Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Gastrointest Liver Physiol ; 318(6): G1070-G1087, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32390462

RESUMO

Lipopolysaccharides (LPS) are potent pro-inflammatory molecules that enter the systemic circulation from the intestinal lumen by uncertain mechanisms. We investigated these mechanisms and the effect of exogenous glucagon-like peptide-2 (GLP-2) on LPS transport in the rodent small intestine. Transmucosal LPS transport was measured in Ussing-chambered rat jejunal mucosa. In anesthetized rats, the appearance of fluorescein isothiocyanate (FITC)-LPS into the portal vein (PV) and the mesenteric lymph was simultaneously monitored after intraduodenal perfusion of FITC-LPS with oleic acid and taurocholate (OA/TCA). In vitro, luminally applied LPS rapidly appeared in the serosal solution only with luminal OA/TCA present, inhibited by the lipid raft inhibitor methyl-ß-cyclodextrin (MßCD) and the CD36 inhibitor sulfosuccinimidyl oleate (SSO), or by serosal GLP-2. In vivo, perfusion of FITC-LPS with OA/TCA rapidly increased FITC-LPS appearance into the PV, followed by a gradual increase of FITC-LPS into the lymph. Rapid PV transport was inhibited by the addition of MßCD or by SSO, whereas transport into the lymph was inhibited by chylomicron synthesis inhibition. Intraveous injection of the stable GLP-2 analog teduglutide acutely inhibited FITC-LPS transport into the PV, yet accelerated FITC-LPS transport into the lymph via Nω-nitro-l-arginine methyl ester (l-NAME)- and PG97-269-sensitive mechanisms. In vivo confocal microscopy in mouse jejunum confirmed intracellular FITC-LPS uptake with no evidence of paracellular localization. This is the first direct demonstration in vivo that luminal LPS may cross the small intestinal barrier physiologically during fat absorption via lipid raft- and CD36-mediated mechanisms, followed by predominant transport into the PV, and that teduglutide inhibits LPS uptake into the PV in vivo.NEW & NOTEWORTHY We report direct in vivo confirmation of transcellular lipopolysaccharides (LPS) uptake from the intestine into the portal vein (PV) involving CD36 and lipid rafts, with minor uptake via the canonical chylomicron pathway. The gut hormone glucagon-like peptide-2 (GLP-2) inhibited uptake into the PV. These data suggest that the bulk of LPS absorption is via the PV to the liver, helping clarify the mechanism of LPS transport into the PV as part of the "gut-liver" axis. These data do not support the paracellular transport of LPS, which has been implicated in the pathogenesis of the "leaky gut" syndrome.


Assuntos
Gorduras/metabolismo , Intestino Delgado/metabolismo , Lipopolissacarídeos/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Fármacos Gastrointestinais/farmacologia , Células HEK293 , Humanos , Intestino Delgado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/química , Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
2.
Dig Dis Sci ; 65(9): 2605-2618, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32006214

RESUMO

BACKGROUND: Circulating endotoxin (lipopolysaccharide, LPS) increases the gut paracellular permeability. We hypothesized that glucagon-like peptide-2 (GLP-2) acutely reduces LPS-related increased intestinal paracellular permeability by a mechanism unrelated to its intestinotrophic effect. METHODS: We assessed small intestinal paracellular permeability in vivo by measuring the appearance of intraduodenally perfused FITC-dextran 4000 (FD4) into the portal vein (PV) in rats 1-24 h after LPS treatment (5 mg/kg, ip). We also examined the effect of a stable GLP-2 analog teduglutide (TDG) on FD4 permeability. RESULTS: FD4 movement into the PV was increased 6 h, but not 1 or 3 h after LPS treatment, with increased PV GLP-2 levels and increased mRNA expressions of proinflammatory cytokines and proglucagon in the ileal mucosa. Co-treatment with a GLP-2 receptor antagonist enhanced PV FD4 concentrations. PV FD4 concentrations 24 h after LPS were higher than FD4 concentrations 6 h after LPS, reduced by exogenous GLP-2 treatment given 6 or 12 h after LPS treatment. FD4 uptake measured 6 h after LPS was reduced by TDG 3 or 6 h after LPS treatment. TDG-associated reduced FD4 uptake was reversed by the VPAC1 antagonist PG97-269 or L-NAME, not by EGF or IGF1 receptor inhibitors. CONCLUSIONS: Systemic LPS releases endogenous GLP-2, reducing LPS-related increased permeability. The therapeutic window of exogenous GLP-2 administration is at minimum within 6-12 h after LPS treatment. Exogenous GLP-2 treatment is of value in the prevention of increased paracellular permeability associated with endotoxemia.


Assuntos
Endotoxemia/prevenção & controle , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 2/agonistas , Absorção Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Dextranos/sangue , Modelos Animais de Doenças , Endotoxemia/sangue , Endotoxemia/induzido quimicamente , Fluoresceína-5-Isotiocianato/análogos & derivados , Receptor do Peptídeo Semelhante ao Glucagon 2/metabolismo , Mediadores da Inflamação/metabolismo , Intestino Delgado/metabolismo , Lipopolissacarídeos , Masculino , Permeabilidade , Veia Porta , Ratos Sprague-Dawley , Fatores de Tempo
3.
Am J Physiol Gastrointest Liver Physiol ; 316(6): G785-G796, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30978113

RESUMO

Xenin-25 is a neurotensin-like peptide that is secreted by enteroendocrine cells in the small intestine. Xenin-8 is reported to augment duodenal anion secretion by activating afferent neural pathways. The intrinsic neuronal circuits mediating the xenin-25-induced anion secretion were characterized using the Ussing-chambered, mucosa-submucosa preparation from the rat ileum. Serosal application of xenin-25 increased the short-circuit current in a concentration-dependent manner. The responses were abolished by the combination of Cl--free and HCO3- -free solutions. The responses were almost completely blocked by TTX (10-6 M) but not by atropine (10-5 M) or hexamethonium (10-4 M). The selective antagonists for neurotensin receptor 1 (NTSR1), neurokinin 1 (NK1), vasoactive intestinal polypeptide (VIP) receptors 1 and 2 (VPAC1 and VPAC2, respectively), and capsaicin, but not 5-hydroxyltryptamine receptors 3 and 4 (5-HT3 and 5-HT4), NTSR2, and A803467, inhibited the responses to xenin-25. The expression of VIP receptors (Vipr) in rat ileum was examined using RT-PCR. The Vipr1 PCR products were detected in the submucosal plexus and mucosa. Immunohistochemical staining showed the colocalization of NTSR1 and NK1 with substance P (SP)- and calbindin-immunoreactive neurons in the submucosal plexus, respectively. In addition, NK1 was colocalized with noncholinergic VIP secretomotor neurons. Based on the results from the present study, xenin-25-induced Cl-/ HCO3- secretion is involved in NTSR1 activation on intrinsic and extrinsic afferent neurons, followed by the release of SP and subsequent activation of NK1 expressed on noncholinergic VIP secretomotor neurons. Finally, the secreted VIP may activate VPAC1 on epithelial cells to induce Cl-/ HCO3- secretion in the rat ileum. Activation of noncholinergic VIP secretomotor neurons by intrinsic primary afferent neurons and extrinsic afferent neurons by postprandially released xenin-25 may account for most of the neurogenic secretory response induced by xenin-25. NEW & NOTEWORTHY This study is the first to investigate the intrinsic neuronal circuit responsible for xenin-25-induced anion secretion in the rat small intestine. We have found that nutrient-stimulated xenin-25 release may activate noncholinergic vasoactive intestinal polypeptide (VIP) secretomotor neurons to promote Cl-/ HCO3- secretion through the activation of VIP receptor 1 on epithelial cells. Moreover, the xenin-25-induced secretory responses are mainly linked with intrinsic primary afferent neurons, which are involved in the activation of neurotensin receptor 1 and neurokinin 1 receptor.


Assuntos
Ânions/metabolismo , Sistema Nervoso Entérico/metabolismo , Íleo , Vias Neurais/metabolismo , Neurotensina/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Animais , Hormônios Gastrointestinais/metabolismo , Íleo/inervação , Íleo/fisiologia , Mucosa Intestinal/metabolismo , Ratos , Receptores de Neurotensina/metabolismo
4.
Int J Mol Sci ; 19(3)2018 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-29510573

RESUMO

The diffuse chemosensory system (DCS) is well developed in the apparatuses of endodermal origin like gastrointestinal (GI) tract. The primary function of the GI tract is the extraction of nutrients from the diet. Therefore, the GI tract must possess an efficient surveillance system that continuously monitors the luminal contents for beneficial or harmful compounds. Recent studies have shown that specialized cells in the intestinal lining can sense changes in the luminal content. The chemosensory cells in the GI tract belong to the DCS which consists of enteroendocrine and related cells. These cells initiate various important local and remote reflexes. Although neural and hormonal involvements in ion transport in the GI tract are well documented, involvement of the DCS in the regulation of intestinal ion transport is much less understood. Since activation of luminal chemosensory receptors is a primary signal that elicits changes in intestinal ion transport and motility and failure of the system causes dysfunctions in host homeostasis, as well as functional GI disorders, study of the regulation of GI function by the DCS has become increasingly important. This review discusses the role of the DCS in epithelial ion transport, with particular emphasis on the involvement of free fatty acid receptor 2 (FFA2) and free fatty acid receptor 3 (FFA3).


Assuntos
Bicarbonatos/metabolismo , Células Quimiorreceptoras/metabolismo , Cloretos/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Humanos , Mucosa Intestinal/citologia , Transporte de Íons
5.
Am J Physiol Gastrointest Liver Physiol ; 313(2): G117-G128, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28526687

RESUMO

Serotonin (5-HT), predominantly synthesized and released by enterochromaffin cells, is implicated in gastrointestinal symptoms such as emesis, abdominal pain, and diarrhea. Because luminal short-chain fatty acids (SCFAs) release 5-HT from enterochromaffin cells, which express the SCFA receptor free fatty acid receptor 2 (FFA2) in rat duodenum, we examined the effects of the selective FFA2 agonist phenylacetamide-1 (PA1) on duodenal 5-HT release with consequent bicarbonate secretion [duodenal bicarbonate secretion (DBS)] and on indomethacin (IND)-induced enteropathy. Intestinal injury was induced by IND (10 mg/kg sc) with or without PA1. We measured DBS in vivo in a duodenal loop perfused with PA1 while measuring 5-HT released in the portal vein. Duodenal blood flow was measured by laser-Doppler flowmetry. IND induced small intestinal ulcers with duodenal sparing. PA1 given with IND (IND + PA1) dose dependently induced duodenal erosions. IND + PA1-induced duodenal lesions were inhibited by the FFA2 antagonist GLPG-0974, ondansetron, or omeprazole but not by RS-23597 or atropine. Luminal perfusion of PA1 augmented DBS accompanied by increased portal blood 5-HT concentrations with approximately eight times more release at 0.1 mM than at 1 µM, with the effects inhibited by coperfusion of GLPG-0974. Luminal PA1 at 1 µM increased, but at 0.1 mM diminished, duodenal blood flow. Cosuperfusion of PA1 (0.1 mM) decreased acid-induced hyperemia, further reduced by IND pretreatment but restored by ondansetron. These results suggest that, although FFA2 activation enhances duodenal mucosal defenses, FFA2 overactivation during ulcerogenic cyclooxygenase inhibition may increase the vulnerability of the duodenal mucosa to gastric acid via excessive 5-HT release and 5-HT3 receptor activation, implicated in foregut-related symptoms such as emesis and epigastralgia.NEW & NOTEWORTHY Luminal free fatty acid receptor 2 agonists stimulate enterochromaffin cells and release serotonin, which enhances mucosal defenses in rat duodenum. However, overdriving serotonin release with high luminal concentrations of free fatty acid 2 ligands such as short-chain fatty acids injures the mucosa by decreasing mucosal blood flow. These results are likely implicated in serotonin-related dyspeptic symptom generation because of small intestinal bacterial overgrowth, which is hypothesized to generate excess SCFAs in the foregut, overdriving serotonin release from enterochromaffin cells.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Duodeno/efeitos dos fármacos , Indometacina/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Receptores Acoplados a Proteínas G/agonistas , Serotonina/metabolismo , Animais , Bicarbonatos/metabolismo , Duodeno/metabolismo , Células Enterocromafins/efeitos dos fármacos , Células Enterocromafins/metabolismo , Mucosa Intestinal/metabolismo , Ratos
6.
J Pharmacol Exp Ther ; 361(1): 151-161, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28115552

RESUMO

Xenin-25, a neurotensin (NT)-related anorexigenic gut hormone generated mostly in the duodenal mucosa, is believed to increase the rate of duodenal ion secretion, because xenin-induced diarrhea is not present after Roux-en-Y gastric bypass surgery. Because the local effects of xenin on duodenal ion secretion have remained uninvestigated, we thus examined the neural pathways underlying xenin-induced duodenal anion secretion. Intravenous infusion of xenin-8, a bioactive C-terminal fragment of xenin-25, dose dependently increased the rate of duodenal HCO3- secretion in perfused duodenal loops of anesthetized rats. Xenin was immunolocalized to a subset of enteroendocrine cells in the rat duodenum. The mRNA of the xenin/NT receptor 1 (NTS1) was predominantly expressed in the enteric plexus, nodose and dorsal root ganglia, and in the lamina propria rather than in the epithelium. The serosal application of xenin-8 or xenin-25 rapidly and transiently increased short-circuit current in Ussing-chambered mucosa-submucosa preparations in a concentration-dependent manner in the duodenum and jejunum, but less so in the ileum and colon. The selective antagonist for NTS1, substance P (SP) receptor (NK1), or 5-hydroxytryptamine (5-HT)3, but not NTS2, inhibited the responses to xenin. Xenin-evoked Cl- secretion was reduced by tetrodotoxin (TTX) or capsaicin-pretreatment, and abolished by the inhibitor of TTX-resistant sodium channel Nav1.8 in combination with TTX, suggesting that peripheral xenin augments duodenal HCO3- and Cl- secretion through NTS1 activation on intrinsic and extrinsic afferent nerves, followed by release of SP and 5-HT. Afferent nerve activation by postprandial, peripherally released xenin may account for its secretory effects in the duodenum.


Assuntos
Duodeno/efeitos dos fármacos , Duodeno/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Neurotensina/farmacologia , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/metabolismo , Animais , Relação Dose-Resposta a Droga , Hormônios Gastrointestinais/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
7.
Dig Dis Sci ; 62(8): 1944-1952, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28523577

RESUMO

BACKGROUND: Therapy with nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with enteropathy in humans and experimental animals, a cause of considerable morbidity. Unlike foregut NSAID-associated mucosal lesions, most treatments for this condition are of little efficacy. We propose that the endogenously released intestinotrophic hormone glucagon-like peptide-2 (GLP-2) prevents the development of NSAID-induced enteropathy. Since the short-chain fatty acid receptor FFA3 is expressed on enteroendocrine L cells and on enteric nerves in the gastrointestinal tract, we further hypothesized that activation of FFA3 on L cells protects the mucosa from injury via GLP-2 release with enhanced duodenal HCO3- secretion. We thus investigated the effects of synthetic selective FFA3 agonists with consequent GLP-2 release on NSAID-induced enteropathy. METHODS: We measured duodenal HCO3- secretion in isoflurane-anesthetized rats in a duodenal loop perfused with the selective FFA3 agonists MQC or AR420626 (AR) while measuring released GLP-2 in the portal vein (PV). Intestinal injury was produced by indomethacin (IND, 10 mg/kg, sc) with or without MQC (1-10 mg/kg, ig) or AR (0.01-0.1 mg/kg, ig or ip) treatment. RESULTS: Luminal perfusion with MQC or AR (0.1-10 µM) dose-dependently augmented duodenal HCO3- secretion accompanied by increased GLP-2 concentrations in the PV. The effect of FFA3 agonists was inhibited by co-perfusion of the selective FFA3 antagonist CF3-MQC (30 µM). AR-induced augmented HCO3- secretion was reduced by iv injection of the GLP-2 receptor antagonist GLP-2(3-33) (3 nmol/kg), or by pretreatment with the cystic fibrosis transmembrane conductance regulator (CFTR) inhibitor CFTRinh-172 (1 mg/kg, ip). IND-induced small intestinal ulcers were dose-dependently inhibited by intragastric administration of MQC or AR. GLP-2(3-33) (1 mg/kg, ip) or CF3-MQC (1 mg/kg, ig) reversed AR-associated reduction in IND-induced enteropathy. In contrast, ip injection of AR had no effect on enteropathy. CONCLUSION: These results suggest that luminal FFA3 activation enhances mucosal defenses and prevents NSAID-induced enteropathy via the GLP-2 pathway. The selective FFA3 agonist may be a potential therapeutic candidate for NSAID-induced enteropathy.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Bicarbonatos/metabolismo , Duodeno/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Enteropatias/prevenção & controle , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Indometacina/efeitos adversos , Enteropatias/induzido quimicamente , Mucosa Intestinal/metabolismo , Masculino , Quinolonas/farmacologia , Ratos , Ratos Sprague-Dawley , Úlcera/induzido quimicamente , Úlcera/prevenção & controle
8.
J Physiol ; 594(12): 3339-52, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-26854275

RESUMO

KEY POINTS: Luminal short-chain fatty acids (SCFAs) influence gut physiological function via SCFA receptors and transporters. The contribution of an SCFA receptor, free fatty acid receptor (FFA)3, to the enteric nervous system is unknown. FFA3 is expressed in enteric cholinergic neurons. Activation of neural FFA3 suppresses Cl(-) secretion induced by nicotinic ACh receptor activation via a Gi/o pathway. Neural FFA3 may have an anti-secretory function by modulating cholinergic neural reflexes in the enteric nervous system. ABSTRACT: The proximal colonic mucosa is constantly exposed to high concentrations of microbially-produced short-chain fatty acids (SCFAs). Although luminal SCFAs evoke electrogenic anion secretion and smooth muscle contractility via neural and non-neural cholinergic pathways in the colon, the involvement of the SCFA receptor free fatty acid receptor (FFA)3, one of the free fatty acid receptor family members, has not been clarified. We investigated the contribution of FFA3 to cholinergic-mediated secretory responses in rat proximal colon. FFA3 was immunolocalized to enteroendocrine cells and to the enteric neural plexuses. Most FFA3-immunoreactive nerve fibres and nerve endings were cholinergic, colocalized with protein gene product (PGP)9.5, the vesicular ACh transporter, and the high-affinity choline transporter CHT1. In Ussing chambered mucosa-submucosa preparations (including the submucosal plexus) of rat proximal colon, carbachol (CCh)-induced Cl(-) secretion was decreased by TTX, hexamethonium, and the serosal FFA3 agonists acetate or propionate, although not by an inactive analogue 3-chloropropionate. Serosal application of a selective FFA3 agonist (N-[2-methylphenyl]-[4-furan-3-yl]-2-methyl-5-oxo-1,4,5,6,7,8-hexahydro-quinoline-3-carboxamide; MQC) dose-dependently suppressed the response to CCh but not to forskolin, with an IC50 of 13 µm. Pretreatment with MQC inhibited nicotine-evoked but not bethanechol-evoked secretion. The inhibitory effect of MQC was reversed by pretreatment with pertussis toxin, indicating that FFA3 acts via the Gi/o pathway. Luminal propionate induced Cl(-) secretion via the cholinergic pathway, which was reduced by MQC, as well as by TTX, hexamethonium or removal of the submucosal plexus. These results suggest that the SCFA-FFA3 pathway has a novel anti-secretory function in that it inhibits cholinergic neural reflexes in the enteric nervous system.


Assuntos
Colo/fisiologia , Sistema Nervoso Entérico/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Carbacol/farmacologia , Cloretos/metabolismo , Colo/efeitos dos fármacos , Colo/metabolismo , Células HeLa , Humanos , Técnicas In Vitro , Masculino , Neurônios/fisiologia , Propionatos/farmacologia , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/fisiologia
9.
J Physiol ; 593(3): 585-99, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25433076

RESUMO

KEY POINTS: Luminal lipid in the duodenum modulates gastroduodenal functions via the release of gut hormones and mediators such as cholecystokinin and 5-HT. The effects of luminal short-chain fatty acids (SCFAs) in the foregut are unknown. Free fatty acid receptors (FFARs) for long-chain fatty acids (LCFAs) and SCFAs are expressed in enteroendocrine cells. SCFA receptors, termed FFA2 and FFA3, are expressed in duodenal enterochromaffin cells and L cells, respectively. Activation of LCFA receptor (FFA1) and presumed FFA3 stimulates duodenal HCO3(-) secretion via a glucagon-like peptide (GLP)-2 pathway, whereas FFA2 activation induces HCO3(-) secretion via muscarinic and 5-HT4 receptor activation. The presence of SCFA sensing in the duodenum with GLP-2 and 5-HT signals further supports the hypothesis that luminal SCFA in the foregut may contribute towards the generation of functional symptoms. ABSTRACT: Intraduodenal fatty acids (FA) and bacterial overgrowth, which generate short-chain FAs (SCFAs), have been implicated in the generation of functional dyspepsia symptoms. We studied the mechanisms by which luminal SCFA perfusion affects duodenal HCO3(-) secretion (DBS), a measure of mucosal neurohumoral activation. Free fatty acid receptor (FFAR) 1 (FFA1), which binds long-chain FA (LCFA), and SCFA receptors FFA2 and FFA3 were immunolocalised to duodenal enteroendocrine cells. FFA3 colocalised with glucagon-like peptide (GLP)-1, whereas FFA2 colocalised with 5-HT. Luminal perfusion of the SCFA acetate or propionate increased DBS, enhanced by dipeptidyl peptidase-IV (DPPIV) inhibition, at the same time as increasing GLP-2 portal blood concentrations. Acetate-induced DBS was partially inhibited by monocarboxylate/HCO3(-) exchanger inhibition without affecting GLP-2 release, implicating acetate absorption in the partial mediation of DBS. A selective FFA2 agonist dose-dependently increased DBS, unaffected by DPPIV inhibition or by cholecystokinin or 5-HT3 receptor antagonists, but was inhibited by atropine and a 5-HT4 antagonist. By contrast, a selective FFA1 agonist increased DBS accompanied by GLP-2 release, enhanced by DPPIV inhibition and inhibited by a GLP-2 receptor antagonist. Activation of FFA1 by LCFA and presumably FFA3 by SCFA increased DBS via GLP-2 release, whereas FFA2 activation stimulated DBS via muscarinic and 5-HT4 receptor activation. SCFA/HCO3(-) exchange also appears to be present in the duodenum. The presence of duodenal fatty acid sensing receptors that signal hormone release and possibly signal neural activation may be implicated in the pathogenesis of functional dyspepsia.


Assuntos
Duodeno/metabolismo , Ácidos Graxos Voláteis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Animais , Colecistocinina/metabolismo , Células Cromafins/metabolismo , Duodeno/citologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Ratos , Serotonina/metabolismo
10.
Digestion ; 89(1): 31-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24458110

RESUMO

BACKGROUND: Gut microbiota affects host homeostasis and dysbiosis causes host diseases. Therefore, uncovering the sensing mechanism of bacterial metabolites such as short-chain fatty acid (SCFA) may help us to understand the host-microbiota interaction both in physiological and nonphysiological conditions. SUMMARY: The colonic lumen is continually exposed to many kinds of chemicals, including beneficial and harmful compounds that are produced by gut microbiota in addition to ingested nutrients. In the mammalian colon SCFAs such as acetate, propionate and butyrate are produced by bacterial fermentation and reach about 100 mM under physiological conditions. In this decade, SCFA receptor genes and their expression in the intestine have been identified as free fatty acid receptor (FFA)2 and FFA3. The FFAs are located in colonic enteroendocrine L cells producing and releasing an insulinotropic hormone, glucagon-like peptide-1 (GLP-1), and an anorectic hormone, peptide YY. Recent in vivo and in vitro studies suggest that SCFAs stimulate gut hormone secretion. Therefore, the SCFA-FFA signal is likely to be important for gut physiological functions. KEY MESSAGE: Colonic epithelial cells express chemical receptors that detect the luminal contents, particularly bacterial metabolites, and may be involved in the host's energy metabolism via GLP-1 release, as well as the mucosal defense system.


Assuntos
Colo/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptores de Superfície Celular/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Colo/microbiologia , Fibras na Dieta , Ácidos Graxos Voláteis/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Microbiota
11.
Biomed Res ; 44(1): 17-29, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36682797

RESUMO

The present study tried to clarify if mumefural would prevent hyperglycemia, one of the typical symptoms of type 2 diabetes mellitus (T2DM), since mumefural is an extract from Japanese apricots preventing hyperglycemia. To clarify if mumefural would prevent T2DM pathogenesis, we used Otsuka Long-Evans Tokushima fatty (OLETF) rats, T2DM model. Mumefural diminished hyperglycemia, HOMA-IR and plasma triglyceride concentration in OLETF rats under fasting conditions. In addition, mumefural elevated protein expression of sodium-coupled monocarboxylate transporter 1 (SMCT1) in the distal colon participating in absorption of weak organic acids, which behave as bases but not acids after absorption into the body. Mumefural also increased the interstitial fluid pH around the brain hippocampus lowered in OLETF rats compared with non-T2DM LETO rats used as control for OLETF rats. Amyloid-beta accumulation in the brain decreased in accordance with the pH elevation. On the one hand, mumefural didn't affect plasma concentrations of glucagon, GLP-1, GIP or PYY under fasting conditions. Taken together, these observations indicate that: 1) mumefural would be a useful functional food improving hyperglycemia, insulin resistance and the lowered interstitial fluid pH in T2DM; 2) the interstitial fluid pH would be one of key factors influencing the accumulation of amyloid-beta.


Assuntos
Diabetes Mellitus Tipo 2 , Hiperglicemia , Resistência à Insulina , Ratos , Animais , Ratos Endogâmicos OLETF , Glicemia/metabolismo , Insulina , Líquido Extracelular/metabolismo , Encéfalo/metabolismo , Concentração de Íons de Hidrogênio
12.
Am J Physiol Gastrointest Liver Physiol ; 302(7): G690-701, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22207576

RESUMO

In gastrointestinal (GI) physiology, anion and fluid secretion is an important function for host defense and is induced by changes in the luminal environment. The transient receptor potential A1 (TRPA1) channel is considered to be a chemosensor in several sensory tissues. Although the function of TRPA1 has been studied in GI motility, its contribution to the transepithelial ion transport system has rarely been discussed. In the present study, we investigated the secretory effect of the potential TRPA1 agonist allyl isothiocyanate (AITC) in rat and human colon using an Ussing chamber. The mucosal application of AITC (10(-6)-10(-3) M) induced Cl(-) and HCO(3)(-) secretion in a concentration-dependent manner, whereas the serosal application induced a significantly weaker effect. AITC-evoked anion secretion was attenuated by tissue pretreatment with piroxicam and prostaglandin (PG) E(2); however, this secretion was not affected by TTX, atropine, or extracellular Ca(2+) depletion. These experiments indicate that TRPA1 activation induces anion secretion through PG synthesis, independent of neural pathways in the colon. Further analysis also indicates that AITC-evoked anion secretion is mediated mainly by the EP(4) receptor subtype. The magnitude of the secretory response exhibited segmental heterogeneity in rat colon. Real-time PCR analysis showed the segmental difference was corresponding to the differential expression of EP(4) receptor and cyclooxygenase-1 and -2. In addition, RT-PCR, in situ hybridization, and immunohistochemical studies showed TRPA1 expression in the colonic epithelia. Therefore, we conclude that the activation of TRPA1 in colonic epithelial cells is likely involved in the host defense mechanism through rapid anion secretion.


Assuntos
Canais de Cálcio/metabolismo , Colo/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Canais de Cátion TRPC/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Idoso , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Canais de Cálcio/genética , Fenômenos Eletrofisiológicos/fisiologia , Feminino , Humanos , Masculino , Proteínas do Tecido Nervoso/genética , Piroxicam/farmacologia , Transporte Proteico , Ratos , Ratos Wistar , Receptores de Prostaglandina E Subtipo EP4/genética , Canal de Cátion TRPA1 , Canais de Cátion TRPC/genética , Canais de Potencial de Receptor Transitório/genética
13.
Med Sci Monit ; 18(5): BR181-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22534700

RESUMO

BACKGROUND: Previous studies have shown that administration of ghrelin exhibits protective and therapeutic effects in the gut. The aim of the present investigation was to examine the influence of ghrelin administration on the course of cysteamine-induced duodenal ulcers, as well as effects on mucosal production of oxygen free radicals and duodenal antioxidant defense. MATERIAL/METHODS: Duodenal ulcers were induced in male Wistar rats by cysteamine administered intragastrically at the dose of 200 mg/kg in 1 ml of saline, 3 times at 4-h intervals. Starting 24 h after the first dose of cysteamine, rats were treated intraperitoneally twice a day with saline or ghrelin given at the dose of 4, 8 or 16 nmol/kg/dose. Seven days after administration of the first dose of cysteamine, the study was terminated. RESULTS: Induction of ulcers by cysteamine was accompanied by a reduction in duodenal blood flow, mucosal DNA synthesis and mucosal activity of superoxide dismutase (SOD); whereas mucosal concentration of interleukin-1ß and malonyldialdehyde (MDA - an index of lipid peroxidation) were increased. Treatment with ghrelin increased healing rate of duodenal ulcers and enhanced duodenal blood flow, mucosal DNA synthesis and mucosal activity of SOD, and reduced mucosal concentration of interleukin-1ß and MDA. CONCLUSIONS: Treatment with ghrelin increases the healing rate of duodenal ulcers and this effect is related, at least in part, to improvement of duodenal mucosal blood flow, mucosal cell proliferation and antioxidant defense, as well as being related to reduction in mucosal oxidative stress and inflammatory response.


Assuntos
Cisteamina/efeitos adversos , Úlcera Duodenal/tratamento farmacológico , Grelina/administração & dosagem , Cicatrização/efeitos dos fármacos , Animais , Grelina/farmacologia , Grelina/uso terapêutico , Masculino , Ratos , Ratos Wistar
14.
Biomed Res ; 43(6): 223-239, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36517024

RESUMO

The inhibitory action of the secondary bile acid lithocholic acid (LCA) on neurally evoked Cl-/HCO3- secretion was investigated using the Ussing-chambered mucosal-submucosal preparation from the rat distal colon. Electrical field stimulation (EFS) evoked cholinergic and noncholinergic secretory responses in the rat distal colon. The responses were almost completely blocked by TTX (10-6 M) but not atropine (10-5 M) or hexamethonium (10-4 M). The selective antagonist for VIP receptor 1 (VPAC1) greatly reduced the EFS-evoked response. Thus, the rat distal colon may be predominantly innervated by noncholinergic VIP secretomotor neurons. Basolateral addition of 6 × 10-5 M LCA inhibited the EFS-evoked response. The inhibitory action of LCA was partly rescued by the Y2R antagonist BIIE0246. The bile acid receptor TGR5 agonist INT-777 mimicked the LCA-induced inhibitory action. Immunohistochemical staining showed the colocalization of TGR5 and PYY on L cells. TGR5 immunoreactivity was also found in VIP-immunoreactive submucosal neurons which also expressed the PYY receptor, Y2R. These results suggest that LCA inhibits neurally evoked Cl-/HCO3- secretion through the activation of TGR5 on L cells and cholinergic- and VIP-secretomotor neurons in the submucosal plexus. Furthermore, the inhibitory mechanism may involve TGR5-stimulated PYY release from L cells and Y2R activation in VIP-secretomotor neurons.


Assuntos
Ácidos e Sais Biliares , Ácido Litocólico , Ratos , Animais , Ácido Litocólico/farmacologia , Ácido Litocólico/metabolismo , Mucosa Intestinal/metabolismo , Cloretos/metabolismo , Transporte de Íons , Colo/metabolismo , Colinérgicos/metabolismo
15.
Peptides ; 147: 170680, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34757144

RESUMO

Xenin-25 has a variety of physiological functions in the gastrointestinal tract, including ion transport and motility. Xenin-25 and neurotensin show sequence homology, especially near their C-terminal regions. The sequence similarity between xenin-25 and neurotensin indicates that the effects of xenin-25 is mediated by the neurotensin receptor but some biological actions of xenin-25 are independent. We have previously reported that xenin-25 modulates intestinal ion transport and colonic smooth muscle activity. However, minimal biological domain of xenin-25 to induce ion transport was not clear. To improve the mechanistic understanding of xenin-25 and to gain additional insights into the functions of xenin-25, the present study was designed to determine the minimal biological domain of xenin-25 required for ion transport in the rat ileum using various truncated xenin fragments and analogues in an Ussing chamber system. The present results demonstrate that the minimum biological domain of xenin-25 to induce Cl-/HCO3- secretion in the ileum contains the C-terminal pentapeptide. Furthermore, Arg at position 21 is important to retain the biological activity of xenin-25 and induces Cl-/HCO3- secretion in the rat ileum.


Assuntos
Ânions/metabolismo , Íleo/metabolismo , Neurotensina/metabolismo , Animais , Íleo/efeitos dos fármacos , Masculino , Neurotensina/análogos & derivados , Neurotensina/genética , Neurotensina/farmacologia , Domínios Proteicos , Pirazóis/farmacologia , Quinolinas/farmacologia , Ratos Sprague-Dawley , Receptores de Neurotensina/antagonistas & inibidores
16.
Pflugers Arch ; 461(1): 141-52, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20945073

RESUMO

Propionate, a fermented product in the lumen of the large intestine, is a short-chain fatty acid (SCFA) known to have a variety of localized physiological and pathophysiological functions (e.g., luminal fluid secretion and anti-inflammatory response). In the present study, we investigated propionate-induced transepithelial ion transport and the expression of SCFA receptor, free fatty acid receptor 2 (FFA2, otherwise known as GPR43) in the guinea pig distal colon utilizing the Ussing chamber technique and immunohistochemistry. The addition of propionate to the luminal bathing solution concentration-dependently induced transient K(+) and Cl(-) and/or bicarbonate secretion within approximately 30 s and long-lasting Cl(-) secretion for approximately 60 min was first identified in the present study. The transient anion secretion was tetrodotoxin (TTX)-sensitive and mediated through the cholinergic (both nicotinic and muscarinic) neural pathway, but the transient K(+) and long-lasting Cl(-) secretion were due to TTX-insensitive mechanism. Immunohistochemistry studies showed that some chromogranin A-immunoreactive enteroendocrine cells were also immunoreactive for FFA2 but not colocalized with 5-hydroxytryptamine. In conclusion, the propionate-induced secretion consisted of the neural and non-neural three-phase secretory manner possibly mediated by the stimulation of FFA2 expressed by enteroendocrine cells.


Assuntos
Bicarbonatos/metabolismo , Cloretos/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Potássio/metabolismo , Propionatos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Atropina/farmacologia , Bumetanida/farmacologia , Capsaicina/farmacologia , Colo/metabolismo , Dioxanos/farmacologia , Células Enteroendócrinas/metabolismo , Granisetron/farmacologia , Cobaias , Hexametônio/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Masculino , Piperidinas/farmacologia , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacologia , Membrana Serosa/efeitos dos fármacos , Suramina/farmacologia , Tetrodotoxina/farmacologia , ômega-Conotoxina GVIA/farmacologia
17.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G1132-43, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21372164

RESUMO

Gut lumen is continually exposed to a great variety of agents, including noxious compounds. Chemical receptors that detect the luminal environment are thought to play an important role as sensors and to modulate gastrointestinal functions. Recently, it has been reported that odorant receptors (ORs) are expressed in the small intestinal mucosa and that odorants stimulate serotonin secretion. However, ion transport in the responses to odorants has rarely been discussed, particularly in relation to the large intestine. In the present study, we examined the effects of the OR ligand thymol on ion transport in human and rat colonic epithelia using an Ussing chamber. In the mucosal-submucosal preparations, the mucosal addition of thymol evoked anion secretion concentration dependently. In addition, dextran (4 kDa) permeability was enhanced by the mucosal treatment with thymol. The response to thymol was not affected by tetrodotoxin (TTX) or piroxicam treatments in human or rat colon. Thymol-evoked electrogenic anion secretion was abolished under Ca(2+)-free conditions or mucosal treatment with transient receptor potential (TRP) A1 blocker (HC-030031). Pretreatment of thymol did not affect electrical field stimulation-evoked anion secretion but significantly attenuated short-chain fatty acid-evoked secretion in a concentration-dependent manner. OR1G1 and TRPA1 expression was investigated in isolated colonic mucosa by RT-PCR. The present results provide evidence that the OR ligand thymol modulates epithelial permeability and electrogenic anion secretion in human and rat colon. The anion secretion by luminal thymol is most likely mediated by direct activation of TRPA1 channel. We suggest that the sensing and responding to odorants in the colon also plays a role in maintaining intestinal homeostasis.


Assuntos
Colo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Receptores Odorantes/efeitos dos fármacos , Timol/farmacologia , Idoso , Análise de Variância , Animais , Anquirinas/genética , Anquirinas/metabolismo , Bicarbonatos/metabolismo , Transporte Biológico , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Cloretos/metabolismo , Colo/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Dextranos/metabolismo , Relação Dose-Resposta a Droga , Estimulação Elétrica , Células Epiteliais/metabolismo , Ácidos Graxos/metabolismo , Feminino , Humanos , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Masculino , Potenciais da Membrana , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Permeabilidade , Inibidores de Fosfodiesterase/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Antagonistas da Serotonina/farmacologia , Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Canal de Cátion TRPA1 , Canais de Cátion TRPC , Fatores de Tempo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo , Fosfolipases Tipo C/metabolismo
18.
Physiol Rep ; 9(4): e14752, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33600071

RESUMO

Xenin25 has a variety of physiological functions in the Gastrointestinal (GI) tract, including ion transport and motility. However, the motility responses in the colon induced by Xenin25 remain poorly understood. Therefore, the effect of Xenin25 on the spontaneous circular muscle contractions of the rat distal colon was investigated using organ bath chambers and immunohistochemistry. Xenin25 induced the inhibition followed by postinhibitory spontaneous contractions with a higher frequency in the rat distal colon. This inhibitory effect of Xenin25 was significantly suppressed by TTX but not by atropine. The inhibitory time (the duration of inhibition) caused by Xenin25 was shortened by the NTSR1 antagonist SR48692, the NK1R antagonist CP96345, the VPAC2 receptor antagonist PG99-465, the nitric oxide-sensitive guanylate-cyclase inhibitor ODQ, and the Ca2+ -dependent K+ channel blocker apamin. The higher frequency of postinhibitory spontaneous contractions induced by Xenin25 was also attenuated by ODQ and apamin. SP-, NOS-, and VIP-immunoreactive neurons were detected in the myenteric plexus (MP) of the rat distal colon. Small subsets of the SP-positive neurons were also Calbindin positive. Most of the VIP-positive neurons were also NOS positive, and small subsets of the NK1R-positive neurons were also VIP positive. Based on the present results, we propose the following mechanism. Xenin25 activates neuronal NTSR1 on the SP neurons of IPANs, and transmitters from the VIP and apamin-sensitive NO neurons synergistically inhibit the spontaneous circular muscle contractions via NK1R. Subsequently, the postinhibitory spontaneous contractions are induced by the offset of apamin-sensitive NO neuron activation via the interstitial cells of Cajal. In addition, Xenin25 also activates the muscular NTSR1 to induce relaxation. Thus, Xenin25 is considered to be an important modulator of post prandial circular muscle contraction of distal colon since the release of Xenin25 from enteroendocrine cells is stimulated by food intake.


Assuntos
Colo/inervação , Sistema Nervoso Entérico/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/inervação , Neurotensina/farmacologia , Animais , Sistema Nervoso Entérico/metabolismo , Técnicas In Vitro , Masculino , Inibição Neural/efeitos dos fármacos , Neurônios Nitrérgicos/efeitos dos fármacos , Neurônios Nitrérgicos/metabolismo , Ratos Sprague-Dawley , Receptores de Neurotensina/metabolismo , Substância P/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo
19.
Biomed Res ; 41(5): 199-216, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33071256

RESUMO

The microbiota-gut-brain axis transmits bidirectional communication between the gut and the central nervous system and links the emotional and cognitive centers of the brain with peripheral gut functions. This communication occurs along the axis via local, paracrine, and endocrine mechanisms involving a variety of gut-derived peptide/amine produced by enteroendocrine cells. Neural networks, such as the enteric nervous system, and the central nervous system, including the autonomic nervous system, also transmit information through the microbiota-gut-brain axis. Recent advances in research have described the importance of the gut microbiota in influencing normal physiology and contributing to disease. We are only beginning to understand this bidirectional communication system. In this review, we summarize the available data supporting the existence of these interactions, highlighting data related to the contribution of enteroendocrine cells and the enteric nervous system as an interface between the gut microbiota and brain.


Assuntos
Encéfalo/fisiologia , Sistema Nervoso Central/fisiologia , Sistema Nervoso Entérico/fisiologia , Microbioma Gastrointestinal/fisiologia , Animais , Ansiedade/complicações , Ácidos e Sais Biliares/química , Depressão/complicações , Células Enteroendócrinas/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Imuno-Histoquímica , Ligantes , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA