Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
2.
Article in English | MEDLINE | ID: mdl-28714277

ABSTRACT

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.


Subject(s)
Colon/physiology , Gastrointestinal Motility , Neurons/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Cholinergic Neurons/metabolism , Colon/metabolism , Defecation , Male , Muscle Contraction , Myenteric Plexus/physiology , Neurons/metabolism , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Nitrergic Neurons/metabolism , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Serotonin/administration & dosage , Serotonin Antagonists/administration & dosage
3.
Br J Pharmacol ; 172(19): 4655-70, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26061462

ABSTRACT

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.


Subject(s)
Bicarbonates/metabolism , Colon/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Serotonin/metabolism , Animals , Intestinal Mucosa/metabolism , Male , Nitric Oxide/metabolism , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology
4.
J Anim Sci ; 91(5): 1957-62, 2013 May.
Article in English | MEDLINE | ID: mdl-23345558

ABSTRACT

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.


Subject(s)
Amino Acids/metabolism , Bile Acids and Salts/metabolism , Duodenum/chemistry , Duodenum/physiology , Intestinal Mucosa/metabolism , Swine/physiology , Amino Acids/chemistry , Animals , Bicarbonates/metabolism , Bile Acids and Salts/chemistry , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 2/blood , Rats
5.
Curr Med Chem ; 19(1): 28-34, 2012.
Article in English | MEDLINE | ID: mdl-22300073

ABSTRACT

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.


Subject(s)
Food , Gastrointestinal Tract/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Humans
6.
Acta Physiol (Oxf) ; 201(1): 109-16, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20560899

ABSTRACT

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.


Subject(s)
Adenosine Triphosphate/metabolism , Cystic Fibrosis/metabolism , Duodenum/metabolism , Hydrogen-Ion Concentration , Intestinal Mucosa/metabolism , Lipid Metabolism , Purines/metabolism , Animals , Bicarbonates/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Duodenum/anatomy & histology , Humans , Signal Transduction/physiology
7.
Acta Physiol (Oxf) ; 201(1): 77-84, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20518751

ABSTRACT

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.


Subject(s)
Chemoreceptor Cells/metabolism , Duodenum/innervation , Duodenum/metabolism , Intestinal Mucosa/metabolism , Animals , Bicarbonates/metabolism , Calcium/metabolism , Carbon Dioxide/metabolism , Gastric Acid/metabolism , Glutamic Acid/metabolism , Humans , Hydrogen-Ion Concentration , Protons , Receptors, Calcium-Sensing/metabolism , Receptors, Glutamate/metabolism
8.
Gut ; 57(12): 1654-64, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18682519

ABSTRACT

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.


Subject(s)
Carbon Dioxide/metabolism , Esophagus/metabolism , Gastroesophageal Reflux/metabolism , Hyperemia/metabolism , TRPV Cation Channels/metabolism , Acid Sensing Ion Channels , Amiloride/pharmacology , Animals , Benzolamide/pharmacology , Capsaicin/analogs & derivatives , Capsaicin/antagonists & inhibitors , Carbon Dioxide/pharmacokinetics , Carbonic Anhydrase Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Esophageal pH Monitoring , Esophagus/blood supply , Gastroesophageal Reflux/complications , Hyperemia/chemically induced , Male , Methazolamide/pharmacology , Mucous Membrane/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors
9.
J Physiol Pharmacol ; 58(4): 767-91, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18195487

ABSTRACT

We measured villous cell intracellular pH (pH(i)) and solute diffusion between the bathing media and the epithelial cells in stripped, chambered mouse duodenum. Apical perfusion of a high CO2 solution rapidly acidified the upper villous cells with recovery after its removal. Apical zoniporide (ZP) enhanced CO(2)-induced acidification. Serosal ZP, dimethylamiloride (DMA) or stilbene anion transport inhibitors failed to alter CO(2)-induced acidification, whereas serosal high CO(2) buffer acidified the upper villous cells. Serosal 5-hydroxytryptamine rapidly acidified the upper villous cells. All serosally-perfused fluorescent compounds stained the crypt area, but not the villi or villous cells. In contrast, intravenous carboxyfluorescein quickly diffused into the interstitial space of the entire mucosa, and mucosally perfused fluorescent compound rapidly penetrated the epithelial cell layer. In muscle-stripped duodenum mounted in a small-aperture perfusion chamber, serosal solutes can readily diffuse only to the crypt cell region, whereas access to the villous epithelial cells is diffusion-limited. In contrast, rapid villous cell responses to serosally applied solutes are best explained by neural reflexes. Limited viability of the villous cells and impaired structural stability of the villi further limit long-term, villous cell functional studies of mucosal preparations mounted in small aperture diffusion chambers.


Subject(s)
Duodenum/metabolism , Intestinal Mucosa/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Anions , Biological Transport/drug effects , Carbon Dioxide/metabolism , Diffusion , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Guanidines/pharmacology , Hydrogen-Ion Concentration , Male , Mice , Mice, Inbred C57BL , Pyrazoles/pharmacology , Serotonin/pharmacology , Sodium-Bicarbonate Symporters/antagonists & inhibitors , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Solutions , Stilbenes/metabolism
10.
Aliment Pharmacol Ther ; 24 Suppl 4: 169-76, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17209861

ABSTRACT

The duodenum serves as a buffer zone between the stomach and the jejunum. Over a length of only 25 cm, large volumes of strong acid secreted by the stomach must be converted to the neutral-alkaline chyme of the hindgut lumen, generating large volumes of CO(2). The duodenal mucosa consists of epithelial cells connected by low-resistance tight junctions, forming a leaky epithelial barrier. Despite this permeability, the epithelial cells, under intense stress from luminal mineral acid and highly elevated Pco(2), maintain normal functioning. Bicarbonate ion uniquely protects the duodenal epithelial cells from acid-related injury. The specific protective mechanisms likely involve luminal bicarbonate secretion, intracellular pH buffering and interstitial buffering. Furthermore, the duodenum plays an active role in foregut acid-base homeostasis, absorbing large amounts of H(+) and CO(2). We have studied mucosal protection and acid-base balance using live-animal fluorescence ratio microimaging and by performing H(+) and CO(2) balance studies on duodenal perfusates. On the basis of these data, we have formulated novel hypotheses with regard to mucosal protection.


Subject(s)
Acid-Base Equilibrium , Bicarbonates/metabolism , Duodenum/metabolism , Intestinal Mucosa/metabolism , Animals , Antiporters/metabolism , Duodenum/pathology , Humans , Intestinal Mucosa/pathology , Peptic Ulcer/metabolism , TRPV Cation Channels/metabolism
11.
Gut ; 52(6): 775-83, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12740330

ABSTRACT

BACKGROUND AND AIMS: The early responses of the oesophageal mucosa to acid perfusion may predict subsequent pathology. Mucosal responses to luminal acid may result either from acid permeating through the mucosa or from other unknown transduction mechanisms. In order to better understand the dynamics of acid permeation into the oesophageal mucosa, we measured interstitial pH (pH(int)) of the oesophageal basal epithelial layer, pre-epithelial layer thickness, and blood flow in rats in vivo during luminal acid challenge. A novel confocal microscopic technique was used in vitro to measure pH(int) from defined cellular sites in response to luminal and basolateral acidification. METHODS: 5-(and-6)-Carboxyfluorescein (CF) and carboxy-seminapthorhodofluor-1 (SNARF-1) fluorescence was used to measure pH(int) by conventional and confocal microscopy, respectively, in urethane anaesthetised rats. Pre-epithelial layer thickness was measured optically with carbon particles as markers. Blood flow was measured with laser Doppler flowmetry. RESULTS: Luminal acidification failed to alter pH(int) in vivo and in vitro, but pH(int) was lowered by modest serosal acidification. Pre-epithelial layer thickness and blood flow increased significantly during luminal surface acid perfusion. Indomethacin had no effect on any acid related response. CONCLUSION: In this first dynamic measurement of oesophageal acid permeation and pre-epithelial layer thickness, pH(int) was preserved in spite of high luminal acidity by two complementary techniques. Despite the apparent permeability barrier to acid permeation, oesophageal blood flow and thickness responded to luminal acid perfusion.


Subject(s)
Acids/pharmacokinetics , Esophagus/metabolism , Animals , Benzopyrans , Esophagus/anatomy & histology , Esophagus/blood supply , Fluorescent Dyes , Hydrogen-Ion Concentration , Laser-Doppler Flowmetry , Male , Microscopy, Confocal , Mucous Membrane/metabolism , Permeability , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects
12.
J Physiol Pharmacol ; 54 Suppl 4: 19-26, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15075446

ABSTRACT

The proximal duodenal mucosa, exposed to frequent pulses of gastric acid, is functionally "leaky", increasing the importance of defense mechanisms such as the mucus gel layer, cellular acid/base transporters, bicarbonate secretion, and mucosal blood flow. Our laboratory has used a unique in vitro perfused microscopic system to measure thickness of the adherent mucus gel (MGT), intracellular pH (pHi), bicarbonate secretion, and mucosal blood flow in anesthetized rats. Exposure to pulses of luminal acid, mimicking the rapid physiologic shifts of luminal pH, increases MGT and blood flow, and induces cellular bicarbonate loading, the latter followed by augmented bicarbonate secretion. The mechanism by which the epithelium senses luminal acid includes capsazepine-inhibitable vanilloid receptors, presumably similar to the vanilloid receptor TPVR-1. CFTR, the cAMP-regulated anion channel mutated in the disease cystic fibrosis, plays an essential role in duodenal bicarbonate secretion. Our data are consistent with the hypothesis that cellular bicarbonate loading is an important means of preserving epithelial pHi during luminal acid challenge. Increased MGT may damp rapid shifts of luminal pH. Enhanced mucosal blood flow plays a significant role in the removal of back-diffusing acid. These neurally coordinated systems act coherently to defend the vulnerable duodenal epithelial cells from concentrated gastric acid.


Subject(s)
Duodenum/physiology , Gastric Acid/physiology , Intestinal Mucosa/physiology , Animals , Duodenum/cytology , Duodenum/metabolism , Gastric Acid/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mucous Membrane/cytology , Mucous Membrane/metabolism , Mucous Membrane/physiology
13.
Dig Dis Sci ; 47(5): 967-73, 2002 May.
Article in English | MEDLINE | ID: mdl-12018922

ABSTRACT

Gastroduodenal mucus may play a critical role in defending the epithelium from luminal acid and in the creation of a microenvironment suitable for H. pylori. We measured transmucus permeation of H+, HCO3-, and CO2 with an in vitro perfusion chamber through freshly harvested or partially purified porcine gastric mucin. pH and CO2 concentrations were measured with selective ion electrodes; HCO3- and CO2 concentrations were derived. Viscosity was measured by rotational microviscometry. Mucin viscosity was directly related to concentration. There was a large variation in viscosity among native mucus from antrum, corpus, and duodenum. The highest viscosity was found in the antral mucus; duodenal mucus had the lowest. Diffusion coefficients of duodenal mucus for H+ and HCO3- were significantly lower than those from corpus and antrum. CO2 diffusion coefficients were invariant. In conclusion, despite large variations in viscosity, antral and corpus gastric mucus were similar in terms of ion diffusion. Surprisingly, the low viscosity duodenal mucus was a more potent barrier to ion diffusion than was gastric mucus. Consequently, duodenal mucus may play a more important role in inhibiting ion diffusion than its gastric counterpart.


Subject(s)
Bicarbonates/metabolism , Carbon Dioxide/metabolism , Duodenum/metabolism , Gastric Mucosa/metabolism , Hydrogen/metabolism , Intestinal Mucosa/metabolism , Animals , Diffusion , Hydrogen-Ion Concentration , In Vitro Techniques , Mucins/metabolism , Pyloric Antrum/metabolism , Swine , Viscosity
14.
J Clin Invest ; 108(12): 1807-16, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11748264

ABSTRACT

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.


Subject(s)
Bicarbonates/metabolism , Cytoprotection , Duodenum/metabolism , Gastric Acid/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Hydrogen-Ion Concentration , Intestinal Mucosa/metabolism , Nitrobenzoates/pharmacology , Rats , Rats, Sprague-Dawley , Sodium-Bicarbonate Symporters/analysis , Staining and Labeling
15.
Am J Physiol Gastrointest Liver Physiol ; 280(6): G1083-92, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352800

ABSTRACT

We studied the role of duodenal cellular ion transport in epithelial defense mechanisms in response to rapid shifts of luminal pH. We used in vivo microscopy to measure duodenal epithelial cell intracellular pH (pH(i)), mucus gel thickness, blood flow, and HCO secretion in anesthetized rats with or without the Na(+)/H(+) exchange inhibitor 5-(N,N-dimethyl)-amiloride (DMA) or the anion transport inhibitor DIDS. During acid perfusion pH(i) decreased, whereas mucus gel thickness and blood flow increased, with pH(i) increasing to over baseline (overshoot) and blood flow and gel thickness returning to basal levels during subsequent neutral solution perfusion. During a second brief acid challenge, pH(i) decrease was lessened (adaptation). These are best explained by augmented cellular HCO uptake in response to perfused acid. DIDS, but not DMA, abolished the overshoot and pH(i) adaptation and decreased acid-enhanced HCO secretion. In perfused duodenum, effluent total CO(2) output was not increased by acid perfusion, despite a massive increase of titratable alkalinity, consistent with substantial acid back diffusion and modest CO(2) back diffusion during acid perfusions. Rapid shifts of luminal pH increased duodenal epithelial buffering power, which protected the cells from perfused acid, presumably by activation of Na(+)-HCO cotransport. This adaptation may be a novel, important, and early duodenal protective mechanism against rapid physiological shifts of luminal acidity.


Subject(s)
Bicarbonates/metabolism , Duodenum/metabolism , Intestinal Mucosa/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Acids/metabolism , Acids/pharmacology , Adaptation, Physiological , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Carbon Dioxide/metabolism , Diffusion , Duodenum/drug effects , Hydrogen-Ion Concentration , In Vitro Techniques , Intestinal Mucosa/blood supply , Intestinal Mucosa/drug effects , Rats , Regional Blood Flow/drug effects
16.
Am J Physiol Gastrointest Liver Physiol ; 280(3): G470-4, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11171630

ABSTRACT

We previously showed that the duodenal hyperemic response to acid occurs through activation of capsaicin-sensitive afferent nerves with subsequent release of vasodilatory substances such as calcitonin gene-related peptide (CGRP) and nitric oxide. We then tested the hypothesis that similar factors regulate duodenal mucus gel thickness. Gel thickness was optically measured using in vivo microscopy in anesthetized rats. Duodenal mucosae were superfused with pH 7.0 buffer with vanilloid receptor agonist capsaicin, bradykinin, or PGE(2) injection or were challenged with pH 2.2 solution, with or without the vanilloid antagonist capsazepine, human CGRP-(8-37), N(G)-nitro-L-arginine methyl ester, and indomethacin. Other rats underwent sensory ablation with high-dose capsaicin pretreatment. Acid, bradykinin, capsaicin, and PGE(2) all quickly thickened the gel. Antagonism of vanilloid and CGRP receptors, inhibition of nitric oxide synthase, and sensory deafferentation delayed gel thickening, suggesting that the capsaicin pathway mediated the initial burst of mucus secretion that thickened the gel. Indomethacin abolished gel thickening due to acid, bradykinin, and capsaicin. Inhibition of gel thickening by indomethacin in response to multiple agonists suggests that cyclooxygenase activity is essential for duodenal gel thickness regulation. Duodenal afferent neural pathways play an important role in the modulation of cyclooxygenase-mediated physiological control of gel thickness.


Subject(s)
Capsaicin/analogs & derivatives , Duodenum/enzymology , Intestinal Mucosa/enzymology , Mucus/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Signal Transduction/physiology , Animals , Bradykinin/administration & dosage , Capsaicin/administration & dosage , Cyclooxygenase Inhibitors/administration & dosage , Dinoprostone/administration & dosage , Dose-Response Relationship, Drug , Duodenum/drug effects , Enzyme Inhibitors/administration & dosage , Injections, Intravenous , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mucus/drug effects , Perfusion , Rats , Signal Transduction/drug effects
17.
JOP ; 2(4 Suppl): 268-73, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11875270

ABSTRACT

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.


Subject(s)
Bicarbonates/metabolism , Cystic Fibrosis/metabolism , Duodenal Ulcer/metabolism , Duodenum/metabolism , Intracellular Fluid/metabolism , Peptic Ulcer/metabolism , Animals , Cystic Fibrosis/complications , Duodenal Ulcer/etiology , Duodenum/cytology , Humans , Hydrogen-Ion Concentration , Peptic Ulcer/etiology
18.
Life Sci ; 69(25-26): 3073-81, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11758832

ABSTRACT

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.


Subject(s)
Duodenum/physiology , Gastric Acid/physiology , Intestinal Mucosa/physiology , Animals , Bicarbonates/metabolism , Epithelium/physiology , Humans , Hydrogen-Ion Concentration , Intestinal Mucosa/blood supply , Microcirculation/physiology , Mucus/physiology , Rats
19.
J Gastrointest Surg ; 4(5): 458-63, 2000.
Article in English | MEDLINE | ID: mdl-11077319

ABSTRACT

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.


Subject(s)
Gastric Mucosa/pathology , Hypertension, Portal/pathology , Animals , Epithelium/pathology , Gastric Mucosa/blood supply , Hydrogen-Ion Concentration , Rats , Rats, Sprague-Dawley , Regional Blood Flow
20.
Am J Physiol Gastrointest Liver Physiol ; 279(2): G437-47, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915654

ABSTRACT

We examined the dynamic regulation of mucus gel thickness (MGT) in vivo in rat duodenum in response to luminal acid, cyclooxygenase (COX) inhibition, and exogenous PGE(2). An in vivo microscopic technique was used to measure MGT with fluorescent microspheres in urethan-anesthetized rats. Duodenal mucosa was topically superfused with pH 7.0 or pH 2.2 solutions with or without PGE(2) and indomethacin treatments. Glycoprotein concentration of duodenal loop perfusates was measured with periodic acid/Schiff (PAS) or Alcian blue (AB) staining. MGT and perfusate glycoprotein concentration were stable during a 35-min perfusion with pH 7.0 solution. Acid exposure increased MGT and PAS- and AB-positive perfusate glycoprotein concentrations. Indomethacin pretreatment increased both PAS- and AB-positive perfusate glycoprotein at baseline; subsequent acid superfusion decreased perfusate glycoproteins and gel thickness. PGE(2) (1 mg/kg iv) simultaneously increased MGT and PAS-positive perfusate glycoprotein concentrations followed by a transient increase in AB-positive glycoprotein concentration, suggesting contributions from goblet cells and Brunner's glands. Parallel changes in MGT and perfusate glycoprotein concentration in response to luminal acid and PGE(2) suggest that rapid MGT variations reflect alterations in the balance between mucus secretion and exudation, which in turn are regulated by a COX-related pathway. Luminal acid and PGE(2) augment mucus secretion from goblet cells and Brunner's glands.


Subject(s)
Brunner Glands/enzymology , Duodenum/enzymology , Mucus/metabolism , Acids/pharmacology , Alcian Blue , Animals , Brunner Glands/chemistry , Brunner Glands/cytology , Coloring Agents , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Duodenum/chemistry , Duodenum/cytology , Fluoresceins , Fluorescent Dyes , Frozen Sections , Glycoproteins/analysis , Glycoproteins/metabolism , Goblet Cells/enzymology , Hydrogen-Ion Concentration , Immunoblotting , Indomethacin/pharmacology , Male , Microspheres , Mucous Membrane/chemistry , Mucous Membrane/cytology , Mucous Membrane/enzymology , Mucus/drug effects , Periodic Acid-Schiff Reaction , Polyvinyls , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...