Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Gastroenterology ; 162(7): 1962-1974.e6, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35167867

RESUMO

BACKGROUND & AIMS: Increased colonic serotonin (5-HT) level and decreased serotonin reuptake transporter (SERT) expression in irritable bowel syndrome (IBS) may contribute to diarrhea and visceral hypersensitivity. We investigated whether mucosal SERT is modulated by gut microbiota via a mast cell-prostaglandin E2 (PGE2) pathway. METHODS: C57Bl/6 mice received intracolonic infusion of fecal supernatant (FS) from healthy controls or patients with diarrhea-predominant irritable bowel syndrome (IBS-D). The role of mast cells was studied in mast cell-deficient mice. Colonic organoids and/or mast cells were used for in vitro experiments. SERT expression was measured by quantitative polymerase chain reaction and Western blot. Visceromotor responses to colorectal distension and colonic transit were assessed. RESULTS: Intracolonic infusion of IBS-D FS in mice caused an increase in mucosal 5-HT compared with healthy control FS, accompanied by ∼50% reduction in SERT expression. Mast cell stabilizers, cyclooxygenase-2 inhibitors, and PGE2 receptor antagonist prevented SERT downregulation. Intracolonic infusion of IBS-D FS failed to reduce SERT expression in mast cell-deficient (W/Wv) mice. This response was restored by mast cell reconstitution. The downregulation of SERT expression evoked by IBS FS was prevented by lipopolysaccharide (LPS) antagonist LPS from Rhodobacter sphaeroides and a bacterial trypsin inhibitor. In vitro LPS treatment caused increased cyclooxygenase-2 expression and PGE2 release from cultured mouse mast cells. Intracolonic infusion of IBS-D FS in mice reduced colonic transit, increased fecal water content, and increased visceromotor responses to colorectal distension. Ondansetron prevented these changes. CONCLUSIONS: Fecal LPS acting in concert with trypsin in patients with IBS-D stimulates mucosal mast cells to release PGE2, which downregulates mucosal SERT, resulting in increased mucosal 5-HT. This may contribute to diarrhea and abdominal pain common in IBS.


Assuntos
Neoplasias Colorretais , Microbioma Gastrointestinal , Síndrome do Intestino Irritável , Animais , Neoplasias Colorretais/metabolismo , Diarreia/metabolismo , Dinoprostona/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Síndrome do Intestino Irritável/complicações , Lipopolissacarídeos , Mastócitos/metabolismo , Camundongos , Serotonina/metabolismo , Serotonina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo
2.
Gastroenterology ; 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-35934059

RESUMO

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal

3.
Gastroenterology ; 158(8): 2195-2207.e6, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32084424

RESUMO

BACKGROUND AND AIMS: Visceral hypersensitivity is common in patients with irritable bowel syndrome (IBS). We investigated whether inflammatory molecules, such as histamine and proteases, activate prostaglandin-endoperoxide synthase 2 (also called COX2) to increase the synthesis of prostaglandin E2 (PGE2) by mast cells, which activates the receptor PTGER2 (also called EP2) in the dorsal root ganglia to promote visceral hypersensitivity. METHODS: We used an enzyme-linked immunosorbent assay to measure levels of spontaneous release of molecules from mast cells in colonic mucosa from patients with IBS with diarrhea (IBS-D; 18 women and 5 men; aged 28-60 years), healthy individuals (controls, n = 24), mice, and rats. We measured visceromotor responses to colorectal distension in rodents after intracolonic administration of colon biopsy supernatants, histamine, PGE2, a small interfering RNA against EP2, or an agonist of F2R like trypsin receptor 1 (F2RL1, also called protease-activated receptor 2 [PAR2]). We investigated the role of COX2, produced by mast cells, in mediation of visceral hypersensitivity using mice with the Y385F substitution in Ptgs2 (Ptgs2Y385F mice), mast cell-deficient (W/WV) mice, and W/WV mice given injections of mast cells derived from wild-type or Ptgs2Y385F mice. RESULTS: Colon biopsies from patients with IBS-D had increased levels of PGE2, based on enzyme-linked immunosorbent assay, and COX2 messenger RNA and protein, compared with control biopsies. Immunohistochemistry showed that most of the COX2 was in mast cells. Intracolonic infusions of rats with IBS-D biopsy supernatants generated a 3- to 4-fold increase in visceromotor responses to colorectal distension; this was associated with significant increases in PGE2, histamine, and tryptase in the colonic mucosa. These increases were prevented by a mast cell stabilizer, COX2 inhibitor, or knockdown of EP2. Intracolonic administration of supernatants from biopsies of patients with IBS-D failed to induce visceral hypersensitivity or increase the level of PGE2 in W/WV and Ptgs2Y385Fmice. Reconstitution of mast cells in W/WV mice restored the visceral hypersensitivity response. CONCLUSIONS: Abnormal synthesis of PGE2 by colonic mast cells appears to induce visceral hypersensitivity in patients with IBS-D.


Assuntos
Colo/metabolismo , Dinoprostona/metabolismo , Mucosa Intestinal/metabolismo , Síndrome do Intestino Irritável/complicações , Mastócitos/metabolismo , Extratos de Tecidos/metabolismo , Dor Abdominal/etiologia , Dor Abdominal/metabolismo , Dor Abdominal/fisiopatologia , Adulto , Animais , Estudos de Casos e Controles , Células Cultivadas , Colo/inervação , Ciclo-Oxigenase 2/deficiência , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Diarreia/etiologia , Diarreia/metabolismo , Diarreia/fisiopatologia , Feminino , Humanos , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Mucosa Intestinal/inervação , Síndrome do Intestino Irritável/metabolismo , Síndrome do Intestino Irritável/fisiopatologia , Masculino , Mastócitos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Ratos Wistar , Células Receptoras Sensoriais/metabolismo , Extratos de Tecidos/administração & dosagem
4.
Am J Respir Cell Mol Biol ; 62(2): 191-203, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31486669

RESUMO

The differentiation of fibroblasts into myofibroblasts is critical for the development of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF). Previously, we demonstrated that fibroblasts from patients with IPF exhibit changes in DNA methylation across the genome that contribute to a profibrotic phenotype. One of the top differentially methylated genes identified in our previous study was KCNMB1, which codes for the ß subunit of the large-conductance potassium (BK, also known as MaxiK or KCa1.1) channel. Here, we examined how the expression of KCNMB1 differed between IPF fibroblasts and normal cells, and how BK channels affected myofibroblast differentiation. Fibroblasts from patients with IPF exhibited increased expression of KCNMB1, which corresponded to increased DNA methylation within the gene body. Patch-clamp experiments demonstrated that IPF fibroblasts had increased BK channel activity. Knockdown of KCNMB1 attenuated the ability of fibroblasts to contract collagen gels, and this was associated with a loss of α-smooth muscle actin (SMA) expression. Pharmacologic activation of BK channels stimulated α-SMA expression, whereas BK channel inhibitors blocked the upregulation of α-SMA. The ability of BK channels to enhance α-SMA expression was dependent on intracellular calcium, as activation of BK channels resulted in increased levels of intracellular calcium and the effects of BK agonists were abolished when calcium was removed. Together, our findings demonstrate that epigenetic upregulation of KCNMB1 contributes to increased BK channel activity in IPF fibroblasts, and identify a newfound role for BK channels in myofibroblast differentiation.


Assuntos
Fibrose Pulmonar Idiopática/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Miofibroblastos/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Metilação de DNA/fisiologia , Fibroblastos/metabolismo , Humanos , Fibrose Pulmonar Idiopática/genética , Pulmão/metabolismo
6.
Gastroenterology ; 151(5): 910-922.e7, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27475306

RESUMO

BACKGROUND & AIMS: Patients with diabetes have defects in the vagal afferent pathway that result in abnormal gastrointestinal function. We investigated whether selective increased activation of the 2-pore domain potassium channel TRESK (2-pore-domain weak inward-rectifying potassium channel-related spinal cord potassium channel) contributes to nodose ganglia (NG) malfunction, disrupting gastrointestinal function in diabetic rats. METHODS: We conducted whole-cell current-clamp and single-unit recordings in NG neurons from diabetes-prone BioBreeding/Worcester rats and streptozotocin-induced diabetic (STZ-D) rats and compared them with control rats. NG neurons in rats or cultured NG neurons were exposed to pharmacologic antagonists and/or transfected with short hairpin or small interfering RNAs that reduced expression of TRESK. We then made electrophysiologic recordings and studied gastrointestinal functions. RESULTS: We observed reduced input resistance, hyperpolarized membrane potential, and increased current threshold to elicit action potentiation in NG neurons of STZ-D rats compared with controls. NG neuron excitability was similarly altered in diabetes-prone rats. In vivo single-unit NG neuronal discharges in response to 30 and 60 pmol cholecystokinin octapeptide were significantly lower in STZ-D rats compared with controls. Reducing expression of the TRESK K+ channel restored NG excitability in vitro and in vivo, as well as cholecystokinin 8-stimulated secretion of pancreatic enzymes and secretin-induced gastrointestinal motility, which are mediated by vago-vagal reflexes. These abnormalities resulted from increased intracellular Ca2+ in the NG, activating calcineurin, which, in turn, bound to an nuclear factor of activated T cell-like docking site on the TRESK protein, resulting in neuronal membrane hyperpolarization. CONCLUSIONS: In 2 rate models of diabetes, we found that activation of the TRESK K+ channel reduced NG excitability and disrupted gastrointestinal functions.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Motilidade Gastrointestinal/fisiologia , Gânglio Nodoso/fisiopatologia , Canais de Potássio/metabolismo , Animais , Biomarcadores/metabolismo , Diabetes Mellitus Experimental/metabolismo , Masculino , Potenciais da Membrana , Técnicas de Patch-Clamp , Ratos , Ratos Endogâmicos BB , Reflexo
7.
Medicina (Kaunas) ; 53(2): 73-84, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28454890

RESUMO

Vagal sensory neurons mediate the vago-vagal reflex which, in turn, regulates a wide array of gastrointestinal functions including esophageal motility, gastric accommodation and pancreatic enzyme secretion. These neurons also transmit sensory information from the gut to the central nervous system, which then mediates the sensations of nausea, fullness and satiety. Recent research indicates that vagal afferent neurons process non-uniform properties and a significant degree of plasticity. These properties are important to ensure that vagally regulated gastrointestinal functions respond rapidly and appropriately to various intrinsic and extrinsic factors. Similar plastic changes in the vagus also occur in pathophysiological conditions, such as obesity and diabetes, resulting in abnormal gastrointestinal functions. A clear understanding of the mechanisms which mediate these events may provide novel therapeutic targets for the treatment of gastrointestinal disorders due to vago-vagal pathway malfunctions.


Assuntos
Vias Aferentes/fisiologia , Trato Gastrointestinal/inervação , Plasticidade Neuronal/fisiologia , Células Receptoras Sensoriais/fisiologia , Nervo Vago/fisiologia , Nervo Vago/fisiopatologia , Animais , Diabetes Mellitus/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Humanos , Hormônios Peptídicos/fisiologia
8.
J Physiol ; 593(17): 3973-89, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26174421

RESUMO

Ghrelin, a hunger signalling peptide derived from the peripheral tissues, overcomes the satiety signals evoked by anorexigenic molecules, such as cholecystokinin (CCK) and leptin, to stimulate feeding. Using in vivo and in vitro electrophysiological techniques, we show that ghrelin hyperpolarizes neurons and inhibits currents evoked by leptin and CCK-8. Administering a KATP channel antagonist or silencing Kir6.2, a major subunit of the KATP channel, abolished ghrelin inhibition. The inhibitory actions of ghrelin were also abolished by treating the vagal ganglia neurons with pertussis toxin, as well as phosphatidylinositol 3-kinase (PI3K) or extracellular signal-regulated kinase 1 and 2 (Erk1/2) small interfering RNA. Feeding experiments showed that silencing Kir6.2 in the vagal ganglia abolished the orexigenic actions of ghrelin. These data indicate that ghrelin modulates vagal ganglia neuron excitability by activating KATP conductance via the growth hormone secretagogue receptor subtype 1a-Gαi -PI3K-Erk1/2-KATP pathway. This provides a mechanism to explain the actions of ghrelin with respect to overcoming anorexigenic signals that act via the vagal afferent pathways. Ghrelin is the only known hunger signal derived from the peripheral tissues. Ghrelin overcomes the satiety signals evoked by anorexigenic molecules, such as cholecystokinin (CCK) and leptin, to stimulate feeding. The mechanisms by which ghrelin reduces the sensory signals evoked by anorexigenic hormones, which act via the vagus nerve to stimulate feeding, are unknown. Patch clamp recordings of isolated rat vagal neurons show that ghrelin hyperpolarizes neurons by activating K(+) conductance. Administering a KATP channel antagonist or silencing Kir6.2, a major subunit of the KATP channel, abolished ghrelin inhibition in vitro and in vivo. Patch clamp studies show that ghrelin inhibits currents evoked by leptin and CCK-8, which operate through independent ionic channels. The inhibitory actions of ghrelin were abolished by treating the vagal ganglia neurons with pertussis toxin, as well as phosphatidylinositol 3-kinase (PI3K) or extracellular signal-regulated kinase 1 and 2 (Erk1/2) small interfering RNA. In vivo gene silencing of PI3K and Erk1/2 in the nodose ganglia prevented ghrelin inhibition of leptin- or CCK-8-evoked vagal firing. Feeding experiments showed that silencing Kir6.2 in the vagal ganglia abolished the orexigenic actions of ghrelin. These data indicate that ghrelin modulates vagal ganglia neuron excitability by activating KATP conductance via the growth hormone secretagogue receptor subtype 1a-Gαi -PI3K-Erk1/2-KATP pathway. The resulting hyperpolarization renders the neurons less responsive to signals evoked by anorexigenic hormones. This provides a mechanism to explain the actions of ghrelin with respect to overcoming anorexigenic signals that act via the vagal afferent pathways.


Assuntos
Grelina/farmacologia , Canais KATP/fisiologia , Gânglio Nodoso/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Colecistocinina/farmacologia , Ingestão de Alimentos , Canais KATP/antagonistas & inibidores , Canais KATP/genética , Leptina/farmacologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Gânglio Nodoso/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , RNA Interferente Pequeno/genética , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos
9.
Gut ; 62(10): 1466-74, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22833396

RESUMO

OBJECTIVE: Increased faecal butyrate levels have been reported in irritable bowel syndrome. Rectal instillation of sodium butyrate (NaB) increases visceral sensitivity in rats by an unknown mechanism. We seek to examine the signal transduction pathways responsible for the enhanced neuronal excitability in the dorsal root ganglion (DRG) following NaB enemas and demonstrate that this is responsible for the colonic hypersensitivity reported in this animal model. DESIGN: Colorectal distention (CRD) studies were performed in rats treated with NaB rectal instillation with/without intrathecal or intravenous administration of mitogen-activated protein (MAP) kinase kinase inhibitor U0126. Western blot analysis and immunocytochemistry studies elucidated intracellular signalling pathways that modulate IA. Patch-clamp recordings were performed on isolated DRG neurons treated with NaB, with/without U0126. RESULTS: Visceromotor responses (VMR) were markedly enhanced in NaB-treated rats. Western blot analysis of DRG neurons from NaB-treated rats showed a 2.2-fold increase in phosphorylated ERK1/2 (pEKR1/2) and 1.9-fold increase in phosphorylated voltage-gated potassium channel subunit 4.2 (pKv4.2). Intrathecal or intravenous administration of U0126 reduced VMR to CRD in NaB-treated rats and prevented increases in pERK1/2 and pKv4.2. Patch-clamp recordings of isolated DRG neurons showed that NaB caused a reduction in IA to 48.9%±1.4% of control and an increase in neuronal excitability, accompanied by a twofold increase in pERK1/2 and pKv4.2. Concurrent U0126 administration prevented these changes. CONCLUSIONS: Visceral hypersensitivity induced by colonic NaB treatment is mediated by activation of the MAP kinase-ERK1/2 pathway, which phosphorylates Kv4.2. This results in a reduction in IA and an enhancement of DRG neuronal excitability.


Assuntos
Butiratos/toxicidade , Gânglios Espinais/efeitos dos fármacos , Síndrome do Intestino Irritável/induzido quimicamente , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Butadienos/farmacologia , Células Cultivadas , Colo/efeitos dos fármacos , Colo/inervação , Dilatação , Enema , Ativação Enzimática/efeitos dos fármacos , Gânglios Espinais/enzimologia , Síndrome do Intestino Irritável/enzimologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Nitrilas/farmacologia , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/metabolismo , Dor Visceral/induzido quimicamente , Dor Visceral/enzimologia
10.
Am J Physiol Gastrointest Liver Physiol ; 304(10): G908-16, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23471341

RESUMO

Sensitization of esophageal afferents plays an important role in esophageal nociception, but the mechanism is less clear. Our previous studies demonstrated that mast cell (MC) activation releases the preformed mediators histamine and tryptase, which play important roles in sensitization of esophageal vagal nociceptive C fibers. PGD2 is a lipid mediator released by activated MCs. Whether PGD2 plays a role in this sensitization process has yet to be determined. Expression of the PGD2 DP1 and DP2 receptors in nodose ganglion neurons was determined by immunofluorescence staining, Western blotting, and RT-PCR. Extracellular recordings were performed in ex vivo esophageal-vagal preparations. Action potentials evoked by esophageal distension were compared before and after perfusion of PGD2, DP1 and DP2 receptor agonists, and MC activation, with or without pretreatment with antagonists. The effect of PGD2 on 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI)-labeled esophageal nodose neurons was determined by patch-clamp recording. Our results demonstrate that DP1 and DP2 receptor mRNA and protein were expressed mainly in small- and medium-diameter neurons in nodose ganglia. PGD2 significantly increased esophageal distension-evoked action potential discharges in esophageal nodose C fibers. The DP1 receptor agonist BW 245C mimicked this effect. PGD2 directly sensitized DiI-labeled esophageal nodose neurons by decreasing the action potential threshold. Pretreatment with the DP1 receptor antagonist BW A868C significantly inhibited PGD2 perfusion- or MC activation-induced increases in esophageal distension-evoked action potential discharges in esophageal nodose C fibers. In conclusion, PGD2 plays an important role in MC activation-induced sensitization of esophageal nodose C fibers. This adds a novel mechanism of visceral afferent sensitization.


Assuntos
Esôfago/inervação , Mastócitos/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Prostaglandina D2/farmacologia , Nervo Vago/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Western Blotting , Carbocianinas , Interpretação Estatística de Dados , Esôfago/efeitos dos fármacos , Imunofluorescência , Cobaias , Masculino , Fibras Nervosas/fisiologia , Fibras Nervosas Amielínicas/efeitos dos fármacos , Gânglio Nodoso/citologia , Gânglio Nodoso/efeitos dos fármacos , Técnicas de Patch-Clamp , Estimulação Física , Prostaglandina D2/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Imunológicos/agonistas , Receptores Imunológicos/efeitos dos fármacos , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/efeitos dos fármacos
11.
J Biol Chem ; 286(13): 11707-15, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270124

RESUMO

Research has shown that the synergistic interaction between vagal cholecystokinin-A receptors (CCKARs) and leptin receptors (LRbs) mediates short term satiety. We hypothesize that this synergistic interaction is mediated by cross-talk between signaling cascades used by CCKARs and LRbs, which, in turn, activates closure of K(+) channels, leading to membrane depolarization and neuronal firing. Whole cell patch clamp recordings were performed on isolated rat nodose ganglia neurons. Western immunoblots elucidated the intracellular signaling pathways that modulate leptin/CCK synergism. In addition, STAT3, PI3K, Src, and MAPK genes were silenced by lentiviral infection and transient Lipofectamine transfection of cultured rat nodose ganglia to determine the effect of these molecules on leptin/CCK synergism. Patch clamp studies showed that a combination of leptin and CCK-8 caused a significant increase in membrane input resistance compared with leptin or CCK-8 alone. Silencing the STAT3 gene abolished the synergistic action of leptin/CCK-8 on neuronal firing. Leptin/CCK-8 synergistically stimulated a 7.7-fold increase in phosphorylated STAT3 (pSTAT3), which was inhibited by AG490, C3 transferase, PP2, LY294002, and wortmannin, but not PD98059. Silencing the Src and PI3K genes resulted in a loss of leptin/CCK-stimulated pSTAT3. We conclude that the synergistic interaction between vagal CCKARs and LRbs is mediated by the phosphorylation of STAT3, which, in turn, activates closure of K(+) channels, leading to membrane depolarization and neuronal firing. This involves the interaction between CCK/Src/PI3K cascades and leptin/JAK2/PI3K/STAT3 signaling pathways. Malfunctioning of these signaling molecules may result in eating disorders.


Assuntos
Leptina/farmacologia , Gânglio Nodoso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Resposta de Saciedade/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sincalida/farmacologia , Animais , Inativação Gênica , Ativação do Canal Iônico/efeitos dos fármacos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Leptina/genética , Leptina/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Gânglio Nodoso/citologia , Fosfatidilinositol 3-Quinases/genética , Fosforilação/efeitos dos fármacos , Canais de Potássio/genética , Canais de Potássio/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores da Colecistocinina/genética , Receptores da Colecistocinina/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Fator de Transcrição STAT3/genética , Sincalida/genética , Sincalida/metabolismo , Fatores de Tempo
12.
Am J Physiol Gastrointest Liver Physiol ; 303(9): G1042-51, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22936273

RESUMO

Vagal CCK-A receptors (CCKARs) and leptin receptors (LRbs) interact synergistically to mediate short-term satiety. Cocaine- and amphetamine-regulated transcript (CART) peptide is expressed by vagal afferent neurons. We sought to demonstrate that this neurotransmitter regulates CCK and leptin actions on short-term satiety. We also examined the signal transduction pathways responsible for mediating the CART release from the nodose ganglia (NG). ELISA studies coupled with gene silencing of NG neurons by RNA interference elucidated intracellular signaling pathways responsible for CCK/leptin-stimulated CART release. Feeding studies followed by gene silencing of CART in NG established the role of CART in mediating short-term satiety. Immunohistochemistry was performed on rat NG neurons to confirm colocalization of CCKARs and LRbs; 63% of these neurons contained CART. Coadministration of CCK-8 and leptin caused a 2.2-fold increase in CART release that was inhibited by CCK-OPE, a low-affinity CCKAR antagonist. Transfection of cultured NG neurons with steroid receptor coactivator (SRC) or phosphatidylinositol 3-kinase (PI3K) small-interfering RNA (siRNA) or STAT3 lentiviral short hairpin RNA inhibited CCK/leptin-stimulated CART release. Silencing the expression of the EGR-1 gene inhibited the CCK/leptin-stimulated CART release but had no effect on CCK/leptin-stimulated neuronal firing. Electroporation of NG with CART siRNA inhibited CCK/leptin stimulated c-Fos expression in rat hypothalamus. Feeding studies following electroporation of the NG with CART or STAT3 siRNA abolished the effects of CCK/leptin on short-term satiety. We conclude that the synergistic interaction of low-affinity vagal CCKARs and LRbs mediates CART release from the NG, and CART is the principal neurotransmitter mediating short-term satiety. CART release from the NG involves interaction between CCK/SRC/PI3K cascades and leptin/JAK2/PI3K/STAT3 signaling pathways.


Assuntos
Leptina , Proteínas do Tecido Nervoso , Gânglio Nodoso , Saciação/efeitos dos fármacos , Sincalida , Animais , Regulação para Baixo , Eletroporação , Inativação Gênica , Imuno-Histoquímica , Leptina/metabolismo , Leptina/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Gânglio Nodoso/efeitos dos fármacos , Gânglio Nodoso/metabolismo , Coativadores de Receptor Nuclear/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Receptor de Colecistocinina A/metabolismo , Receptores para Leptina/metabolismo , Sincalida/metabolismo , Sincalida/farmacologia , Transmissão Sináptica/genética
14.
Neurogastroenterol Motil ; 34(7): e14339, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35315179

RESUMO

Abnormalities of mast cell structure or function may play prominent roles in irritable bowel syndrome (IBS) symptom genesis. Mast cells show close apposition to sensory nerves and release bioactive substances in response to varied stimuli including infection, stress, and other neuroendocrine factors. Most studies focus on patients who develop IBS after enteric infection or who report diarrhea-predominant symptoms. Three topics underlying IBS pathogenesis have been emphasized in recent investigations. Visceral hypersensitivity to luminal stimulation is found in most IBS patients and may contribute to abdominal pain. Mast cell dysfunction also may disrupt epithelial barrier function which alters mucosal permeability potentially leading to altered bowel function and pain. Mast cell products including histamine, proteases, prostaglandins, and cytokines may participate in hypersensitivity and permeability defects, especially with diarrhea-predominant IBS. Recent experimental evidence indicates that the pronociceptive effects of histamine and proteases are mediated by the generation of prostaglandins in the mast cell. Enteric microbiome interactions including increased mucosal bacterial translocation may activate mast cells to elicit inflammatory responses underlying some of these pathogenic effects. Therapies to alter mast cell activity (mast cell stabilizers) or function (histamine antagonists) have shown modest benefits in IBS. Future investigations will seek to define patient subsets with greater potential to respond to therapies that address visceral hypersensitivity, epithelial permeability defects, and microbiome alterations secondary to mast cell dysfunction in IBS.


Assuntos
Síndrome do Intestino Irritável , Diarreia/etiologia , Histamina , Humanos , Mucosa Intestinal/patologia , Mastócitos/patologia , Peptídeo Hidrolases , Permeabilidade , Prostaglandinas , Sensação
15.
JCI Insight ; 6(22)2021 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-34618688

RESUMO

Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) are carbohydrates thought to contribute to the symptoms of IBS. A diet in high in FODMAPs (HFM) induces gastrointestinal symptoms in patients with irritable bowel syndrome (IBS), and a diet low in FODMAPs (LFM) improves symptoms in up to 60% of patients with IBS. However, the mechanism by which FODMAPs affect IBS symptoms is unclear. We showed that mice fed on a HFM diet have mast cell activation and colonic barrier loss. Using mast cell-deficient mice with and without mast cell reconstitution, we showed that HFM-mediated colonic barrier loss is dependent on TLR4-dependent mast cell activation. In in vitro studies, we demonstrated that IBS fecal supernatant stimulates mast cells significantly more compared with fecal supernatant from healthy controls. This effect of IBS fecal supernatant on mast cell stimulation is ameliorated in the absence of the TLR4 receptor and after a LFM diet. We found that a LFM diet improves colonic barrier function and reduces mast cell activation while decreasing fecal LPS levels. Our findings indicate that a HFM diet causes mast cell activation via LPS, which in turn leads to colonic barrier loss, and a LFM diet reverses these pathophysiologic mucosal changes.


Assuntos
Fermentação/fisiologia , Gastroenteropatias/dietoterapia , Síndrome do Intestino Irritável/dietoterapia , Lipopolissacarídeos/metabolismo , Mastócitos/metabolismo , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Resultado do Tratamento
16.
J Physiol ; 588(Pt 4): 617-32, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20008464

RESUMO

The vagal afferent system is strategically positioned to mediate rapid changes in motility and satiety in response to systemic glucose levels. In the present study we aimed to identify glucose-excited and glucose-inhibited neurons in nodose ganglia and characterize their glucose-sensing properties. Whole-cell patch-clamp recordings in vagal afferent neurons isolated from rat nodose ganglia demonstrated that 31/118 (26%) neurons were depolarized after increasing extracellular glucose from 5 to 15 mm; 19/118 (16%) were hyperpolarized, and 68/118 were non-responsive. A higher incidence of excitatory response to glucose occurred in gastric- than in portal vein-projecting neurons, the latter having a higher incidence of inhibitory response. In glucose-excited neurons, elevated glucose evoked membrane depolarization (11 mV) and an increase in membrane input resistance (361 to 437 M). Current reversed at 99 mV. In glucose-inhibited neurons, membrane hyperpolarization (13 mV) was associated with decreased membrane input resistance (383 to 293 M). Current reversed at 97 mV. Superfusion of tolbutamide, a K(ATP) channel sulfonylurea receptor blocker, elicited identical glucose-excitatory but not glucose-inhibitory responses. Kir6.2 shRNA transfection abolished glucose-excited but not glucose-inhibited responses. Phosphatidylinositol bisphosphate (PIP(2)) depletion using wortmannin increased the fraction of glucose-excited neurons from 26% to 80%. These results show that rat nodose ganglia have glucose-excited and glucose-inhibited neurons, differentially distributed among gastric- and portal vein-projecting nodose neurons. In glucose-excited neurons, glucose metabolism leads to K(ATP) channel closure, triggering membrane depolarization, whereas in glucose-inhibited neurons, the inhibitory effect of elevated glucose is mediated by an ATP-independent K(+) channel. The results also show that PIP(2) can determine the excitability of glucose-excited neurons.


Assuntos
Glucose/fisiologia , Gânglio Nodoso/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Células Receptoras Sensoriais/fisiologia , Transportadores de Cassetes de Ligação de ATP/fisiologia , Androstadienos/farmacologia , Animais , Feminino , Glucose/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Gânglio Nodoso/citologia , Gânglio Nodoso/efeitos dos fármacos , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/fisiologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Ratos , Ratos Sprague-Dawley , Receptores de Droga/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Receptores de Sulfonilureias , Tolbutamida/farmacologia , Nervo Vago/citologia , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia , Wortmanina
17.
JCI Insight ; 4(17)2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31484832

RESUMO

Research shows that rats and humans on a high-fat diet (HFD) are less sensitive to satiety signals known to act via vagal afferent pathways. We hypothesize that HFD causes an upregulation of 2-pore domain potassium channels, resulting in hyperpolarization of nodose ganglia (NG) and decreased vagal response to satiety signals, which contribute to hyperphagia. We show that a 2-week HFD caused an upregulation of 2-pore domain TWIK-related spinal cord K+ (TRESK) and TWIK-related acid-sensitive K+ 1 (TASK1) channels by 330% ± 50% and 60% ± 20%, respectively, in NG. Patch-clamp studies of isolated NG neurons demonstrated a decrease in excitability. In vivo single-unit NG recordings showed that a 2-week HFD led to a 55% reduction in firing frequency in response to CCK-8 or leptin stimulation. NG electroporation with TRESK siRNA restored NG responsiveness to CCK-8 and leptin. Rats fed a 2-week HFD consumed ~40% more calories compared with controls. Silencing NG TRESK but not TASK1 channel expression in HFD-fed rats restored normal calorie consumption. In conclusion, HFD caused upregulation of TRESK channels, resulting in NG hyperpolarization and decreased vagal responsiveness to satiety signals. This finding provides a pharmacological target to prevent or treat HFD-induced hyperphagia.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Canais de Potássio/metabolismo , Nervo Vago/metabolismo , Animais , Ingestão de Energia , Inativação Gênica , Homeostase , Humanos , Leptina/metabolismo , Masculino , Proteínas do Tecido Nervoso , Neurônios/metabolismo , Obesidade/metabolismo , Potássio/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Sincalida/metabolismo , Medula Espinal/metabolismo , Transcriptoma , Regulação para Cima
19.
Neurosci Lett ; 373(1): 10-5, 2005 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-15555768

RESUMO

Concentration and time course of neurotransmitter in the synaptic cleft determines the amplitude and the duration of the resulting postsynaptic current. However, technical limitations involved in monitoring the time course of neurotransmitter concentration in the extra-cellular space have prevented direct evaluation of factors that influence neurotransmitter level in the cleft. Tetanic stimulation results in saturation of postsynaptic GABA(A) receptors in the rostral nucleus of the solitary tract (rNST) and GABA diffusion defines the decay time course of the inhibitory potentials or currents (IPSP/Cs). By applying a GABA concentration-response curve to these data it is possible to calculate the GABA concentration transient in the clefts of rNST inhibitory synapses. The analysis indicates that tetanic stimulation produces a GABA concentration that exceeds the concentration of neurotransmitter required to activate all postsynaptic GABA(A) receptors, resulting in short-term modification of the IPSP/Cs decay time. Moreover, the results also demonstrate that the rate of diffusion of GABA from the synaptic cleft is defined by two exponentials. A mathematical model of this process has been developed that supports these conclusions.


Assuntos
Modelos Teóricos , Núcleo Solitário/metabolismo , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Encéfalo/metabolismo , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Sinapses/química , Fatores de Tempo , Ácido gama-Aminobutírico/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA