Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
1.
Acta Physiol (Oxf) ; 201(1): 141-50, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20726845

ABSTRACT

AIMS: Apelin peptides are the endogenous ligand of the G protein-coupled receptor APJ. Proposed actions include involvement in control of cardiovascular functions, appetite and body metabolism. We have investigated the effects of apelin peptides on duodenal bicarbonate secretion in vivo and the release of cholecystokinin (CCK) from acutely isolated mucosal cells and the neuroendocrine cell line STC-1. METHODS: Lewis × Dark Agouti rats had free access to water and, unless fasted overnight, free access to food. A segment of proximal duodenum was cannulated in situ in anaesthetized animals. Mucosal bicarbonate secretion was titrated (pH stat) and apelin was administered to the duodenum by close intra-arterial infusion. Total RNA was extracted from mucosal specimens, reverse transcripted to cDNA and the expression of the APJ receptor measured by quantitative real-time PCR. Apelin-induced release of CCK was measured using (1) cells prepared from proximal small intestine and (2) STC-1 cells. RESULTS: Even the lowest dose of apelin-13 (6 pmol kg⁻¹ h⁻¹) caused a significant rise in bicarbonate secretion. Stimulation occurred only in continuously fed animals and even a 100-fold greater dose (600 pmol kg⁻¹ h⁻¹) of apelin was without effect in overnight food-deprived animals. Fasting also induced an eightfold decrease in the expression of APJ receptor mRNA. Apelin induced significant release of CCK from both mucosal and STC-1 cells, and the CCK(A) receptor antagonist devazepide abolished bicarbonate secretory responses to apelin. CONCLUSION: Apelin-induced stimulation of duodenal electrolyte secretion is feeding-dependent and mediated by local mucosal release of CCK.


Subject(s)
Bicarbonates/metabolism , Cholecystokinin/metabolism , Duodenum/drug effects , Duodenum/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Animals , Apelin Receptors , Atropine/pharmacology , Cell Line , Cholecystokinin/antagonists & inhibitors , Devazepide/pharmacology , Dinoprostone/pharmacology , Eating , Food Deprivation/physiology , Humans , Infusions, Intra-Arterial , Intestinal Mucosa/cytology , Male , Parasympatholytics/pharmacology , Rats , Rats, Inbred Lew , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Tryptamines/pharmacology
2.
Acta Physiol (Oxf) ; 198(3): 373-80, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20003099

ABSTRACT

Studies of gastrointestinal physiology in humans and intact animals are usually conducted after overnight fast. We compared the effects of orexin-A, vasoactive intestinal polypeptide (VIP), melatonin, serotonin, uroguanylin, ghrelin and prostaglandin E(2) (PGE(2)) on duodenal bicarbonate secretion in fed and overnight fasted animals. This review is a summary of our findings. Secretagogues were administered by intra-arterial infusion or luminally (PGE(2)). Enterocyte intracellular calcium ([Ca(2+)](i)) signalling was studied by fluorescence imaging. Total RNA was extracted, reverse transcripted to cDNA and expression of orexin receptors measured by quantitative real-time PCR. Orexin-A stimulates the duodenal secretion in continuously fed animals but not in food-deprived animals. Similarly, short-term fasting causes a 100-fold decrease in the amount of the muscarinic agonist bethanechol required for stimulation of secretion. In contrast, fasting does not affect secretory responses to intra-arterial VIP, melatonin, serotonin, uroguanylin and ghrelin, or that to luminal PGE(2). Orexin-A induces [Ca(2+)](i) signalling in enterocytes from fed rats but no significant [Ca(2+)](i) responses occur in enterocytes from fasted animals. In addition, overnight fasting decreases the expression of mucosal orexin receptors. Short-term food deprivation thus decreases duodenal expression of orexin receptors and abolishes the secretory response to orexin-A as well as orexin-A-induced [Ca(2+)](i) signalling. Fasting, furthermore, decreases mucosal sensitivity to bethanechol. The absence of declines in secretory responses to other secretagogues tested strongly suggests that short-term fasting does not affect the secretory capacity of the duodenal mucosa in general. Studies of intestinal secretion require particular evaluation with respect to feeding status.


