Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Gut ; 57(12): 1654-64, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18682519

RESUMO

BACKGROUND: Acid in the oesophageal lumen is often sensed as heartburn. It was hypothesised that luminal CO(2), a permeant gas, rather than H(+), permeates through the epithelium, and is converted to H(+), producing an afferent neural signal by activating chemosensors. METHODS: The rat lower oesophageal mucosa was superfused with pH 7.0 buffer, and pH 1.0 or pH 6.4 high CO(2) (P(CO2) = 260 Torr) solutions with or without the cell-permeant carbonic anhydrase (CA) inhibitor methazolamide (MTZ, 1 mM), the cell-impermeant CA inhibitor benzolamide (BNZ, 0.1 mM), the transient receptor potential vanilloid 1 (TRPV1) antagonist capsazepine (CPZ, 0.5 mM) or the acid-sensing ion channel (ASIC) inhibitor amiloride (0.1 mM). Interstitial pH (pH(int)) was measured with 5',6'-carboxyfluorescein (5 mg/kg intravenously) loaded into the interstitial space, and blood flow was measured with laser-Doppler. RESULTS: Perfusion of a high CO(2) solution induced hyperaemia without changing pH(int), mimicking the effect of pH 1.0 perfusion. Perfused MTZ, BNZ, CPZ and amiloride all inhibited CO(2)-induced hyperaemia. CA XIV was expressed in the prickle cells, with CA XII in the basal cells. TRPV1 was expressed in the stratum granulosum and in the muscularis mucosa, whereas all ASICs were expressed in the prickle cells, with ASIC3 additionally in the muscularis mucosa. CONCLUSIONS: The response to CO(2) perfusion suggests that CO(2) diffuses through the stratum epithelium, interacting with TRPV1 and ASICs in the epithelium or in the submucosa. Inhibition of the hyperaemic response to luminal CO(2) by CA, TRPV1 and ASIC inhibitors implicates CA and these chemosensors in transduction of the luminal acid signal. Transepithelial CO(2) permeation may explain how luminal H(+) equivalents can rapidly be transduced into hyperaemia, and the sensation of heartburn.


Assuntos
Dióxido de Carbono/metabolismo , Esôfago/metabolismo , Refluxo Gastroesofágico/metabolismo , Hiperemia/metabolismo , Canais de Cátion TRPV/metabolismo , Canais Iônicos Sensíveis a Ácido , Amilorida/farmacologia , Animais , Benzolamida/farmacologia , Capsaicina/análogos & derivados , Capsaicina/antagonistas & inibidores , Dióxido de Carbono/farmacocinética , Inibidores da Anidrase Carbônica/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Monitoramento do pH Esofágico , Esôfago/irrigação sanguínea , Refluxo Gastroesofágico/complicações , Hiperemia/induzido quimicamente , Masculino , Metazolamida/farmacologia , Mucosa/metabolismo , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Ratos , Ratos Sprague-Dawley , Canais de Sódio/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores
2.
Artigo em Inglês | MEDLINE | ID: mdl-28714277

RESUMO

BACKGROUND: Short-chain fatty acids (SCFA) are microbial fermentation products absorbed by the colon. We recently reported that activation of the SCFA receptor termed free fatty acid receptor 3 (FFA3), expressed on cholinergic nerves, suppresses nicotinic acetylcholine receptor (nAChR)-mediated transepithelial anion secretion. This study aimed to clarify how activation of neurally expressed FFA3 affects colonic motor function. METHODS: FFA3-expressing myenteric neurons were identified by immunostaining; contractions of isolated circular muscle strips obtained from rat proximal colon were measured by isometric transducers. The effect of FFA3 agonists on defecation in vivo was examined in an exogenous serotonin-induced defecation model. KEY RESULTS: FFA3 immunoreactivity was located in nitrergic and cholinergic neurons in the myenteric plexus. In isolated circular muscle strips without mucosa and submucosa, the addition of nicotine (10 µM) or serotonin transiently relaxed the muscle through nitrergic neurons, whereas high concentrations of nicotine (100 µM) induced large-amplitude contractions that were mediated by cholinergic neurons. Pretreatment with FFA3 agonists inhibited nicotine- or serotonin-induced motility changes but had no effect on bethanechol-induced direct muscle contractions. The Gi/o inhibitor pertussis toxin reversed the inhibitory effect of an FFA3 agonist AR420626 on nicotine-evoked contractions, suggesting that FFA3 activation suppresses nAChR-mediated neural activity in myenteric neurons, consistent with an FFA3-mediated antisecretory effect. In conscious rats, exogenous serotonin increased the volume of fecal output, compared with the vehicle- or AR420626-treated groups. Pretreatment with AR420626 significantly suppressed serotonin-induced fecal output. CONCLUSION AND INFERENCES: FFA3 is a promising target for the treatment of neurogenic diarrheal disorders by suppressing nAChR-mediated neural pathways.


Assuntos
Colo/fisiologia , Motilidade Gastrointestinal , Neurônios/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Neurônios Colinérgicos/metabolismo , Colo/metabolismo , Defecação , Masculino , Contração Muscular , Plexo Mientérico/fisiologia , Neurônios/metabolismo , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Neurônios Nitrérgicos/metabolismo , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Serotonina/administração & dosagem , Antagonistas da Serotonina/administração & dosagem
3.
J Clin Invest ; 103(5): 605-12, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10074477

RESUMO

In vivo confocal imaging of the mucosal surface of rat stomach was used to measure pH noninvasively under the mucus gel layer while simultaneously imaging mucus gel thickness and tissue architecture. When tissue was superfused at pH 3, the 25 microm adjacent to the epithelial surface was relatively alkaline (pH 4.1 +/- 0.1), and surface alkalinity was enhanced by topical dimethyl prostaglandin E2 (pH 4.8 +/- 0.2). Luminal pH was changed from pH 3 to pH 5 to mimic the fasted-to-fed transition in intragastric pH in rats. Under pH 5 superfusion, surface pH was relatively acidic (pH 4.2 +/- 0.2). This surface acidity was enhanced by pentagastrin (pH 3.5 +/- 0.2) and eliminated by omeprazole, implicating parietal cell H,K-ATPase as the dominant regulator of surface pH under pH 5 superfusion. With either pH 5 or pH 3 superfusion (a) gastric pit lumens had the most divergent pH from luminal superfusates; (b) qualitatively similar results were observed with and without superfusion flow; (c) local mucus gel thickness was a poor predictor of surface pH values; and (d) no channels carrying primary gastric gland fluid through the mucus were observed. The model of gastric defense that includes an alkaline mucus gel and viscous fingering of secreted acid through the mucus may be appropriate at the intragastric pH of the fasted, but not fed, animal.


Assuntos
Mucosa Gástrica/metabolismo , Animais , Feminino , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Concentração de Íons de Hidrogênio , Masculino , Microscopia Confocal , Ratos
4.
J Clin Invest ; 108(12): 1807-16, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11748264

RESUMO

Secretion of bicarbonate from epithelial cells is considered to be the primary mechanism by which the duodenal mucosa is protected from acid-related injury. Against this view is the finding that patients with cystic fibrosis, who have impaired duodenal bicarbonate secretion, are paradoxically protected from developing duodenal ulcers. Therefore, we hypothesized that epithelial cell intracellular pH regulation, rather than secreted extracellular bicarbonate, was the principal means by which duodenal epithelial cells are protected from acidification and injury. Using a novel in vivo microscopic method, we have measured bicarbonate secretion and epithelial cell intracellular pH (pH(i)), and we have followed cell injury in the presence of the anion transport inhibitor DIDS and the Cl(-) channel inhibitor, 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB). DIDS and NPPB abolished the increase of duodenal bicarbonate secretion following luminal acid perfusion. DIDS decreased basal pH(i), whereas NPPB increased pH(i); DIDS further decreased pH(i) during acid challenge and abolished the pH(i) overshoot over baseline observed after acid challenge, whereas NPPB attenuated the fall of pH(i) and exaggerated the overshoot. Finally, acid-induced epithelial injury was enhanced by DIDS and decreased by NPPB. The results support the role of intracellular bicarbonate in the protection of duodenal epithelial cells from luminal gastric acid.


Assuntos
Bicarbonatos/metabolismo , Citoproteção , Duodeno/metabolismo , Ácido Gástrico/metabolismo , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Concentração de Íons de Hidrogênio , Mucosa Intestinal/metabolismo , Nitrobenzoatos/farmacologia , Ratos , Ratos Sprague-Dawley , Simportadores de Sódio-Bicarbonato/análise , Coloração e Rotulagem
5.
Neuroscience ; 134(1): 39-49, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15939544

RESUMO

Intestinal ischemia impairs gastrointestinal motility. The aims of this study were to investigate the effect of intestinal ischemia on gastrointestinal transit and on the expression of enteric transmitters in the rat, and whether the glutamate N-methyl-d-aspartate receptors influence these effects. Ischemia (1 h), induced by occluding the superior mesenteric artery, was followed by 0 or 24 h of reperfusion. Normal and sham-operated rats served as controls. Serosal blood flow was measured with laser Doppler flow meter. Gastrointestinal transit was measured as time of appearance of a marker in fecal pellets. Immunohistochemistry was used to evaluate the number of neurons immunoreactive for neuronal nitric oxide synthase (NOS) or vasoactive intestinal polypeptide and the density of substance P immunoreactive fibers in the myenteric plexus. The N-methyl-d-aspartate receptors antagonist, (+)-5-methyl-10,11-dihydro-5HT-[a,b] cyclohepten-5,10-imine (MK-801) (1 mg/kg i.v.) or the NOS inhibitor, N-nitro-l-arginine (10 mg/kg i.v.) was administered prior to ischemia. Serosal blood flow was decreased by 70% during ischemia, but it was not altered in sham-operated rats. Gastrointestinal transit was significantly prolonged in ischemic/reperfused rats compared with controls. There was a significant increase in the number of vasoactive intestinal polypeptide and neuronal nitric oxide synthase immunoreactive neurons, and a marked decrease of substance P immunoreactive fibers in ischemia followed by 24 h of reperfusion animals compared with controls. These alterations were not observed in ischemia without reperfusion. A significant delay of gastrointestinal transit and increase of vasoactive intestinal polypeptide neurons were also observed in sham-operated rats. The changes in transmitter expression and gastrointestinal transit in ischemic/reperfused rats were prevented by pre-treatment with the NOS inhibitor, N-nitro-l-arginine or the N-methyl-d-aspartate receptors antagonist, MK-801. This study suggests an involvement of the glutamatergic system and its interaction with nitric oxide in intestinal ischemia/reperfusion. Ischemia/reperfusion might induce local release of glutamate that activates N-methyl-d-aspartate receptors leading to increased production of nitric oxide and adaptive changes in enteric transmitters that might contribute to gastrointestinal dysmotility.


Assuntos
Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Trânsito Gastrointestinal/fisiologia , Isquemia/fisiopatologia , Plasticidade Neuronal/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/fisiologia , Traumatismo por Reperfusão/fisiopatologia , Análise de Variância , Animais , Arginina/farmacologia , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Trânsito Gastrointestinal/efeitos dos fármacos , Imuno-Histoquímica/métodos , Isquemia/metabolismo , Fluxometria por Laser-Doppler/métodos , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Traumatismo por Reperfusão/metabolismo , Fatores de Tempo , Peptídeo Intestinal Vasoativo/metabolismo
6.
Br J Pharmacol ; 172(19): 4655-70, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26061462

RESUMO

BACKGROUND AND PURPOSE: The bioactive monoamine 5-HT, implicated in the pathogenesis of functional gastrointestinal disorders, is abundantly synthesized and stored in rat proximal colonic mucosa and released to the gut lumen and subepithelial space. Despite much data regarding its expression and function, the effects of luminal 5-HT on colonic anion secretion have not been fully investigated. EXPERIMENTAL APPROACH: We measured short-circuit current (Isc ) as an indicator of ion transport in mucosa-submucosa or mucosa-only preparations of rat proximal colon. Total CO2 output was measured in vitro and in vivo. Immunohistochemistry was performed to investigate the localization of 5-HT4 , NOS1 and NOS2. KEY RESULTS: Luminal 5-HT gradually increased the amplitude and sustained the elevation of Isc . Luminal 5-HT-evoked ΔIsc was acetazolamide sensitive and HCO3 (-) dependent, consistent with cytosolic carbonic anhydrase-dependent electrogenic HCO3 (-) secretion, while not affected by tetrodotoxin (TTX), atropine or indomethacin. Pretreatment with the selective 5-HT4 antagonist GR113808, but not antagonists for 5-HT3 , 5-HT6 or 5-HT7 , inhibited luminal 5-HT-evoked ΔIsc . Furthermore, luminal cisapride and tegaserod increased Isc to the same extent as did 5-HT in the presence of indomethacin and TTX. Removal of the submucosa or pretreatment with NOS inhibitors enhanced luminal 5-HT-evoked ΔIsc , suggesting that NO synthesized in the submucosa suppresses mucosal anion secretion. NOS1 and NOS2 were immunostained in the submucosal neurons and glial cells respectively. Luminal 5-HT-evoked HCO3 (-) secretion was confirmed in vivo, inhibited by co-perfusion of GR113808, but not by ondansetron. CONCLUSIONS AND IMPLICATIONS: A novel apical 5-HT4 -mediated HCO3 (-) secretory pathway and an NO-dependent inhibitory mechanism are present in the proximal colon. Luminal 5-HT-evoked HCO3 (-) secretion may be important for the maintenance of mucosal integrity by regulating luminal pH.


Assuntos
Bicarbonatos/metabolismo , Colo/metabolismo , Receptores 5-HT4 de Serotonina/metabolismo , Serotonina/metabolismo , Animais , Mucosa Intestinal/metabolismo , Masculino , Óxido Nítrico/metabolismo , Ratos Sprague-Dawley , Antagonistas da Serotonina/farmacologia
7.
J Med Chem ; 42(19): 3899-909, 1999 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-10508438

RESUMO

We report herein the synthesis and biological testing of several glycosylated derivatives of some fluoroquinolone antibiotics. In particular, we have prepared several glycosylated derivatives of ciprofloxacin (2) in which the carbohydrate units are linked to the free secondary amine of the piperazine unit by: (a) no linker (e.g., a glycosylamine), (b) a beta-oxyethyl linker, and (c) a gamma-oxypropyl linker. Both glucose and galactose were used as carbohydrates so that six compounds of this type were prepared, e.g., no linker 4a,b, oxyethyl linker 5a,b, and oxypropyl linker 6a,b. In addition the aryl glycosides of glucose and galactose (7a,b) were prepared from the active 1-(4-hydroxyphenyl)fluoroquinolone (3.) The syntheses of the glycosylamines 4a,b involved the direct condensation of glucose and galactose with the hydrochloride salt of ciprofloxacin (2). For the oxyalkyl-linked compounds, we first prepared the peracetylated omega-bromoalkyl glycopyranosides 14a,b and 15a,b and then coupled them to the allyl ester of ciprofloxacin (11) to give, after saponification to remove all of the esters, the desired fluoroquinolone carbohydrates 5a,b and 6a,b. The final series was prepared from 2,4,5-trifluorobenzoyl chloride (22) which gave 3 in four precedented steps. Coupling of 3 with the peracetylated glucosyl and galactosyl halides 12a,b and 26 afforded, after saponification, the desired aryl glycosides 7a,b. Six of these derivatives of ciprofloxacin-4a,b, 5a,b, and 6a,b-were subjected to microbiological screening. Of the six, compound 6a showed the highest activity. Since 6a would give the hydroxypropyl-substituted ciprofloxacin on hydrolysis and its activity is approximately 4-8 times less than that of ciprofloxacin (2), this implies that compound 6a is probably being actively transported. Thus preliminary results suggest that some of the compounds are stable in culture conditions and may be differentially transported by multiple resistant organisms. In some cases, the addition of a linker and a carbohydrate to ciprofloxacin lessens, but does not eliminate, antimicrobial activity.


Assuntos
Anti-Infecciosos/metabolismo , Transporte Biológico Ativo , Ciprofloxacina/análogos & derivados , Ciprofloxacina/metabolismo , Galactose/metabolismo , Glucose/metabolismo , Glicosilação , Meia-Vida , Espectroscopia de Ressonância Magnética , Testes de Sensibilidade Microbiana , Piperazinas
8.
Aliment Pharmacol Ther ; 11(3): 605-12, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9218090

RESUMO

BACKGROUND: Bismuth compounds prevent gastric injury from the short-term administration of nonsteroidal anti-inflammatory drugs. We studied the mechanisms underlying the gastroprotective actions of bismuth subsalicylate against indomethacin-induced injury in rats. METHODS: An in vivo microscopic technique was used in which acid output, surface cell intracellular pH (pHi), gastric mucus gell thickness and mucosal blood flow were measured simultaneously. Concentrations of bismuth in mucus were measured by atomic absorption. RESULTS: Indomethacin (60 mg/kg) significantly thinned the mucus gel layer and augmented the decrease of pHi during luminal acid superfusion, consistent with a weakened gastric mucosal barrier to acid. Bismuth subsalicylate partially reversed this effect of indomethacin on pHi, consistent with gastroprotection. Neither a prostaglandin-inhibiting but non-injurious dose of indomethacin (5 mg/kg), bismuth subsalicylate, or their combination affected mucus gel thickness or pHi homeostasis. In separate experiments, indomethacin (60 mg/kg) significantly increased gastric mucus bismuth concentration in rats given bismuth subsalicylate. CONCLUSION: Bismuth accumulation in the gastric mucus during the evolution of mucosal injury may play an important role in the gastroprotective effect of bismuth subsalicylate against indomethacin injury.


Assuntos
Bismuto/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Compostos Organometálicos/farmacologia , Salicilatos/farmacologia , Animais , Bismuto/metabolismo , Velocidade do Fluxo Sanguíneo , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Indometacina/efeitos adversos , Masculino , Compostos Organometálicos/metabolismo , Ratos , Ratos Sprague-Dawley , Salicilatos/metabolismo
9.
Keio J Med ; 48(2): 63-8, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10405521

RESUMO

A viscoelastic mucus gel layer covers the gastric mucosa in a continuous sheet. The functions of the mucus gel have been one of the least studied aspects of gastric barrier function. Although the role of gastric mucus in providing physical protection against ingested particles, and preventing contact between digestive enzymes such as pepsin and the underlying mucosa is generally accepted, the barrier role function of gastric mucus with regard to luminal acid is still conjectural. The modest proton diffusion barrier that mucus provides is negligible in relation to the overall barrier properties of the gastric mucosa; nevertheless, stabilization of unstirred layers and damping of rapid shifts in luminal pH are potentially important functions. Associative studies have suggested a possible role of a hydrophobic barrier in strengthening the barrier functions of mucus. One of the most actively investigated areas of mucus function in recent times has been the mechanism by which secreted acid traverses the gel. Although compelling and complementary data obtained in vivo and in vitro have been consistent with secretion of acid under pressure, creating temporary viscous fingers through the gel, recent evidence obtained with in vivo confocal microscopy suggests that secreted acid diffuses through the gel. Since Helicobacter pylori exists solely in the juxtamucosal portion of the gastric mucus gel, detailed knowledge concerning the pH microenvironment in which the organism thrives is important in understanding the pathophysiology of peptic ulcer disease and related conditions.


Assuntos
Mucosa Gástrica/metabolismo , Muco/metabolismo , Animais , Difusão , Humanos , Permeabilidade
10.
Keio J Med ; 45(3): 155-60, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8897758

RESUMO

The gastric mucosa is constantly exposed to conditions that would normally be damaging to living cells. A complex defensive system has evolved that involves multiple mechanisms arranged in a laminar fashion, that as a whole constitute the gastric mucosal barrier to acid. As antisecretory therapy becomes perfected, more attention has been focused on these defensive components of the gastric mucosal barrier in disease. Recently, our laboratory has developed a means of measuring intracellular pH (pHi), mucosal blood flow, acid secretion, surface cell acidification rate, and acid secretion simultaneously in vivo. This system has enabled our laboratory to explore how the different components of the gastric mucosal barrier interact so as to protect the pHi of the surface cells under a variety of conditions. Analysis of these studies has revealed a significant inverse correlation between the initial fall in pHi of surface cells during luminal acid exposure and the thickness of the mucus gel, suggestive of a role of adherent gastric mucus in retarding the permeation of luminal protons into the epithelial cells. Another correlation has been between recovery of pHi and the presence of a hyperemic response to luminal acid, which suggests that the hyperemic response is an important defense mechanism in the intact mucosa. Our data is consistent with the hypothesis that gastric mucosal defense mechanisms, like gastric acid secretion, are dynamically regulated according to need. Disturbance of the regulation of these mechanisms, for example by cirrhosis, might be one of the major factors underlying clinical ulcer disease.


Assuntos
Mucosa Gástrica/fisiologia , Gastropatias/prevenção & controle , Ácidos , Animais , Estudos de Avaliação como Assunto , Mucosa Gástrica/citologia , Humanos , Concentração de Íons de Hidrogênio
11.
J Clin Pharmacol ; 33(1): 63-9, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8429116

RESUMO

Gentamicin uptake and toxicity was studied in a nontransformed cell line obtained from the S1 segment of the proximal tubule epithelium of a transgenic mouse. Cytotoxicity was assayed using the dye 3-(4,-5dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). Gentamicin uptake was assayed by a fluorescence polarization assay. No differences in toxicity were found among cells incubated for 4 hours in complete culture medium, enriched Kreb's buffer alone, or enriched Krebs' buffer with added 300 micrograms/mL gentamicin, 0.5 mmol/L L-lysine, or gentamicin plus L-lysine. Uptake of 300 micrograms/mL gentamicin was minimal at zero time and increased as a function of time. Uptake of gentamicin at 4 hours was positively correlated with medium gentamicin concentration. Addition of 0.5 mmol/L L-lysine inhibited uptake of 300 micrograms/mL gentamicin 38.9 +/- 10.2%. No other amino acid, including D-lysine or arginine, significantly changed gentamicin uptake. The authors conclude that gentamicin and L-lysine share a specific uptake mechanism located in the apical membrane of renal proximal tubule cells.


Assuntos
Gentamicinas/metabolismo , Túbulos Renais Proximais/metabolismo , Lisina/farmacologia , Animais , Linhagem Celular , Corantes , Meios de Cultura , Epitélio , Gentamicinas/toxicidade , Túbulos Renais Proximais/citologia , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Sais de Tetrazólio , Tiazóis
12.
Peptides ; 17(1): 155-9, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8822525

RESUMO

We studied the effect of the prostaglandin synthesis inhibitor, indomethacin, on pentagastrin-associated enhancements of gastric mucosal defense mechanisms, using a microfluorometric technique in which mucus gel thickness, intracellular pH (pHi), gastric mucosal blood flow, and acid secretion were measured simultaneously in vivo. Intravenous infusion with pentagastrin (80 micrograms/kg/h) increased mucus gel thickness, induced a hyperemic response to luminal acid, and enhanced pHi homeostasis during acid superfusion. Indomethacin, (5 mg/kg, IP) did not alter the effects of pentagastrin on acid output, mucus gel thickness, mucosal blood flow, and pHi homeostasis. Indomethacin alone did not affect any of these measures. We conclude that the enhancement of gastric defense mechanisms due to pentagastrin is independent of prostaglandin synthesis.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/fisiologia , Indometacina/farmacologia , Pentagastrina/farmacologia , Animais , Mucosa Gástrica/irrigação sanguínea , Homeostase/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Hiperemia/fisiopatologia , Masculino , Muco/efeitos dos fármacos , Muco/metabolismo , Prostaglandinas/biossíntese , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos
13.
J Gastrointest Surg ; 4(5): 458-63, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11077319

RESUMO

Portal hypertension (PHT) increases susceptibility of the gastric mucosa to injury. The aim of this study was to investigate whether PHT affects rat gastric mucosal defense mechanisms in vivo at the pre-epithelial, epithelial, and/or post-epithelial levels. PHT was produced in rats by staged portal vein ligation and sham-operated (SO) rats served as controls. The gastric mucosa was exposed, chambered, and continuously superfused with buffers under in vivo microscopy. We measured gastric mucosal gel layer thickness, surface epithelial cell intracellular pH (pHi), mucosal blood flow, and mucosal/serosal oxygenation. In PHT rats, gastric mucosal gel layer thickness was significantly reduced (88 +/- 16 microm in PHT rats vs. 135 +/- 25 microm in SO rats; P <0.0001), and the surface epithelial cell pHi was significantly decreased (6.80 +/- 0.11 in PHT rats vs. 7.09 +/- 0.21 in SO rats; P <0.01). Although total gastric mucosal blood flow was significantly increased in PHT rats by 72% (P <0.05), the oxygenation of the gastric mucosal surface was decreased by 42% (P <0.05) compared with SO rats. PHT impairs pre-epithelial (mucosal gel layer thickness), epithelial (pHi), and post-epithelial (maldistribution of blood flow) components of the gastric mucosal barrier. These findings can explain the increased susceptibility of portal hypertensive gastric mucosa to injury.


Assuntos
Mucosa Gástrica/patologia , Hipertensão Portal/patologia , Animais , Epitélio/patologia , Mucosa Gástrica/irrigação sanguínea , Concentração de Íons de Hidrogênio , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional
14.
Life Sci ; 69(25-26): 3073-81, 2001 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-11758832

RESUMO

The proximal duodenum is unique in that it is the only leaky epithelium regularly exposed to concentrated gastric acid. To prevent injury from occurring, numerous duodenal defense mechanisms have evolved. The most studied is bicarbonate secretion, which is presumed to neutralize luminal acid. Less well studied in their protective roles are the mucus gel layer and blood flow. Measuring duodenal epithelial intracellular pH [pHi], blood flow and mucus gel thickness (MGT), we studied duodenal defense mechanisms in vivo so as to more fully understand the mucosal response to luminal acid. Exposure of the mucosa to physiologic acid solutions promptly lowered pHi, followed by recovery after acid was removed, indicating that acid at physiologic concentrations readily diffuses into, but does not damage duodenal epithelial cells. Cellular acid then exits the cell via an amiloride-inhibitable process, presumably sodium-proton exchange (NHE). MGT and blood flow increase promptly during acid perfusion; both decrease after acid challenge and are inhibited by vanilloid receptor antagonists or by sensory afferent denervation. Bicarbonate secretion is not affected by acid superfusion but increases after challenge. Inhibition of cellular base loading lowers pHi, whereas inhibition of apical base extrusion alkalinizes pHi. These observations support the following hypothesis: luminal acid diffuses into the epithelial cells, lowering pHi. Acidic pHi increases the activity of a basolateral NHE, acidifying the submucosal space and increasing cellular base loading. The acidic submucosal space activates capsaicin receptors on afferent nerves, increasing MGT and blood flow. With concontinued acid exposure, a new steady state with thickened mucus gel, increased blood flow, and a higher cellular buffering power protects against acid injury. After acid challenge, mucus secretion decreases, blood flow slows, and pHi returns to normal, the latter occurring via apical bicarbonate extrusion, increasing bicarbonate secretion. Through these integrated mechanisms, the epithelial cells are protected from damage due to repeated pulses of concentrated gastric acid.


Assuntos
Duodeno/fisiologia , Ácido Gástrico/fisiologia , Mucosa Intestinal/fisiologia , Animais , Bicarbonatos/metabolismo , Epitélio/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Mucosa Intestinal/irrigação sanguínea , Microcirculação/fisiologia , Muco/fisiologia , Ratos
15.
JOP ; 2(4 Suppl): 268-73, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11875270

RESUMO

HCO(3)(-) secretion, which is believed to neutralize acid within the mucus gel, is the most studied duodenal defense mechanism. In general, HCO(3)(-) secretion rate and mucosal injury susceptibility correlate closely. Recent studies suggest that luminal acid can lower intracellular pH (pH(i)) of duodenal epithelial cells and that HCO(3)(-) secretion is unchanged during acid stress. Furthermore, peptic ulcers are rare in cystic fibrosis (CF), although, with impaired HCO(3)(-) secretion, increased ulcer prevalence is predicted, giving rise to the 'CF Paradox'. We thus tested the hypothesis that duodenal epithelial cell protection occurs as the result of pH(i) regulation rather than by neutralization of acid by HCO(3)(-) in the pre-epithelial mucus. Cellular acidification during luminal acid perfusion, and unchanged HCO(3)(-) secretion during acid stress are inconsistent with pre-epithelial acid neutralization by secreted HCO(3)(-). Furthermore, inhibition of HCO(3)(-) secretion by 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB) despite preservation of pH(i) and protection from acid-induced injury further question the pre-epithelial acid neutralization hypothesis. This decoupling of HCO(3)(-) secretion and injury susceptibility by NPPB (and possibly by CF) further suggest that cellular buffering, rather than HCO(3)(-) exit into the mucus, is of primary importance for duodenal mucosal protection, and may account for the lack of peptic ulceration in CF patients.


Assuntos
Bicarbonatos/metabolismo , Fibrose Cística/metabolismo , Úlcera Duodenal/metabolismo , Duodeno/metabolismo , Líquido Intracelular/metabolismo , Úlcera Péptica/metabolismo , Animais , Fibrose Cística/complicações , Úlcera Duodenal/etiologia , Duodeno/citologia , Humanos , Concentração de Íons de Hidrogênio , Úlcera Péptica/etiologia
17.
J Anim Sci ; 91(5): 1957-62, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23345558

RESUMO

Meal ingestion is followed by release of numerous hormones from enteroendocrine cells interspersed among the epithelial cells lining the intestine. Recently, the de-orphanization of G protein-coupled receptor (GPCR)-type nutrient receptors, expressed on the apical membranes of enteroendocrine cells, has suggested a plausible mechanism whereby luminal nutrients trigger the release of gut hormones. Activation of nutrient receptors triggers intracellular signaling mechanisms that promote exocytosis of hormone-containing granules into the submucosal space. Hormones released by foregut enteroendocrine cells include the glucagon-like peptides (GLP) affecting glycemic control (GLP-1) and releasing pro-proliferative, hypertrophy-inducing growth factors (GLP-2). The foregut mucosa, being exposed to pulses of concentrated HCl, is protected by a system of defense mechanisms, which includes epithelial bicarbonate and mucus secretion and augmentation of mucosal blood flow. We have reported that luminal co-perfusion of AA with nucleotides in anesthetized rats releases GLP-2 into the portal vein, associated with increased bicarbonate and mucus secretion and mucosal blood flow. The GLP-2 increases bicarbonate secretion via release of vasoactive intestinal peptide (VIP) from myenteric nerves. Luminal bile acids also release gut hormones due to activation of the bile-acid receptor known as G Protein-Coupled Receptor (GPR) 131, G Protein Bile Acid Receptor (GPBAR) 1, or Takeda G Protein-Coupled Receptor (TGR) 5, also expressed on enteroendocrine cells. The GLP are metabolized by dipeptidyl peptidase IV (DPPIV), an enzyme of particular interest to pharmaceutical, because its inhibition increases plasma concentrations of GLP-1 to treat diabetes. We have also reported that DPPIV inhibition enhances the secretory effects of nutrient-evoked GLP-2. Understanding the release mechanism and the metabolic pathways of gut hormones is of potential utility to the formulation of feedstuff additives that, by increasing nutrient absorption due to increased mucosal mass, can increase yields.


Assuntos
Aminoácidos/metabolismo , Ácidos e Sais Biliares/metabolismo , Duodeno/química , Duodeno/fisiologia , Mucosa Intestinal/metabolismo , Suínos/fisiologia , Aminoácidos/química , Animais , Bicarbonatos/metabolismo , Ácidos e Sais Biliares/química , Peptídeo 1 Semelhante ao Glucagon/sangue , Peptídeo 2 Semelhante ao Glucagon/sangue , Ratos
18.
Curr Med Chem ; 19(1): 28-34, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22300073

RESUMO

The field of gut nutrient chemosensing is evolving rapidly. Recent advances have uncovered the mechanism by which specific nutrient components evoke multiple metabolic responses. Deorphanization of G protein-coupled receptors (GPCRs) in the gut has helped identify previously unliganded receptors and their cognate ligands. In this review, we discuss nutrient receptors, their ligand preferences, and the evoked neurohormonal responses. Family A GPCRs includes receptor GPR93, which senses protein and proteolytic degradation products, and free fatty acid-sensing receptors. Short-chain free fatty acids are ligands for FFA2, previously GPR43, and FFA3, previously GPR41. FFA1, previously GPR40, is activated by long-chain fatty acids with GPR120 activated by medium- and long-chain fatty acids. The GPR119 agonist ethanolamide oleoylethanolamide (OEA) and bile acid GPR131 agonists have also been identified. Family C receptors ligand preferences include L-amino acids, carbohydrate, and tastants. The metabotropic glutamate receptor (mGluR), calcium-sensing receptor (CaR), and GPCR family C, group 6, subtype A receptor (GPRC6A) mediate L-amino acid-sensing. Taste receptors have a proposed role in intestinal chemosensing; sweet, bitter, and umami evoke responses in the gut via GPCRs. The mechanism of carbohydrate-sensing remains controversial: the heterodimeric taste receptor T1R2/T1R3 and sodium glucose cotransporter 1 (SGLT-1) expressed in L cells are the two leading candidates. Identification of specific nutrient receptors and their respective ligands can provide novel therapeutic targets for the treatment of diabetes, acid reflux, foregut mucosal injury, and obesity.


Assuntos
Alimentos , Trato Gastrointestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Humanos
19.
Acta Physiol (Oxf) ; 201(1): 77-84, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20518751

RESUMO

The upper gastrointestinal (GI) mucosa is exposed to endogenous and exogenous chemicals, including gastric acid, CO2 and nutrients. Mucosal chemical sensors are necessary to exert physiological responses such as secretion, digestion, absorption and motility. We propose the mucosal chemosensing system by which luminal chemicals are sensed to trigger mucosal defence mechanisms via mucosal acid sensors and taste receptors. Luminal acid/CO2 is sensed via ecto- and cytosolic carbonic anhydrases and ion transporters in the epithelial cells and via acid sensors on the afferent nerves in the duodenum and the oesophagus. Gastric acid sensing is differentially mediated via endocrine cell acid sensors and afferent nerves. Furthermore, a luminal l-glutamate signal is mediated via epithelial l-glutamate receptors, including metabotropic glutamate receptors and taste receptor 1 family heterodimers, with activation of afferent nerves and cyclooxygenase, whereas luminal Ca²(+) is differently sensed via the calcium-sensing receptor in the duodenum. These luminal chemosensors help to activate mucosal defence mechanisms in order to maintain the mucosal integrity and physiological responses of the upper GI tract. Stimulation of luminal chemosensing in the upper GI mucosa may prevent mucosal injury, affect nutrient metabolism and modulate sensory nerve activity.


Assuntos
Células Quimiorreceptoras/metabolismo , Duodeno/inervação , Duodeno/metabolismo , Mucosa Intestinal/metabolismo , Animais , Bicarbonatos/metabolismo , Cálcio/metabolismo , Dióxido de Carbono/metabolismo , Ácido Gástrico/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Prótons , Receptores de Detecção de Cálcio/metabolismo , Receptores de Glutamato/metabolismo
20.
Acta Physiol (Oxf) ; 201(1): 109-16, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20560899

RESUMO

The duodenum secretes HCO3⁻ as part of a multi-layered series of defence mechanisms against damage from luminal acid. In the 1980s, an alkaline surface layer was measured over the mucosa which correlated with the rate of HCO3⁻ secretion. As all biological processes are regulated, we investigated how the alkaline pH of the surface layer was maintained. As the ecto-phosphorylase alkaline phosphatase (AP) is highly expressed in the duodenal brush border, we hypothesized that its extreme alkaline pH optimum (∼pH 8-9) combined with its ability to hydrolyse regulatory purines such as ATP was part of an ecto-purinergic signalling system, consisting also of brush border P2Y receptors and cystic fibrosis transmembrane regulator-mediated HCO3⁻ secretion. Extracellular ATP increases the rate of HCO3⁻ secretion through this purinergic system. At high surface pH (pH(s)), AP activity is increased, which then increases the rate of ATP hydrolysis, decreasing surface ATP concentration ([ATP](s)), with a resultant decrease in the rate of HCO3⁻ secretion, which subsequently decreases pH(s) . This feedback loop is thus hypothesized to regulate pH(s) over the duodenal mucosa, and in several other HCO3⁻ secretory organs. As AP activity is directly related to pH(s) , and as AP hydrolyses ATP, [ATP](s) and pH(s) are co-regulated. As many essential tissue functions such as ciliary motility and lipid uptake are dependent on [ATP](s) , dysregulation of pH(s) and [ATP](s) may help explain the tissue dysfunction characteristic of diseases such as cystic fibrosis.


Assuntos
Trifosfato de Adenosina/metabolismo , Fibrose Cística/metabolismo , Duodeno/metabolismo , Concentração de Íons de Hidrogênio , Mucosa Intestinal/metabolismo , Metabolismo dos Lipídeos , Purinas/metabolismo , Animais , Bicarbonatos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Duodeno/anatomia & histologia , Humanos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa