-stored choline acetyltransferase and the vesicular acetylcholine transporter, were apposed to a subset of nerve cell bodies in the submucosal and myenteric plexus. Retrograde tracing with FluoroGold demonstrated that VGLUT2 is found in nodose and dorsal root ganglia neurons innervating the stomach. Thus, VGLUT2 is found in intrinsic and extrinsic primary afferent neurons, which suggests that glutamate is as primary afferent neurotransmitter that transfers information from the mucosa to the enteric plexuses and brain.


Assuntos
Proteínas de Transporte/análise , Gânglios Espinais/química , Proteínas de Membrana Transportadoras , Plexo Mientérico/química , Gânglio Nodoso/química , Proteínas de Transporte Vesicular , Animais , Anticorpos , Química Encefálica , Calbindinas , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Células Cultivadas , Colina O-Acetiltransferase/metabolismo , Imunofluorescência , Gânglios Espinais/citologia , Ácido Glutâmico/metabolismo , Cobaias , Masculino , Plexo Mientérico/citologia , Neurônios Aferentes/química , Neurônios Aferentes/citologia , Gânglio Nodoso/citologia , Proteína G de Ligação ao Cálcio S100/análise , Proteína Vesicular 2 de Transporte de Glutamato
5.
Anat Rec ; 262(1): 91-100, 2001 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-11146432

RESUMO

Pancreatic ganglia are innervated by neurons in the gut and are formed by precursor cells that migrate into the pancreas from the bowel. The innervation of the pancreas, therefore, may be considered an extension of the enteric nervous system. Pituitary adenylate cyclase-activating polypeptide (PACAP) is present in a subset of enteric neurons. We investigated the presence of PACAP in the enteropancreatic innervation in guinea pigs, and the response of pancreatic neurons to PACAP-related peptides. PACAP immunoreactivity was found in nerve fibers in both enteric and pancreatic ganglia and in nerve bundles that travelled between the duodenum and pancreas. PACAP-immunoreactive nerve fibers were densely distributed in the pancreatic ganglia, where they surrounded a subset of cholinergic cell bodies. Pancreatic ganglia did not contain PACAP-immunoreactive cell bodies; however, neuronal perikarya with PACAP immunoreactivity were found in the myenteric plexus of the duodenum. These cells co-stored vasoactive intestinal peptide (VIP). PACAP depolarized pancreatic neurons. Pancreatic neurons were also depolarized by VIP; however, PACAP was more efficacious at depolarizing pancreatic cells than VIP. These findings are consistent with the view that the PACAP effects were mediated through PACAP-selective (PAC1) receptors. PACAP-responsive neurons displayed PAC1 receptor immunoreactivity, which was also found in islet cells and enteric neurons. These results provide support for the hypothesis that PACAP modulates reflex activity between the gut and pancreas. The excitatory effect of PACAP would be expected to potentiate pancreatic secretion.


Assuntos
Plexo Mientérico/metabolismo , Neuropeptídeos/metabolismo , Neurotransmissores/metabolismo , Pâncreas/inervação , Animais , Duodeno/citologia , Duodeno/metabolismo , Eletrofisiologia , Técnica Indireta de Fluorescência para Anticorpo , Cobaias , Técnicas In Vitro , Masculino , Plexo Mientérico/citologia , Fibras Nervosas/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores do Hormônio Hipofisário/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo
6.
Curr Opin Pharmacol ; 1(6): 591-6, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11757814

RESUMO

Several lines of evidence indicate a role for glutamate in the regulation of gut motility and secretion; however, the receptor subtypes that mediate the effects of this amino acid are still incompletely understood. There has, however, been recent progress in pharmacological characterization of enteric glutamate receptor subtypes. In the past two years, investigators have demonstrated that in addition to ionotropic glutamate receptors, the enteric nervous system contains functional group I metabotropic glutamate receptors that appear to participate in enteric reflexes. This opens up an entirely new arena in which to study the roles of glutamate in gut function and presents potential new target sites for drug development.


Assuntos
Sistema Nervoso Entérico/metabolismo , Ácido Glutâmico/metabolismo , Receptores de Glutamato/metabolismo , Animais , Sistema Digestório/anatomia & histologia , Fenômenos Fisiológicos do Sistema Digestório , Ácido Glutâmico/fisiologia , Humanos , Receptores de AMPA/metabolismo , Receptores de Glutamato/fisiologia , Receptores de Ácido Caínico/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica
7.
J Neurosci ; 20(9): 3200-5, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10777784

RESUMO

We demonstrate that metabotropic glutamate receptor 5 (mGluR5) is present in the guinea pig ileum. A punctate ring-like distribution of immunoreactivity is found on the soma of a subset of neurons, consistent with an association of mGluR5 with the plasma membrane. mGluR5-containing cells in the submucosal plexus are predominantly noncholinergic and contain vasoactive intestinal peptide, a marker of secretomotor neurons. Using immunocytochemistry in conjunction with confocal microscopy, we show that the mGluR5 undergoes agonist- and reflex-evoked internalization that is inhibited by the group I antagonist 1-aminoindan-1,5-dicarboxylic acid. In addition, group I mGluR antagonists reduce the distension-induced phosphorylation of cAMP-responsive element-binding protein in enteric neurons and attenuate both glutamate- and group I agonist-induced depolarizing responses and slow synaptic events in submucosal neurons. These findings support the idea that mGluRs play a role in enteric reflexes and suggest that internalization might be a major mechanism for regulation of mGluR activity.


Assuntos
Sistema Nervoso Entérico/metabolismo , Neurônios/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Plexo Submucoso/metabolismo , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/farmacologia , Cobaias , Íleo/metabolismo , Masculino , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inibidores
8.
J Neurosci ; 19(23): 10305-17, 1999 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10575028

RESUMO

We tested the hypothesis that a subset of enteric neurons is glucoresponsive and expresses ATP-sensitive K(+) (K(ATP)) channels. The immunoreactivities of the inwardly rectifying K(+) channel 6.2 (Kir6.2) and the sulfonylurea receptor (SUR), now renamed SUR1, subunits of pancreatic beta-cell K(ATP) channels, were detected on cholinergic neurons in the guinea pig ileum, many of which were identified as sensory by their costorage of substance P and/or calbindin. Glucoresponsive neurons were distinguished in the myenteric plexus because of the hyperpolarization and decrease in membrane input resistance that were observed in response to removal of extracellular glucose. The effects of no-glucose were reversed on the reintroduction of glucose or by the K(ATP) channel inhibitor tolbutamide. No reversal of the hyperpolarization was observed when D- mannoheptulose, a hexokinase inhibitor, was present on the reintroduction of glucose. Application of the K(ATP) channel opener diazoxide or the ob gene product leptin mimicked the effect of glucose removal in a reversible manner; moreover, hyperpolarizations evoked by either agent were inhibited by tolbutamide. Glucoresponsive neurons displayed leptin receptor immunoreactivity, which was widespread in both enteric plexuses. Superfusion of diazoxide inhibited fast synaptic activity in myenteric neurons, via activation of presynaptic K(ATP) channels. Diazoxide also produced a decrease in colonic motility. These experiments demonstrate for the first time the presence of glucoresponsive neurons in the gut. We propose that the glucose-induced excitation of these neurons be mediated by inhibition of K(ATP) channels. The results support the idea that enteric K(ATP) channels play a role in glucose-evoked reflexes.


Assuntos
Sistema Nervoso Entérico/fisiologia , Glucose/fisiologia , Neurônios/fisiologia , Trifosfato de Adenosina/fisiologia , Animais , Colo/efeitos dos fármacos , Diazóxido/farmacologia , Eletrofisiologia , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/metabolismo , Motilidade Gastrointestinal/efeitos dos fármacos , Cobaias , Leptina/farmacologia , Masculino , Plexo Mientérico/citologia , Plexo Mientérico/metabolismo , Plexo Mientérico/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Canais de Potássio/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Tolbutamida/farmacologia
9.
J Pharmacol Exp Ther ; 291(1): 374-82, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10490927

RESUMO

The whole-cell patch-clamp technique was used to analyze the effects of 5-hydroxytryptamine (5-HT) and alosetron on cultured myenteric neurons from newborn guinea pigs. All neurons responded to 5-HT (EC(50) approximately 38.7 microM) with a concentration-dependent inward current (reversal potential = 7.1 +/- 1.7 mV) with a short latency and rapid decay. Because the 5-HT-induced inward current was mimicked by 2-methyl-5-hydroxytryptamine (50 microM) and blocked by ondansetron (5.0 microM) and MDL 72222 (0.05 microM), it was 5-HT(3)-mediated. Alosetron blocked (IC(50) approximately 0.05 microM; Hill coefficient approximately 1.24) the 5-HT- and 2-methyl-5-hydroxytryptamine-induced inward currents. This effect was independent of membrane potential and was not seen when alosetron was delivered to the inside of cells. Alosetron-sensitive sites are, thus, accessible only on the ectodomain of the plasmalemma. The effect of alosetron was reversible, but not surmountable. Although nicotine (100 microM) mimicked the 5-HT-induced inward current, the response was antagonized by hexamethonium (100 microM), but not by alosetron, implying its potential to be a selective 5-HT(3) antagonist. Hexamethonium did not affect responses to 5-HT. Most neurons in the cultures were 5-HT-immunoreactive and immunostained with an antibody raised against 5-HT(3) receptors. The 5-HT-selective uptake inhibitor, fluoxetine (30 microM), gradually reduced the amplitude of the current induced by 5-HT; the residual response was abolished by alosetron (0.2 microM). The effect of fluoxetine could have been caused by either the desensitization of 5-HT(3) receptors or by a nonspecific 5-HT(3) antagonistic effect of fluoxetine. It is concluded that alosetron is a potent and noncompetitive 5-HT(3) antagonist on myenteric neurons.


Assuntos
Intestinos/fisiologia , Neurônios/fisiologia , Receptores de Serotonina/metabolismo , Acetilcolina/metabolismo , Animais , Animais Recém-Nascidos , Carbolinas/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Eletrofisiologia , Gânglios/efeitos dos fármacos , Gânglios/fisiologia , Cobaias , Intestinos/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Receptores de Serotonina/efeitos dos fármacos , Receptores 5-HT3 de Serotonina , Serotonina/análogos & derivados , Serotonina/metabolismo , Serotonina/farmacologia , Antagonistas da Serotonina/farmacologia
10.
J Comp Neurol ; 409(1): 85-104, 1999 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-10363713

RESUMO

Immunocytochemistry was employed to locate calcium (Ca2+) channel proteins in the enteric nervous system (ENS) of the rat and guinea pig. Anti-peptide antibodies that specifically recognize the alpha1 subunits of class A (P/Q-type), B (N-type), C and D (L-type) Ca2+ channels were utilized. Alpha1B channel-like immunoreactivity was abundant in both enteric plexuses, the mucosa, and circular and longitudinal muscle layers. Immunoreactivity was predominantly found in cholinergic varicosities, supporting a role for Ca2+ channels, which contain the alpha1B subunit, in acetylcholine release. Immunoreactivity was also associated with the cell soma of calbindin-immunoreactive submucosal and myenteric neurons, cells that have been proposed to be intrinsic primary afferent neurons. Alpha1C channel-like immunoreactivity was distributed diffusely in the cell membrane of a large subset of neuronal cell bodies and processes, whereas alpha1D was found mainly in the cell soma and proximal dendrites ofvasoactive intestinal polypeptide-immunoreactive neurons in the guinea pig gut. Alpha1A channel-like immunoreactivity was found in a small subset of cell bodies and processes in the rat ENS. The differential localization of the alpha1 subunits of Ca2+ channels in the ENS implies that they serve distinct roles in neuronal excitation and signaling within the bowel. The presence of alpha1B channel-like immunoreactivity in putative intrinsic primary afferent neurons suggested that class B Ca2+ channels play a role in enteric sensory neurotransmission; therefore, we determined the effects of the N-type Ca2+ channel blocker, omega-conotoxin GVIA (omega-CTx GVIA), on the reflex-evoked activity of enteric neurons. Demonstrating the phosphorylation of cyclic AMP (cAMP)-responsive element-binding protein (pCREB) identified neurons that became active in response to distension. Distension elicited hexamethonium-resistant pCREB immunoreactivity in calbindin-immunoreactive neurons in each plexus; however, in preparations stimulated in the presence of omega-CTx GVIA, pCREB immunoreactivity was found only in calbindin-immunoreactive neurons in the submucosal plexus and not in myenteric ganglia. These data confirm that intrinsic primary afferent neurons are located in the submucosal plexus and that N-type Ca2+ channels play a role in sensory neurotransmission.


Assuntos
Canais de Cálcio/metabolismo , Intestinos/inervação , Neurônios Aferentes/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Feminino , Imuno-Histoquímica , Sistema Nervoso/citologia , Sistema Nervoso/metabolismo , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Estimulação Física , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Reflexo/fisiologia , Distribuição Tecidual/fisiologia , ômega-Conotoxina GVIA
11.
Neuron ; 24(4): 941-51, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10624957

RESUMO

Orexin (hypocretin) appears to play a role in the regulation of energy balances. Previous reports have indicated that orexin-containing neurons are found only in the lateral hypothalamic (LH) area. We show that a subset of neurons in the gut which also express leptin receptors display orexin-like immunoreactivity and express functional orexin receptors. Orexin excites secretomotor neurons in the guinea pig submucosal plexus and increases motility. Moreover, fasting upregulates the phosphorylated form of cAMP response element-binding protein (pCREB) in orexin-immunoreactive neurons, indicating a functional response to food status in these cells. Together, these data suggest that orexin in the gut may play an even more intimate role in regulating energy homeostasis than it does in the CNS.


Assuntos
Proteínas de Transporte/biossíntese , Proteínas de Transporte/fisiologia , Sistema Digestório/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Neuropeptídeos/biossíntese , Neuropeptídeos/fisiologia , Animais , Axônios/metabolismo , Proteínas de Transporte/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Sistema Digestório/inervação , Fenômenos Fisiológicos do Sistema Digestório , Sistema Nervoso Entérico/fisiologia , Células Enteroendócrinas/metabolismo , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Fome/fisiologia , Imuno-Histoquímica , Intestino Delgado/inervação , Intestino Delgado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neuropeptídeos/farmacologia , Receptores de Orexina , Orexinas , Precursores de Proteínas/biossíntese , Precursores de Proteínas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G , Receptores de Neuropeptídeos/biossíntese , Receptores de Neuropeptídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinapses/metabolismo , Sinapses/fisiologia
12.
J Comp Neurol ; 390(4): 497-514, 1998 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-9450532

RESUMO

Although nicotinic acetylcholine receptors (nAChRs) are known to be present on neural elements in both the bowel and the pancreas, the precise location of these receptors has not previously been determined. Immunocytochemistry, by using a rat monoclonal antibody (mAb35), which recognizes alpha-bungarotoxin (alpha-Bgt)-insensitive nAChRs, and a polyclonal antibody raised against the alpha-Bgt-sensitive receptor subunit, alpha7, was used to locate receptor protein in guinea pig gut and pancreas. mAb35-receptor (mAb35-R) immunoreactivity was abundant in both enteric plexuses, enterochromaffin cells, and pancreatic ganglia. Immunostaining was associated with the cell membrane, and clusters of mAb35-R were observed on cell somas and dendrites. Receptor immunoreactivity was also observed on terminals and axons, suggesting that a subset of nAChRs is presynaptic. Internal sites of mAb35-R were observed in permeabilized ganglia. Cells expressing the receptors were closely associated with ChAT-immunoreactive nerve fibers. In addition, the majority of ChAT-positive neurons expressed both cell surface and internal stores of mAb35-R. In the bowel, clusters of mAb35-R were present on the soma and dendrites of Dogiel type I motorneurons and secretomotor neurons. Receptors were detected at the plasma membrane of calbindin-immunoreactive myenteric neurons. In contrast, calbindin-immunoreactive submucosal neurons did not express cell surface mAb35-R, supporting the idea that they are sensory neurons. A subset of enteric neurons expressed both mAb35-R and glutamate receptor (GluR1) immunoreactivity. In the pancreas, mAb35-R immunoreactivity was only observed in ganglia. Alpha7-immunoreactivity was found on both enteric cell bodies and nerve fibers. Based on these results, it appears that visceral nAChRs are composed of at least four subunits and that both pre- and postsynaptic nAChRs are present in the gut and pancreas.


Assuntos
Cobaias/metabolismo , Intestinos/química , Pâncreas/química , Receptores Nicotínicos/análise , Animais , Anticorpos Monoclonais , Colina O-Acetiltransferase/análise , Sistema Nervoso Entérico/química , Imuno-Histoquímica , Intestinos/citologia , Masculino , Fibras Nervosas/química , Neurônios/química , Receptores de Glutamato/análise
13.
J Neurosci ; 17(22): 8804-16, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9348349

RESUMO

Glutamate, the major excitatory neurotransmitter in the CNS, is also an excitatory neurotransmitter in the enteric nervous system (ENS). We tested the hypothesis that excessive exposure to glutamate, or related agonists, produces neurotoxicity in enteric neurons. Prolonged stimulation of enteric ganglia by glutamate caused necrosis and apoptosis in enteric neurons. Acute and delayed cell deaths were observed. Glutamate neurotoxicity was mimicked by NMDA and blocked by the NMDA antagonist D-2-amino-5-phosphonopentanoate. Excitotoxicity was more pronounced in cultured enteric ganglia than in intact preparations of bowel, presumably because of a reduction in glutamate uptake. Glutamate-immunoreactive neurons were found in cultured myenteric ganglia, and a subset of enteric neurons expressed NMDA (NR1, NR2A/B), AMPA (GluR1, GluR2/3), and kainate (GluR5/6/7) receptor subunits. Glutamate receptors were clustered on enteric neurites. Stimulation of cultured enteric neurons by kainic acid led to the swelling of somas and the growth of varicosities ("blebs") on neurites. Blebs formed close to neurite intersections and were enriched in mitochondria, as revealed by rhodamine 123 staining. Kainic acid also produced a loss of mitochondrial membrane potential in cultured enteric neurons at sites where blebs tended to form. These observations demonstrate, for the first time, excitotoxicity in the ENS and suggest that overactivation of enteric glutamate receptors may contribute to the intestinal damage produced by anoxia, ischemia, and excitotoxins present in food.


Assuntos
Sistema Nervoso Entérico/efeitos dos fármacos , Sistema Nervoso Entérico/patologia , Neurotoxinas/farmacologia , Animais , Especificidade de Anticorpos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Células Cultivadas , Agonistas de Aminoácidos Excitatórios/farmacologia , Gânglios/citologia , Ácido Glutâmico/farmacologia , Ácido Glutâmico/fisiologia , Cobaias , Ácido Caínico/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mitocôndrias/fisiologia , N-Metilaspartato/farmacologia , Necrose , Neuritos/efeitos dos fármacos , Neuritos/patologia , Neurônios/química , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Receptores de Ácido Caínico/análise , Receptores de Ácido Caínico/imunologia , Receptores de N-Metil-D-Aspartato/análise , Receptores de N-Metil-D-Aspartato/imunologia
14.
J Neurosci ; 17(12): 4764-84, 1997 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9169536

RESUMO

We tested the hypothesis that glutamate, the major excitatory neurotransmitter of the CNS, is also an excitatory neurotransmitter in the enteric nervous system (ENS). Glutamate immunoreactivity was found in cholinergic enteric neurons, many of which were identified as sensory by their co-storage of substance P and/or calbindin. Glutamate immunoreactivity was concentrated in terminal varicosities with a majority of small clear synaptic vesicles. The immunoreactivities of both AMPA and NMDA receptor subunits were also detected on neurons in both submucosal and myenteric plexuses. The immunoreactivity of the EAAC1 neuronal glutamate transporter was widespread in both plexuses. Glutamate evoked depolarizing responses in myenteric neurons that had fast and slow components. The fast component was mimicked by AMPA, and the slow component was mimicked by NMDA. The fast component and the response to AMPA mimicked fast EPSPs evoked in 2/AH neurons; moreover, fast EPSPs as well as fast glutamate and AMPA responses were blocked by selective AMPA antagonists and potentiated by the glutamate uptake inhibitor L-(-)-threo-3-hydroxyaspartic acid. These observations demonstrate, for the first time, the presence of glutamatergic neurons and glutamate-mediated neurotransmission in the ENS.


Assuntos
Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/análise , Ácido Glutâmico/farmacologia , Mucosa Intestinal/inervação , Intestino Delgado/inervação , Plexo Mientérico/fisiologia , Neurônios/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Anticorpos , Benzotiadiazinas/farmacologia , Cálcio/farmacologia , Potenciais Evocados/efeitos dos fármacos , Cobaias , Hexametônio/farmacologia , Íleo , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Microscopia Imunoeletrônica , Músculo Liso/inervação , Plexo Mientérico/citologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Nicotina/farmacologia , Organelas/fisiologia , Organelas/ultraestrutura , Ratos , Receptores de AMPA/análise , Receptores de N-Metil-D-Aspartato/análise , Transmissão Sináptica/efeitos dos fármacos , Vesículas Sinápticas/ultraestrutura , Tetrodotoxina/farmacologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
15.
Am J Physiol ; 273(6): G1273-89, 1997 12.
Artigo em Inglês | MEDLINE | ID: mdl-9435552

RESUMO

The morphology, neurochemistry, and electrical properties of guinea pig pancreatic neurons were determined. The majority of neurons expressed choline acetyltransferase (ChAT) immunoreactivity; however, ChAT-negative neurons were also found. Both cholinergic and noncholinergic neurons expressed nitric oxide synthase (NOS) immunoreactivity. Three types of pancreatic neurons were distinguished. Phasic neurons fired action potentials (APs) at the onset of depolarizing current pulse, tonic neurons spiked throughout the duration of a suprathreshold depolarizing pulse, and APs could not be generated in nonspiking neurons, even though they did receive synaptic input. APs were tetrodotoxin sensitive, and all types of neurons received fast and slow excitatory postsynaptic potentials (EPSPs). Fast EPSPs had cholinergic and noncholinergic components. The majority of pancreatic neurons appeared to innervate the acini. NOS- and/or neuropeptide Y-immunoreactive phasic and tonic neurons were found. Microejection of 5-hydroxytryptamine (5-HT) caused a slow depolarization that was inhibited by the 5-HT1P antagonist N-acetyl-5-hydroxytryptophyl-5-hydroxytryptophan amide and mimicked by the 5-HT1P agonist 6-hydroxyindalpine. A pancreatic 5-HT transporter was located, and inhibition of 5-HT uptake by fluoxetine blocked slow EPSPs in 5-HT-responsive neurons by receptor desensitization.


Assuntos
Neurônios/citologia , Neurônios/fisiologia , Pâncreas/inervação , Serotonina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Biotina/análogos & derivados , Colina O-Acetiltransferase/análise , Colina O-Acetiltransferase/biossíntese , Potenciais Pós-Sinápticos Excitadores/fisiologia , Fluoxetina/farmacologia , Cobaias , Hexametônio/farmacologia , Masculino , Potenciais da Membrana , Microeletrodos , Microscopia Confocal , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase/biossíntese , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Sinapses/fisiologia , Tetrodotoxina/farmacologia
16.
J Comp Neurol ; 371(2): 270-86, 1996 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-8835732

RESUMO

To identify neurons participating in enteric and enteropancreatic reflexes, we validated the use of the activity-dependent markers FM1-43 and FM2-10 as "on-line" probes for the visualization of activated guinea pig enteric and pancreatic neurons. FM1-43 or FM2-10 labeling of neuronal perikarya and processes was induced by KCl (70 mM), veratridine (1.0 microM), intracellular injection of depolarizing current pulses, stimulation of afferent inputs, evoking reflexes (by inflating an intraluminal balloon, blowing puffs of N2 at, or applying glucose to, the villous surface of the duodenum), or injury; labeling was prevented by tetrodotoxin (0.5 microM). Intracellular recording and injection of Neurobiotin confirmed that FM1-43 labeled neurons that spike, but not those that exhibit only fast excitatory postsynaptic potentials. Perikarya did not label if axonal transport was blocked by colchicine. When pulses of N2 or glucose were directed at duodenal villi in vitro, labeling by FM1-43 or FM2-10 was observed in myenteric and pancreatic neurons, as well as in subsets of cells in pancreatic islets and intestinal crypts. Hexamethonium blocked the spread of label via nicotinic synapses and thus enabled primary afferent neurons to be located. Balloon distension elicited hexamethonium-resistant labeling of epithelial cells, interstitial cells, and Dogiel type II neurons in each plexus; however, in preparations stimulated with pulses of N2 or glucose, hexamethonium-resistant labeling of neurons occurred only in the submucosal plexus and not in myenteric ganglia. These observations suggest that primary afferent neurons responsible for mucosal pressure- or glucose-induced enteric and enteropancreatic reflexes are submucosal, whereas myenteric afferent neurons become activated only when the wall of the bowel is distended. The data are compatible with the possibility that primary afferent neurons are activated by a signaling molecule released from intestinal epithelial cells.


Assuntos
Sistema Nervoso Entérico/fisiologia , Neurônios Aferentes/química , Pâncreas/inervação , Reflexo/fisiologia , Animais , Axônios/efeitos dos fármacos , Colchicina/farmacologia , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/efeitos dos fármacos , Corantes Fluorescentes , Cobaias , Hibridização In Situ , Masculino , Potenciais da Membrana/fisiologia , Terminações Nervosas/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Compostos de Piridínio , Compostos de Amônio Quaternário , Reflexo/efeitos dos fármacos , Estimulação Química
17.
J Comp Neurol ; 364(3): 439-455, 1996 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-8820876

RESUMO

Although serotonin (5-HT)1A receptors are known to be present on neural elements in both the bowel and the pancreas, the precise location of these receptors has not previously been determined. Earlier investigations have suggested that 5-HT1A receptors are synthesized in enteric, but not pancreatic ganglia, and that they mediate pre-and postjunctional inhibition. Wholemount in situ hybridization was used to identify cells that contain mRNA encoding 5-HT1A receptors, and immunocytochemistry was employed to locate receptor protein. mRNA encoding 5-HT1A receptors was found in the majority of neurons in both submucosal and myenteric plexuses. 5-HT1A immunoreactivity, however, was abundant only on the surfaces of a limited subset of nerve cell bodies and processes. 5-HT-immunoreactive axons were found in close proximity to sites of 5-HT1A immunoreactivity. Myenteric, but not submucosal calbindin-immunoreactive neurons (with Dogiel type II morphology) were surrounded by rings of 5-HT1A immunoreactivity. The cytoplasm of the cell bodies and dendrites of a small subset of Dogiel type I neurons was also intensely 5-HT1A immunoreactive. Most of the Dogiel type I 5-HT1A-immunoreactive myenteric neurons, and some of the type II neurons that were ringed by 5-HT1A immunoreactivity became doubly labeled following injections of the retrograde tracer, FluoroGold (FG), into the submucosal plexus. 5-HT1A-immunoreactive neurons in distant submucosal ganglia also became labeled by retrograde transport of FG. None of the 5-HT1A-immunoreactive cells were labeled by the intraluminal administration of the beta-subunit of cholera toxin, a marker for vasoactive intestinal peptide-containing secretomotor neurons. These observations suggest that some of the myenteric 5-HT1A-immunoreactive neurons project to submucosal ganglia and that the submucosal 5-HT1A-immunoreactive cells are interneurons. In addition to neurons, a subset of 5-HT-containing enterochromaffin cells expressed 5-HT1A immunoreactivity, which was co-localized with 5-HT in secretory granules. In the pancreas, 5-HT1A immunoreactivity was observed in ganglia, acinar nerves, and glucagonimmunoreactive islet cells. Serotonergic enteropancreatic axons have been found to terminate in close proximity to each of these structures, which may thus be the targets of this innervation. The abundance of 5-HT1A receptor immunoreactivity on nerves of the gut and pancreas suggests that drugs designed to interact with these receptors may have unanticipated visceral actions.


Assuntos
Sistema Nervoso Entérico/metabolismo , Intestinos/inervação , Pâncreas/inervação , Receptores de Serotonina/biossíntese , Estilbamidinas , Animais , Calbindinas , Feminino , Corantes Fluorescentes , Cobaias , Imuno-Histoquímica , Hibridização In Situ , Mucosa Intestinal/metabolismo , Masculino , Neurônios Motores/fisiologia , Plexo Mientérico/citologia , Plexo Mientérico/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/metabolismo , RNA Mensageiro/biossíntese , Ratos , Proteína G de Ligação ao Cálcio S100/metabolismo
18.
Am J Physiol ; 268(2 Pt 1): G339-45, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7532365

RESUMO

Enteric cholinergic and serotonergic neurons innervate pancreatic ganglia. Enteropancreatic cholinergic neurons are secretomotor, bu the function of the serotonergic cells is unknown and was investigated. Postganglionic cholinergic nerve-mediated amylase secretion was evoked by veratridine in isolated pancreatic lobules. This concentration-dependent response was inhibited by tetrodotoxin (1.0 microM), atropine (5.0 microM), 5-hydroxytryptamine (5-HT; 5.0 microM), 5-hydroxyindalpine (5-OHIP; 10.0 microM; a 5-HT1P agonist), and 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT; 0.1 microM), but not by hexamethonium (100.0 microM), 2-methyl-5-HT (10 microM), or 5-carboxyamidotryptamine (10 microM). The effects of 5-HT and 5-OHIP were blocked by the 5-HT1P antagonist N-acetyl-5-hydroxytryptophyl-5-hydroxytryptophan amide (5-HTP-DP; 100.0 microM). Carbachol (5.0 microM)-evoked secretion was not affected by 5-HT or 5-OHIP. Veratridine induced c-fos expression in pancreatic neurons and acinar cells. This expression was inhibited by tetrodotoxin, 5-HT, and 5-OHIP. These observations suggest that the serotonergic enteropancreatic innervation inhibits pancreatic secretion via presynaptic receptors on cholinergic nerves. Although the data are consistent with the hypothesis that the inhibitory receptor is a 5-HT1P site, positive identification awaits further study.


Assuntos
Amilases/antagonistas & inibidores , Gânglios/fisiologia , Pâncreas/inervação , Pâncreas/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Serotonina/farmacologia , Amilases/metabolismo , Animais , Feminino , Técnicas In Vitro , Neurônios/metabolismo , Pâncreas/citologia , Terminações Pré-Sinápticas/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Veratridina/farmacologia
19.
Brain Res Dev Brain Res ; 84(1): 26-38, 1995 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-7720214

RESUMO

Pancreatic ganglia are formed by neural crest-derived precursors, are innervated by enteric neurons, and contain neuropeptides. In addition, the enzyme NADPH-diaphorase is located in a subset of enteric and pancreatic neurons. The expression of neural markers (GAP-43 and NC-1), neurotransmitter-related markers (including neuropeptide Y (NPY), vasoactive intestinal peptide (VIP), gastrin-releasing peptide (GRP), galanin (GAL), dopamine beta hydroxylase (DBH), substance P (SP), calcitonin gene-related peptide (CGRP)), and NADPH-diaphorase was studied in the fetal and neonatal rat gut and pancreas (E12-P28) in situ and in vitro. NC-1, GAP-43 and DBH-immunoreactive cells were found in the primordial stomach on day E12, and in the pancreas on day E13, along with NPY in endocrine cells. Pancreatic NPY-immunoreactive neurons were detected by day E18. CGRP was seen in the foregut at day E12 but not in the pancreas until day E14. Other neuropeptides (SP, GAL, GRP and VIP) all appeared in the foregut earlier than in the pancreas. NADPH-diaphorase activity was first found in situ in foregut neurons on day E13, and in the pancreas on day E14, but seen in explants a day earlier. These observations show that development of neurons occurs earlier in the gut than in the pancreas, and that NADPH-diaphorase activity appears earlier than the immunoreactivities of the neuropeptides.


Assuntos
Sistema Nervoso Entérico/fisiologia , NADPH Desidrogenase/fisiologia , Neuropeptídeo Y/farmacologia , Neuropeptídeos/fisiologia , Animais , Biologia do Desenvolvimento , Feminino , Imuno-Histoquímica , Neurônios/fisiologia , Pâncreas/inervação , Pâncreas/fisiologia , Ratos , Ratos Sprague-Dawley , Estômago/imunologia
20.
J Comp Neurol ; 342(1): 115-30, 1994 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-7515904

RESUMO

Pancreatic ganglia are innervated by neurons in the gut and are formed by precursor cells that migrate into the pancreas from the bowel. The innervation of the pancreas, therefore, may be considered an extension of the enteric nervous system. NADPH-diaphorase is present in a subset of enteric neurons. We investigated the presence of NADPH-diaphorase in the enteropancreatic innervation, the contribution of extrinsic nerves to the NADPH-diaphorase-containing fibers of the gut and pancreas, and the coincident expression of NADPH-diaphorase NADPH-diaphorase in intrinsic neurons of these organs with neuropeptides. The possible role of nitric oxide in the neural regulation of pancreatic secretion was studied in isolated pancreatic lobules. Neuronal perikarya with NADPH-diaphorase activity were found in both Dogiel type I and type II neurons of the myenteric plexus of the stomach and duodenum. All galanin (GAL)-immunoreactive neurons and a small subset of vasoactive intestinal polypeptide (VIP)- and neuropeptide Y (NPY)-immunoreactive neurons contained NADPH-diaphorase activity. NADPH-diaphorase activity was also found in a subset of VIP and NPY-immunoreactive pancreatic neurons. Retrograde tracing with FluoroGold established that NADPH-diaphorase-containing neurons in the bowel project to the pancreas. NADPH-diaphorase-containing fibers were also found to be provided to both organs by neurons in dorsal root ganglia. Secretion of amylase was evoked by L-arginine. This effect was prevented by N(G)-nitro-L-arginine (L-NNA), which also inhibited VIP-stimulated secretion of amylase; however, L-NNA had no effect on amylase secretion stimulated by carbachol. These results provide support for the hypothesis that nitric oxide plays a role in the neural regulation of pancreatic secretion.


Assuntos
NADPH Desidrogenase/metabolismo , Neuropeptídeos/biossíntese , Pâncreas/inervação , Estilbamidinas , Amilases/metabolismo , Animais , Arginina/metabolismo , Transporte Axonal/fisiologia , Sistema Nervoso Entérico/enzimologia , Feminino , Corantes Fluorescentes , Imuno-Histoquímica , Fibras Nervosas/enzimologia , Vias Neurais/citologia , Vias Neurais/fisiologia , Óxido Nítrico/metabolismo , Pâncreas/enzimologia , Pâncreas/fisiologia , Ratos , Peptídeo Intestinal Vasoativo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...