Subject(s)
Bicarbonates/metabolism , Duodenum/drug effects , Duodenum/metabolism , Food Deprivation/physiology , Intracellular Signaling Peptides and Proteins/administration & dosage , Neuropeptides/administration & dosage , Neurotransmitter Agents/administration & dosage , Animals , Dinoprostone/administration & dosage , Gastrointestinal Tract/innervation , Ghrelin/administration & dosage , Humans , Infusions, Intra-Arterial , Melatonin/administration & dosage , Natriuretic Peptides/administration & dosage , Orexin Receptors , Orexins , Peripheral Nervous System/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Serotonin/administration & dosage , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Time Factors , Vasoactive Intestinal Peptide/administration & dosage
3.
Acta Physiol (Oxf) ; 191(4): 309-17, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17995576

ABSTRACT

AIM: Uroguanylin and guanylin are endogenous ligands for guanylate cyclase C, an upstream regulator of the cystic fibrosis transmembrane resistance (CFTR) anion channel, and both peptides increase intestinal anion export in vitro. We have compared the effects of close intra-arterial and luminal administration of uroguanylin and guanylin on duodenal bicarbonate secretion in vivo and studied the interactions with melatonin and cholinergic stimulation. METHODS: Lewis x Dark Agouti rats were anaesthetized and a segment of the proximal duodenum with intact blood supply was cannulated in situ. Mucosal bicarbonate secretion (pH stat) was continuously recorded and peptides were infused intra-arterially or added to the luminal perfusate. RESULTS: Intra-arterial (50-1000 pmol kg(-1) h(-1)) as well as luminal administration (50-500 nmol L(-1)) of guanylin or uroguanylin caused dose-dependent increases in the duodenal secretion. Luminal administration induced more rapidly appearing rises in secretion and the two peptides induced secretory responses of similar shape and magnitude. The melatonin MT(2)-selective antagonist luzindole (600 nmol kg(-1)) significantly depressed the response to intra-arterial guanylins but did not affect secretion induced by luminal guanylins. Similarly, the muscarinic antagonist atropine (0.75 micromol kg(-1) followed by 0.15 micromol kg(-1) h(-1)) abolished the response to intra-arterial uroguanylin but caused only slight suppression of the response to luminal uroguanylin. CONCLUSIONS: Intra-arterial as well as luminal uroguanylin and guanylin are potent stimuli of duodenal mucosal bicarbonate secretion in vivo. The response to luminal guanylins reflects an action at apical receptors. Stimulation by parenteral guanylins, in contrast, is under cholinergic influence and interacts with melatonin produced by mucosal enteroendocrine cells.


Subject(s)
Arteries/metabolism , Bicarbonates/metabolism , Cytoprotection/physiology , Duodenum/metabolism , Gastrointestinal Hormones/metabolism , Intestinal Mucosa/metabolism , Natriuretic Peptides/metabolism , Animals , Duodenum/drug effects , Intestinal Mucosa/drug effects , Male , Rats , Rats, Inbred Lew
4.
J Clin Invest ; 108(4): 625-33, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11518737

ABSTRACT

Melatonin is released from intestinal enterochromaffin cells and from the pineal gland, but its role in gastrointestinal function is largely unknown. Our aim was to study the involvement of intestinal and central nervous melatonin in the neurohumoral control of the duodenal mucosa-protective bicarbonate secretion. Working in anesthetized rats, we cannulated a 12-mm segment of duodenum with an intact blood supply and titrated the local bicarbonate secretion with pH-stat. Melatonin and receptor ligands were supplied to the duodenum by close intra-arterial infusion. Even at low doses, melatonin and the full agonist 2-iodo-N-butanoyl-5-methoxytryptamine increased duodenal bicarbonate secretion. Responses were inhibited by the predominantly MT2-selective antagonist luzindole but not by prazosin, acting at MT3 receptors. Also, luzindole almost abolished the marked rise in secretion induced by intracerebroventricular infusion of the adrenoceptor agonist phenylephrine. This response was also abolished by sublaryngeal ligation of all nerves around the carotid arteries. However, it was insensitive to truncal vagotomy alone or sympathectomy alone and was unaffected by removal of either the pineal gland or pituitary gland. Thus, melatonin stimulates duodenal bicarbonate secretion via action at enterocyte MT2-receptors and mediates neural stimulation of the secretion.


Subject(s)
Bicarbonates/metabolism , Duodenum/metabolism , Enterochromaffin Cells/metabolism , Intestinal Mucosa/metabolism , Melatonin/physiology , Neurosecretion/physiology , Adrenocorticotropic Hormone/administration & dosage , Adrenocorticotropic Hormone/pharmacology , Animals , Blood Pressure/drug effects , Circadian Rhythm , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/pharmacology , Duodenum/drug effects , Hypophysectomy , Injections, Intra-Arterial , Injections, Intraventricular , Intestinal Mucosa/drug effects , Melatonin/administration & dosage , Melatonin/antagonists & inhibitors , Melatonin/pharmacology , Neurosecretion/drug effects , Phenylephrine/administration & dosage , Phenylephrine/pharmacology , Pineal Gland/surgery , Prazosin/administration & dosage , Prazosin/pharmacology , Protein Isoforms/drug effects , Protein Isoforms/physiology , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/physiology , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Melatonin , Tetrahydronaphthalenes/administration & dosage , Tetrahydronaphthalenes/pharmacology , Thiopental/administration & dosage , Thiopental/analogs & derivatives , Thiopental/pharmacology , Tryptamines/administration & dosage , Tryptamines/pharmacology , Vagotomy , gamma-MSH/administration & dosage , gamma-MSH/pharmacology
5.
News Physiol Sci ; 16: 23-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11390942

ABSTRACT

The gastroduodenal mucosa is a dynamic barrier restricting entry of gastric acid and other potentially hostile luminal contents. Mucosal HCO3(-) is a key element in preventing epithelial damage, and knowledge about HCO3(-) transport processes, including the role of the cystic fibrosis transmembrane conductance regulator channel, and their neurohumoral control are in rapid progress.


Subject(s)
Alkalies/metabolism , Bicarbonates/metabolism , Duodenum/metabolism , Gastric Mucosa/metabolism , Intestinal Mucosa/metabolism , Animals , Humans
6.
J Physiol Pharmacol ; 52(4 Pt 1): 671-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11787766

ABSTRACT

A number of common diseases in humans, including gastroduodenal ulcer and irritable bowel syndrome, show circadian rhythms in pain and discomfort. The neurohormone melatonin is released from enterochromaffin cells in the intestinal mucosa and from the pineal gland but its role in gastrointestinal function is largely unknown. We have studied the involvement of melatonin in stimulation of the mucosa-protective alkaline secretion by the duodenal mucosa. A 12-mm segment of proximal duodenum with an intact blood supply was cannulated in situ in anesthetized rats and duodenal HCO3- secretion titrated by pH-stat. Duodenal close intra-arterial infusion of melatonin or the full agonist 2-iodo-N-butanoyl- 5-methoxytryptamine significantly increased the secretion and pretreatment with the melatonin (predominantly MT2-receptor specific) antagonist luzindole almost abolished the response. Intracerebroventricular (i.c.v.) infusion of the alpha1-adrenoceptor agonist phenylephrine (12.2 micromol kg(-1) x h(-1)) caused an up to fivefold increased in the alkaline secretion and the melatonin antagonist luzindole or cutting all peri-carotid nerves abolished the duodenal secretory response to i.c.v. phenylephrine. Peripheral melatonin thus stimulates duodenal mucosal HCO3- secretion and endogenous melatonin, very likely released from mucosal enterochromaffin cells, is involved in mediating neural stimulation of the secretion.


Subject(s)
Bicarbonates/metabolism , Brain/physiology , Duodenum/drug effects , Melatonin/pharmacology , Animals , Brain/drug effects , Duodenum/metabolism , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Melatonin
7.
Acta Physiol Scand ; 169(3): 237-47, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10886038

ABSTRACT

After abdominal surgery, luminal HCl fails to induce duodenal contractions in anaesthetized rats. Elevated tissue levels of nitric oxide (NO) and prostaglandins possibly contribute to this observation. The aim of this study was to compare the effects of luminal capsaicin (1.2 mg mL-1), ethanol (15%) and high partial pressure of CO2 (>250 mmHg) with those of HCl (10 mM) in anaesthetized rats. Motility (intraluminal pressure), mucosal permeability [blood-to-lumen clearance of 51Cr-EDTA (51Chromium-labelled ethylenediaminetetraacetate)] and duodenal mucosal bicarbonate secretion (DMBS) were recorded. Three groups of animals were studied: (1) controls, (2) pretreatment with the NO synthase inhibitor N-nitro-L-arginine-methyl-ester (L-NAME) and (3) pretreatment with the cyclo-oxygenase inhibitor indomethacin. Neither capsaicin, ethanol, CO2 nor HCl induced duodenal contractions or affected DMBS in control rats. However, L-NAME induced duodenal contractions that were augmented by capsaicin, ethanol and HCl, but not by CO2. Indomethacin also induced contractions that were reversibly diminished by capsaicin and HCl, but not by ethanol or CO2. Significant increases in mucosal permeability occurred during ethanol perfusion in indomethacin- and L-NAME pretreated rats. In conclusion, NO probably plays a key role in preventing duodenal contractions in response to luminally HCl, capsaicin and ethanol. The HCl-induced effect on motility appears to be independent of CO2 and is not caused by alteration in mucosal integrity.


Subject(s)
Duodenum/drug effects , Gastrointestinal Motility/drug effects , Muscle Contraction/drug effects , Nitric Oxide/pharmacology , Animals , Arginine/metabolism , Bicarbonates/metabolism , Capsaicin/administration & dosage , Carbon Dioxide/administration & dosage , Chromium Radioisotopes , Crosses, Genetic , Edetic Acid/pharmacokinetics , Ethanol/administration & dosage , Hydrochloric Acid/administration & dosage , Indomethacin/administration & dosage , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intubation, Gastrointestinal , Male , Metabolic Clearance Rate/drug effects , NG-Nitroarginine Methyl Ester/administration & dosage , Perfusion/methods , Rats , Rats, Inbred Lew , Rats, Inbred Strains
8.
Scand J Gastroenterol ; 34(11): 1093-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10582759

ABSTRACT

BACKGROUND: The effects of gamma-aminobutyric (GABA(A))-receptor ligands and peripheral-type benzodiazepine (BZ) ligands on duodenal mucosal bicarbonate secretion (DBS) were studied in thiobarbiturate-anesthetized rats. METHODS: A segment of proximal duodenum was perfused, and bicarbonate secretion was continuously titrated by pH-stat. In some experiments the vagus nerves were dissected free and cut at the neck level. RESULTS: Luminal GABA (10(-8) to 10(-6) M) increased (P < 0.01) DBS dose-dependently, and the GABA(A) antagonist (+)-bicuculline (0.1-2.5 mg/kg intravenously) caused a similar increase (P < 0.01) in DBS. Vagotomy abolished the latter response, suggesting a centrally elicited nervous action of bicuculline mediated by the vagal nerves. This was supported by the absence of an effect of bicuculline administered intra-arterially close to the duodenum. The GABA(A) agonist muscimol had no effect on DBS when administered intravenously (0.01-0.25 mg/kg) or into a lateral brain ventricle (2 microg/kg/h) but counteracted any stimulation by subsequent intravenous bicuculline. The 'peripheral-type' BZ agonist 4'-chlorodiazepam increased DBS when infused close intra-arterially but had no effect when administered intravenously. CONCLUSIONS: Inhibition of GABA(A) receptors related to cholinergic excitatory pathways in the central nervous system stimulates duodenal mucosal bicarbonate secretion and may increase the local mucosal defense. The stimulation by luminal GABA may reflect modulation of the local mucosal control of duodenal bicarbonate secretion.


Subject(s)
Benzodiazepines/pharmacology , Bicarbonates/metabolism , Duodenum , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , gamma-Aminobutyric Acid/pharmacology , Analysis of Variance , Animals , Antineoplastic Agents/pharmacology , Bicuculline/pharmacology , Diazepam/pharmacology , Female , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , GABA Modulators/pharmacology , Isoquinolines/pharmacology , Ligands , Male , Muscimol/pharmacology , Rats , Rats, Inbred Lew
9.
Am J Physiol ; 277(2): G375-82, 1999 08.
Article in English | MEDLINE | ID: mdl-10444452

ABSTRACT

The aim of this study was to investigate the permeability of the adherent mucus gel layer in rat duodenum in vivo to macromolecules applied in the lumen. Rats were anesthetized with thiobarbiturate, and the duodenum was perfused with isotonic NaCl solution containing large-molecular-size secretagogues. Effects on mucosal HCO(-)(3) secretion and blood-to-lumen (51)chromium-labeled EDTA clearance were used as indexes that compounds had migrated across the mucus layer. Exposure to a low concentration of papain (10 U/100 ml) for 30 min removed the mucus layer without damage to the epithelium and induced or markedly enhanced HCO(-)(3) secretory responses to cholera toxin (molecular mass of 85 kDa) or glucagon (3.5 kDa). Water extracts from a VacA cytotoxin (89 kDa) producing Helicobacter pylori strain, but not from a toxin-negative isogenic mutant, caused a small increase in HCO(-)(3) secretion but only after the mucus layer had been removed by papain. The duodenal surface mucus gel thus significantly restricts migration of macromolecules to the duodenal surface. Release of bacterial toxins at the cell-mucus interface may enhance or be a prerequisite for their effects on the gastrointestinal mucosa.


Subject(s)
Duodenum/metabolism , Mucus/physiology , Animals , Cholera Toxin/pharmacology , Diffusion , Dinoprostone/pharmacology , Duodenum/drug effects , Gels , Glucagon/pharmacology , Helicobacter pylori/chemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Macromolecular Substances , Male , Rats , Rats, Inbred Lew , Rats, Inbred Strains , Tissue Extracts/pharmacology
10.
Am J Physiol ; 275(2): G296-304, 1998 08.
Article in English | MEDLINE | ID: mdl-9688657

ABSTRACT

Vagal stimuli increase duodenal mucosal HCO-3 secretion and may provide anticipatory protection against acid injury, but duodenal enterocyte (duodenocyte) responses and cholinoceptor selectivity have not been defined. We therefore developed a stable primary culture model of duodenocytes from rats and humans. Brief digestion of scraped rat duodenal mucosa or human biopsies with collagenase/dispase yielded cells that attached to the extracellular matrix Matrigel within a few hours of plating. Columnar cells with villus enterocyte morphology that exhibited spontaneous active movement were evident between 1 and 3 days of culture. Rat duodenocytes loaded with fura 2 responded to carbachol with a transient increase in intracellular calcium concentration ([Ca2+]i), with an apparent EC50 of approximately 3 microM. In a first type of signaling pattern, [Ca2+]i returned to basal or near basal values within 3-5 min. In a second type, observed in cells with enlarged vacuoles characteristic of crypt cell morphology, the initial transient increase was followed by rhythmic oscillations. Human duodenocytes responded with a more sustained increase in [Ca2+]i, and oscillations were not observed. Rat as well as human duodenocytes also responded to CCK-octapeptide but not to vasoactive intestinal polypeptide. Equimolar concentrations (100 nM) of the subtype-independent muscarinic antagonist atropine and the M3 antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide prevented the response to 10 microM carbachol, whereas the M1 antagonist pirenzepine and the M2 antagonists methoctramine and AF-DX 116BS had no effect at similar concentrations. Responses in rat and human duodenocytes were similar. A new agonist-sensitive primary culture model for rat and human duodenocytes has thus been established and the presence of enterocyte CCK and muscarinic M3 receptors demonstrated.


Subject(s)
Calcium/metabolism , Carbachol/pharmacology , Intestinal Mucosa/physiology , Signal Transduction , Animals , Cell Adhesion , Cells, Cultured , Dose-Response Relationship, Drug , Duodenum , Extracellular Matrix/physiology , Fluorescent Dyes , Fura-2 , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Male , Parasympatholytics/pharmacology , Piperidines/pharmacology , Rats , Rats, Inbred Lew , Sincalide/pharmacology , Vacuoles/drug effects , Vacuoles/physiology , Vacuoles/ultrastructure
11.
Am J Physiol ; 275(1): G95-103, 1998 07.
Article in English | MEDLINE | ID: mdl-9655689

ABSTRACT

Neurokinin A (NKA) induces duodenal motility and increases mucosal permeability and bicarbonate secretion in the in situ perfused duodenum in anesthetized rats. In the present study, the NKA-induced increase in mucosal permeability was potentiated by luminal perfusion with lidocaine and diminished by vasoactive intestinal peptide (VIP) but unaltered by elevated intraluminal pressure. Elevation of intraluminal pressure, however, potentiated the stimulatory effect of NKA on bicarbonate secretion. In contrast, the tachykinin decreased the rate of alkalinization in rats subjected to elevated intraluminal pressure and treated with indomethacin. Similarly, NKA partially inhibited the VIP-stimulated bicarbonate secretion. Luminal lidocaine did not affect the secretory response to NKA. The motility induced by NKA was unaffected by VIP or lidocaine but decreased by elevated intraluminal pressure. It is concluded that the NKA-induced increase in duodenal mucosal bicarbonate secretion is independent of neurons and possibly mediated by prostanoids. The increase in mucosal permeability in response to NKA may be suppressed by mucosal nerves, perhaps utilizing VIP as one of the transmitters.


Subject(s)
Duodenum/physiology , Gastrointestinal Motility/physiology , Intestinal Absorption/physiology , Intestinal Mucosa/physiology , Muscle, Smooth/physiology , Neurokinin A/pharmacology , Prostaglandins/physiology , Vasoactive Intestinal Peptide/pharmacology , Analysis of Variance , Animals , Bicarbonates/metabolism , Chromium Radioisotopes/pharmacokinetics , Duodenum/drug effects , Duodenum/innervation , Edetic Acid/pharmacokinetics , Gastrointestinal Motility/drug effects , Indomethacin/pharmacology , Infusions, Intravenous , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Lidocaine/administration & dosage , Lidocaine/pharmacology , Male , Metabolic Clearance Rate , Muscle, Smooth/drug effects , Muscle, Smooth/innervation , Neurokinin A/administration & dosage , Neurons/drug effects , Neurons/physiology , Pressure , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/physiology , Vasoactive Intestinal Peptide/administration & dosage
13.
Am J Physiol ; 273(5): G1077-86, 1997 11.
Article in English | MEDLINE | ID: mdl-9374705

ABSTRACT

The aim of this study was to examine the integrative response to neurokinin A (NKA) on duodenal mucosal permeability, bicarbonate secretion, fluid flux, and motility in an in situ perfusion model in anesthetized rats. Intravenous infusion of NKA (100, 200, and 400 pmol.kg-1.min-1) induced duodenal motility. Furthermore, duodenal mucosal bicarbonate secretion, fluid output, and mucosal permeability increased in response to NKA. Pretreatment with the nicotinic antagonist hexamethonium did not change the response in any of the parameters investigated, whereas the NK2-receptor antagonist MEN 10,627 effectively inhibited all responses to NKA. Indomethacin induced duodenal motility and stimulated bicarbonate secretion. In indomethacin-treated rats, NKA further increased motility but decreased indomethacin-stimulated bicarbonate secretion by 70%. The NKA-induced increase in mucosal permeability was unaltered by indomethacin. It is concluded that NKA not only induces motility but also increases mucosal permeability and fluid output. Furthermore, the neuropeptide may have both stimulative and inhibitory effects on bicarbonate secretion. All responses to NKA are dependent on NK-2 receptor activation but are not mediated through nicotinic receptors.


Subject(s)
Bicarbonates/metabolism , Duodenum/physiology , Gastrointestinal Motility/drug effects , Intestinal Mucosa/physiology , Neurokinin A/pharmacology , Animals , Chromium Radioisotopes/pharmacokinetics , Duodenum/drug effects , Edetic Acid/pharmacokinetics , Hexamethonium/pharmacology , Indomethacin/pharmacology , Infusions, Intravenous , Intestinal Mucosa/drug effects , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Neurokinin A/administration & dosage , Peptides, Cyclic/pharmacology , Permeability , Pressure , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-2/antagonists & inhibitors , Time Factors
14.
Am J Physiol ; 271(5 Pt 1): G831-40, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8944698

ABSTRACT

We have studied the effects of intracerebral administration of selective alpha-adrenergic agonists on duodenal bicarbonate secretion. Duodenum free of Brunner's glands was cannulated in situ in anesthetized rats, and bicarbonate secretion into the luminal reperfusate was continuously titrated by pH stat. Infusion of the alpha 1-selective adrenoceptor agonist, phenylephrine (1,000-2,500 micrograms.kg-1.h-1), into a lateral brain ventricle increased (P < 0.01) duodenal bicarbonate secretion. Pretreatment with prazosin, an alpha 1-antagonist, significantly (P < 0.01) reduced the stimulatory effect when infused into the lateral ventricle (30 micrograms.kg-1.h-1), but not when administered intravenously (1,000 micrograms.kg-1.h-1). Hexamethonium (10 mg.kg-1.h-1 iv) abolished stimulation, whereas cervical vagotomy, epidural blockade, and naloxone were each without effect. Vasopressin, vasopressin antagonists, ts, and oxytocin did not affect basal secretion. Intracerebro-ventricular administration of the alpha 2-adrenoceptor agonist, clonidine (1,000 micrograms.kg-1.h-1), in contrast to alpha 1-receptor activation, decreased (P < 0.01) the secretion. Thus central nervous adrenoceptors influence duodenal mucosal bicarbonate te secretion, and alpha 1-adrenoceptor stimulation may provide protection against luminal acid. This potent stimulation was not mediated by the vagal nerves, spinal cord pathways, or the release of beta-endorphin but involves nicotinic, possibly enteric nervous transmission.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Bicarbonates/metabolism , Cerebral Ventricles/physiology , Duodenum , Intestinal Mucosa/metabolism , Phenylephrine/pharmacology , Adrenergic alpha-Agonists/administration & dosage , Adrenergic alpha-Antagonists/pharmacology , Animals , Cerebral Ventricles/drug effects , Hexamethonium/pharmacology , Infusions, Parenteral , Intestinal Mucosa/drug effects , Kinetics , Male , Naloxone/pharmacology , Phenylephrine/administration & dosage , Prazosin/administration & dosage , Prazosin/pharmacology , Rats , Rats, Sprague-Dawley
15.
Am J Physiol ; 269(2 Pt 1): G246-54, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7653565

ABSTRACT

This study examines the integrative response of several duodenal functions to nitric oxide synthase inhibition. Effects of the nitric oxide synthase inhibitor N-nitro-L-arginine methyl ester (L-NAME) were studied in anesthetized rats, using in situ duodenal perfusion. L-NAME increased bicarbonate secretion, permeability, and fluid secretion and induced motility. Injection of L-arginine abolished L-NAME-induced motility and lowered the secretion of bicarbonate and fluid. Pretreatment with the nicotinic receptor antagonist hexamethonium prevented the rise in bicarbonate secretion and motility in response to L-NAME but did not affect the increase in mucosal permeability. Atropine diminished the L-NAME-induced increases in permeability, motility, and fluid secretion. The adrenolytic drug guanethidine did not alter the responses to the inhibitor. These results suggest that nitric oxide inhibits duodenal motility and bicarbonate secretion by suppressing a stimulatory, nicotinic receptor-dependent, neural mechanism. The L-NAME-induced contractions involve both a cholinergic, atropine-sensitive pathway and nonadrenergic, noncholinergic neural transmission. Muscarinic receptors also mediate part of the L-NAME-induced increases in mucosal permeability and fluid secretion.


Subject(s)
Duodenum/innervation , Duodenum/physiology , Nitric Oxide/antagonists & inhibitors , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Atropine/pharmacology , Bicarbonates/metabolism , Duodenum/drug effects , Gastrointestinal Motility/drug effects , Guanethidine/pharmacology , Hexamethonium/pharmacology , Intestinal Mucosa/metabolism , Male , NG-Nitroarginine Methyl Ester , Nervous System Physiological Phenomena , Permeability/drug effects , Rats , Rats, Sprague-Dawley
16.
Dig Dis Sci ; 39(9): 1839-42, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8082488

ABSTRACT

Duodenal mucosal secretion of bicarbonate is one main mechanism in the protection of this epithelium against luminal acid. The duodenal secretagogue vasoactive intestinal polypeptide, at doses not affecting mucosal blood flow, protects against acid-induced morphological changes. Some sigma receptor ligands, which increase the duodenal alkaline secretion, prevent duodenal but not gastric mucosal ulceration. Dopamine D-1 receptor agonists and the peripherally acting catechol-O-methyl-transferase (COMT) inhibitor nitecapone stimulate the bicarbonate secretion in the rat and a similar increase in secretion has been observed in human volunteers. COMT inhibitors decrease tissue degradation of catecholamines, including dopamine. The D-2 agonist bromocriptine, in contrast, decreases the secretion. These results, indicating that the bicarbonate secretion is stimulated via peripheral dopamine D-1 receptors, are supported by the finding that dopamine D-1 (but not D-2) agonists increase the production of cyclic AMP in isolated duodenal enterocytes. The increase in mucosal alkaline secretion may contribute to the previously observed ulceroprotective actions of dopaminergic compounds.


Subject(s)
Bicarbonates/metabolism , Dopamine/physiology , Duodenum/physiology , Intestinal Mucosa/metabolism , Animals , Dopamine/pharmacology , Duodenum/drug effects , Humans , Intestinal Mucosa/drug effects , Receptors, Dopamine/metabolism
17.
Gastroenterology ; 107(1): 180-8, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8020660

ABSTRACT

BACKGROUND/AIMS: The gastric mucosa is covered by a continuous layer of bicarbonate-containing mucus gel; the question arises how acid, formed in the gastric glands, moves into the lumen. METHODS: The pH in the gastric mucus gel and gel thickness were measured in anesthetized rats with pH-sensitive microelectrodes (tip diameter, 1-5 microns). RESULTS: During pentagastrin (40 micrograms.kg-1.h-1) stimulation of acid secretion, the pH was higher in the gel than in the lumen (pH 2) up to a distance of 115 +/- 18 microns from the epithelial surface and maximal (pH 7.2 +/- 0.1) at the surface. A similar pH gradient was recorded at luminal pH 3. After omeprazole (10 mumol/kg) inhibition of endogenous acid secretion and with exogenous acid in the lumen, the pH profile was broader: 204 +/- 26 microns at luminal pH 2 and 231 +/- 63 microns at luminal pH 3. In contrast, the pH at the epithelial surface was lower (pH 6.8-6.9). The gel thickness (200-300 microns) was similar in all groups. CONCLUSIONS: The significantly higher surface pH in acid-secreting stomachs probably reflects better availability of interstitial mucosal bicarbonate. Bulk transport of secreted acid in channels created by the gland luminal hydrostatic pressure may additionally act to limit acidification of the mucus gel.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/chemistry , Gastric Mucosa/physiology , Hydrogen-Ion Concentration , Omeprazole/pharmacology , Pentagastrin/pharmacology , Animals , Biological Transport/physiology , Congo Red , Gastric Acid/physiology , Gastric Mucosa/metabolism , Male , Microelectrodes , Rats , Rats, Sprague-Dawley
18.
Am J Physiol ; 267(1 Pt 1): G10-7, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8048521

ABSTRACT

Bicarbonate secretion by duodenum distal to the Brunner's glands area was titrated in situ in rats anesthetized with thiobarbiturate. The unselective muscarinic antagonist atropine (0.4 mg/kg) inhibited secretion stimulated by bethanechol (15 micrograms.kg-1.h-1) but not that stimulated by carbachol (15 micrograms.kg-1.h-1). The nicotinic antagonist hexamethonium (10 mg.kg-1.h-1), however, abolished the latter response. The muscarinic M1-selective antagonists pirenzepine and telenzepine (0.025, 0.25 and 2.5 mg/kg) did not decrease but caused a dose-dependent rise in duodenal mucosal HCO3- secretion, an effect abolished by cervical vagotomy or infusion of the alpha-adrenoceptor antagonist phentolamine (0.1 mg.kg-1.h-1). Phentolamine alone caused a sustained increase in secretion. McN-A-343 (0.025, 0.25, and 2.5 mg/kg), an M1-selective agonist and ganglionic stimulator, increased the HCO3- secretion; this effect was not prevented by vagotomy but was attenuated by pirenzepine. Intracerebroventricular infusion of pirenzepine and telenzepine did not cause any changes in secretion. These findings suggest that peripheral muscarinic M1 and nicotinic receptors mediate cholinergic stimulation of duodenal mucosal HCO3- secretion. Pirenzepine and telenzepine may act stimulatory by antagonizing muscarinic M1-transmission in peripheral sympathetic ganglia, thus decreasing postsynaptic adrenergic inhibition.


Subject(s)
Bicarbonates/metabolism , Duodenum/metabolism , Intestinal Mucosa/metabolism , Parasympatholytics/pharmacology , Parasympathomimetics/pharmacology , Anesthesia , Animals , Injections, Intravenous , Injections, Intraventricular , Male , Phentolamine/pharmacology , Rats , Rats, Sprague-Dawley , Sympatholytics/pharmacology , Vagotomy
19.
Am J Physiol ; 265(4 Pt 1): G677-85, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8238351

ABSTRACT

The duodenum, in contrast to the jejunum, actively secretes HCO3- at a high rate, a process that protects the mucosa from acid/peptic injury. Our purpose was to define the mechanisms involved in HCO3- transport by studying the acid-base transport processes in isolated duodenal enterocytes. Individual rat duodenocytes, isolated by a combination of Ca2+ chelation and collagenase, attached to a collagen matrix were loaded with the pH-sensitive fluoroprobe 2',7'-bis(2-carboxyethyl)-5(6)-carboxyfluorescein acetoxymethyl ester (BCECF-AM), and intracellular pH was monitored by microfluorospectrophotometry. To identify Na(+)-H+ transport, cells in N-2-hydroxyethylpiperazine-N'-2-ethanesulfonic acid 1) were pulsed with NH4Cl (40 mM) in the absence and presence of amiloride and 2) were removed of Na+. To examine Cl(-)-HCO3- exchange, Cl- was removed from Ringer-HCO3- superfusate in the presence and absence of dihydro-4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (H2DIDS). The NaHCO3 cotransporter was studied by addition and subtraction of Na+ to amiloride-treated and Cl(-)-depleted enterocytes perfused with Na(+)- and Cl(-)-free Ringer-HCO3- buffer with and without H2DIDS. Mammalian duodenocytes contain at least three acid-base transporters: an amiloride-sensitive Na(+)-H+ exchanger that extrudes acid, a DIDS-sensitive Cl(-)-HCO3- exchanger that extrudes base, and a NaHCO3 cotransporter, also DIDS sensitive, that functions as a base loader. These acid-base transporters likely play a key role in duodenal mucosal HCO3- secretion.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antiporters/metabolism , Carrier Proteins/metabolism , Duodenum/metabolism , Sodium-Hydrogen Exchangers/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/analogs & derivatives , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Amiloride/pharmacology , Animals , Biological Transport/drug effects , Cell Separation , Cell Survival , Chloride-Bicarbonate Antiporters , Duodenum/cytology , Hydrogen-Ion Concentration , Male , Rats , Rats, Sprague-Dawley , Sodium-Bicarbonate Symporters
20.
Physiol Rev ; 73(4): 823-57, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8415927

ABSTRACT

The barrier that protects the undamaged gastroduodenal mucosa from autodigestion by gastric juice is a dynamic multicomponent system. The major elements of this barrier are the adherent mucus gel layer, which is percolated by the HCO3- secretion from the underlying epithelial cells; the epithelial layer itself, which provides a permeability barrier and can rapidly repair superficial damage by a process of cell migration referred to as reepithelization or restitution; and a specially adapted vasculature, which provides a supply of HCO3- for transcellular transport and/or diffusion into the mucus layer. Passive diffusion of intestinal HCO3- into the lumen is particularly important when there is superficial damage resulting in increased leakiness of the mucosal epithelium. The process of reepithelization occurs by the migration of performed cells from gastric pits or duodenal crypts. This process is quite distinct from the wound healing and associated inflammatory response that accompany more severe injury or chronic damage. The adherent mucus gel acts as a physical barrier against luminal pepsin and provides a stable unstirred layer that supports surface neutralization of acid by mucosal HCO3-. Surface neutralization by mucosal HCO3- provides a major mechanism of protection against acid in the proximal duodenum. In the stomach, where luminal acidity can fall to around pH 1, other mechanisms of protection must exist, since the surface pH gradient is reported to collapse when luminal H+ exceeds approximately 10 mM. This collapse of the surface pH gradients may reflect, at least in part, that such studies have been mostly performed on non-acid-secreting mucosa where the supply of HCO3- to the interstitium from the parietal cells will be reduced. However, because the gastric mucosa can withstand prolonged exposure to acid without apparent damage, this implies an intrinsic resistance of the epithelial apical surface. This is amply illustrated within the gastric glands that do not secrete mucus and HCO3- yet are exposed to undiluted pepsin and an isotonic solution of HCl. Bicarbonate and mucus secretions together with mucosal blood flow are under paracrine, endocrine, and neural control. The rate of reepithelialization will depend on local chemotactic factors, adhesion mechanisms, and the creation of an acid/pepsin/irritant-free environment under a protective gelatinous or mucoid cap. If optimal conditions are met, then the rate of reepithelialization appears to depend primarily on the intrinsic properties of the migrating cells themselves rather than control by exogenous mediators.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Duodenum/physiology , Gastric Mucosa/physiology , Intestinal Mucosa/physiology , Acids/antagonists & inhibitors , Acids/metabolism , Animals , Bicarbonates/metabolism , Cell Division , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Gels , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mucus/chemistry , Pepsin A/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...