Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
1.
Sci Rep ; 14(1): 11720, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38778154

ABSTRACT

We studied the inhibitory actions of docosahexaenoic acid (DHA) on the contractions induced by carbachol (CCh), angiotensin II (Ang II), and bradykinin (BK) in guinea pig (GP) gastric fundus smooth muscle (GFSM), particularly focusing on the possible inhibition of store-operated Ca2+ channels (SOCCs). DHA significantly suppressed the contractions induced by CCh, Ang II, and BK; the inhibition of BK-induced contractions was the strongest. Although all contractions were greatly dependent on external Ca2+, more than 80% of BK-induced contractions remained even in the presence of verapamil, a voltage-dependent Ca2+ channel inhibitor. BK-induced contractions in the presence of verapamil were not suppressed by LOE-908 (a receptor-operated Ca2+ channel (ROCC) inhibitor) but were suppressed by SKF-96365 (an SOCC and ROCC inhibitor). BK-induced contractions in the presence of verapamil plus LOE-908 were strongly inhibited by DHA. Furthermore, DHA inhibited GFSM contractions induced by cyclopiazonic acid (CPA) in the presence of verapamil plus LOE-908 and inhibited the intracellular Ca2+ increase due to Ca2+ addition in CPA-treated 293T cells. These findings indicate that Ca2+ influx through SOCCs plays a crucial role in BK-induced contraction in GP GFSM and that this inhibition by DHA is a new mechanism by which this fatty acid inhibits GFSM contractions.


Subject(s)
Angiotensin II , Bradykinin , Carbachol , Docosahexaenoic Acids , Gastric Fundus , Muscle Contraction , Muscle, Smooth , Animals , Guinea Pigs , Docosahexaenoic Acids/pharmacology , Bradykinin/pharmacology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Muscle, Smooth/metabolism , Carbachol/pharmacology , Muscle Contraction/drug effects , Angiotensin II/pharmacology , Gastric Fundus/drug effects , Gastric Fundus/physiology , Gastric Fundus/metabolism , Verapamil/pharmacology , Calcium/metabolism , Male , Humans , Calcium Channels/metabolism , HEK293 Cells , Calcium Channel Blockers/pharmacology , Imidazoles/pharmacology
2.
Am J Physiol Cell Physiol ; 321(4): C684-C693, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34432539

ABSTRACT

Transient outward, or "A-type," currents are rapidly inactivating voltage-gated potassium currents that operate at negative membrane potentials. A-type currents have not been reported in the gastric fundus, a tonic smooth muscle. We used whole cell voltage clamp to identify and characterize A-type currents in smooth muscle cells (SMCs) isolated from murine fundus. A-type currents were robust in these cells with peak amplitudes averaging 1.5 nA at 0 mV. Inactivation was rapid with a time constant of 71 ms at 0 mV; recovery from inactivation at -80 mV was similarly rapid with a time constant of 75 ms. A-type currents in fundus were blocked by 4-aminopyridine (4-AP), flecainide, and phrixotoxin-1 (PaTX1). Remaining currents after 4-AP and PaTX1 displayed half-activation potentials that were shifted to more positive potentials and showed incomplete inactivation. Currents after tetraethylammonium (TEA) displayed half inactivation at -48.1 ± 1.0 mV. Conventional microelectrode and contractile experiments on intact fundus muscles showed that 4-AP depolarized membrane potential and increased tone under conditions in which enteric neurotransmission was blocked. These data suggest that A-type K+ channels in fundus SMCs are likely active at physiological membrane potentials, and sustained activation of A-type channels contributes to the negative membrane potentials of this tonic smooth muscle. Quantitative analysis of Kv4 expression showed that Kcnd3 was dominantly expressed in fundus SMCs. These data were confirmed by immunohistochemistry, which revealed Kv4.3-like immunoreactivity within the tunica muscularis. These observations indicate that Kv4 channels likely form the A-type current in murine fundus SMCs.


Subject(s)
Gastric Fundus/metabolism , Gastrointestinal Motility , Muscle Contraction , Muscle, Smooth/metabolism , Potassium/metabolism , Shal Potassium Channels/metabolism , 4-Aminopyridine/pharmacology , Animals , Gastric Fundus/drug effects , Gastrointestinal Motility/drug effects , Kinetics , Male , Membrane Potentials , Mice, Inbred BALB C , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Potassium Channel Blockers/pharmacology , Shal Potassium Channels/antagonists & inhibitors , Shal Potassium Channels/genetics , Spider Venoms/metabolism
3.
Int J Surg Pathol ; 29(5): 503-505, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33827319

ABSTRACT

Pseudo-signet ring parietal cell vacuolation has been described as a mimic of invasive signet ring cell carcinoma. Moreover, signet ring cell carcinoma has been described in a fundic gland polyp. This case demonstrates parietal cell vacuolation in a fundic gland polyp in a patient on a long-term proton pump inhibitor.


Subject(s)
Carcinoma, Signet Ring Cell/diagnosis , Gastric Fundus/pathology , Polyps/diagnosis , Stomach Neoplasms/diagnosis , Aged , Biopsy , Diagnosis, Differential , Gastric Fundus/diagnostic imaging , Gastric Fundus/drug effects , Gastritis/drug therapy , Gastroscopy , Humans , Male , Polyps/chemically induced , Proton Pump Inhibitors/adverse effects , Stomach Neoplasms/chemically induced
4.
Int J Mol Sci ; 21(24)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348652

ABSTRACT

Some adipokines, such as adiponectin (ADPN), other than being implicated in the central regulation of feeding behavior, may influence gastric motor responses, which are a source of peripheral signals that also influence food intake. The present study aims to elucidate the signaling pathways through which ADPN exerts its actions in the mouse gastric fundus. To this purpose, we used a multidisciplinary approach. The mechanical results showed that ADPN caused a decay of the strip basal tension, which was abolished by the nitric oxide (NO) synthesis inhibitor, L-NG-nitro arginine (L-NNA). The electrophysiological experiments confirmed that all ADPN effects were abolished by L-NNA, except for the reduction of Ca2+ current, which was instead prevented by the inhibitor of AMP-activated protein kinase (AMPK), dorsomorphin. The activation of the AMPK signaling by ADPN was confirmed by immunofluorescence analysis, which also revealed the ADPN R1 receptor (AdipoR1) expression in glial cells of the myenteric plexus. In conclusion, our results indicate that ADPN exerts an inhibitory action on the gastric smooth muscle by acting on AdipoR1 and involving the AMPK signaling pathway at the peripheral level. These findings provide novel bases for considering AMPK as a possible pharmacologic target for the potential treatment of obesity and eating disorders.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/pharmacology , Gastric Mucosa/metabolism , Muscle, Smooth/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Female , Gastric Fundus/drug effects , Gastric Fundus/metabolism , Gastric Mucosa/drug effects , Mice , Mice, Inbred C57BL , Muscle, Smooth/drug effects , Obesity/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Adiponectin/metabolism
5.
Can J Physiol Pharmacol ; 98(10): 691-699, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32365302

ABSTRACT

Endokinin A/B (EKA/B), the common C-terminal decapeptide in endokinins A and B, is a preferred ligand of the NK1 receptor and regulates pain and itch. The study focused on the effects of EKA/B on rat gastric motility in vivo and in vitro. Gastric emptying was measured to evaluate gastric motility in vivo. Intragastric pressure and the contraction of gastric muscle strips were measured to evaluate gastric motility in vitro. Moreover, various neural blocking agents and neurokinin receptor antagonists were applied to explore the mechanisms. TAC4 and TACR1 mRNAs were expressed throughout rat stomach. EKA/B promoted gastric emptying by intraperitoneal injection in vivo. Correspondingly, EKA/B also increased intragastric pressure in vitro. Additionally, EKA/B contracted the gastric muscle strips from the fundus but not from the corpus or antrum. Further studies revealed that the contraction induced by EKA/B on muscle strips from the fundus could be significantly reduced by NK1 receptor antagonist SR140333 but not by NK2 receptor antagonist, NK3 receptor antagonist, or the neural blocking agents used. Our results suggested that EKA/B might stimulate gastric motility mainly through the direct activation of myogenic NK1 receptors located in the fundus.


Subject(s)
Gastric Emptying/drug effects , Gastric Fundus/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Peptide Fragments/pharmacology , Receptors, Neurokinin-1/agonists , Tachykinins/pharmacology , Animals , Gastric Fundus/metabolism , In Vitro Techniques , Male , Muscle, Smooth/metabolism , Pressure , Rats, Wistar , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Signal Transduction
6.
Pflugers Arch ; 471(7): 995-1005, 2019 07.
Article in English | MEDLINE | ID: mdl-31044280

ABSTRACT

Class-I-antiarrhythmics like ajmaline are known to alter smooth muscle function, which may cause alterations in gastrointestinal motility. The effects of ajmaline on isolated gastric and portal vein smooth muscle and the underlying mechanisms are unknown. We studied the effects of ajmaline on the contractile patterns of isolated preparations of gastric antrum and portal vein from Wistar rats. The organ bath technique was used to measure spontaneous or pharmacologically induced isometric contractions. Changes in force observed after application of ajmaline or under control conditions are reported as % of the amplitude of an initial K+-induced contraction. Electric field stimulation was used to study neurogenic relaxations of gastric fundus smooth muscle. Ajmaline increased the amplitude of spontaneous contractions of muscle strips (portal vein: control 31.1 ± 15.2%, with 100 µM ajmaline 76.6 ± 32.3%, n = 9, p < 0.01; gastric antrum: control 9.5 ± 1.6%, with 100 µM ajmaline 63.9 ± 9.96%, n = 14, p < 0.01). The frequency of spontaneous activity was reduced in portal vein, but not in gastric antrum strips. The effects of ajmaline were not blocked by tetrodotoxin, L-nitroarginine methyl ester, or atropine. Ajmaline abolished coordinated neurogenic relaxations triggered by electric field stimulation and partly reversed the inhibition of GA spontaneous activity caused by the gap junction blocker carbenoxolone. Ajmaline enhances the amplitude of spontaneous contractions in rat gastric and portal vein smooth muscle. This effect may be accompanied, but not caused by an inhibition of enteric neurotransmission. Enhanced syncytial coupling as indicated by its ability to antagonize the effects of carbenoxolone is likely to underlie the enhancement of contractility.


Subject(s)
Ajmaline/pharmacology , Gastric Fundus/drug effects , Muscle, Smooth/drug effects , Portal Vein/drug effects , Pyloric Antrum/drug effects , Animals , Atropine/pharmacology , Electric Stimulation/methods , Female , Gastrointestinal Motility/drug effects , Male , Muscle Contraction/drug effects , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Tetrodotoxin/pharmacology
7.
Eur J Pharmacol ; 851: 151-160, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30796903

ABSTRACT

It is generally considered that enteric neuropathy is one of the causative factors in diabetic gastroparesis. Our previous study demonstrated that there is a loss of NOS neurons in diabetic mice. However, the underlying mechanism remains unclear. The present study was designed to clarify the relationship between neuronal P2X7R and NOS neuron damage. The effect of P2X7R on diabetes-induced gastric NOS neurons damage and its mechanism were investigated by using quantitative RT-PCR,immunofluorescence, western blot, isometric force recording, intracellular calcium ([Ca2+]i) measurement and whole-cell patch clamp techniques. The immunohistochemistry and western blot results showed that nNOS expression was significantly down-regulated in diabetic mice, meanwhile, electric field stimulation-induced NOS sensitive relaxation was significantly suppressed. Myenteric neurons expressed P2X7R and pannexin1, and the mRNA and protein level of P2X7R and pannexin1 were up-regulated in diabetic mice. BzATP, a P2X7R activator, evoked [Ca2+]i increase in Hek293 cells with heterologous expression of P2X7R (Hek293-P2X7R cells) and the same dose of ATP-induced [Ca2+]i was more obvious in Hek293-P2X7R cells than in Hek293 cells. Application of BzATP activated an inward current of Hek293-P2X7R in a dose dependent manner. Hek293-P2X7R but not untransfected Hek293 cells could take up of YO-PRO-1. In addition, the uptake of YO-PRO-1 by Hek293-P2X7R was blocked by oxATP, a P2X7 antagonist and CBX, a pannexin1 inhibitor. The results suggest that the P2X7R of enteric neurons may be involved in diabetes-induced NOS neuron damage via combining with pannexin-1 to form transmembrane pores which induce macromolecular substances and calcium into the cells.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gastric Mucosa/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Calcium/metabolism , Gastric Fundus/drug effects , Gastric Fundus/metabolism , Gastric Mucosa/drug effects , Gene Expression Regulation, Enzymologic/drug effects , HEK293 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Mice , Mice, Inbred ICR , Neurons/drug effects , Neurons/pathology , Nitric Oxide Synthase Type I/metabolism
8.
Can J Physiol Pharmacol ; 97(5): 413-421, 2019 May.
Article in English | MEDLINE | ID: mdl-30730760

ABSTRACT

Statins are determined to have various pleiotropic effects apart from their lipid-lowering properties. Herein, we investigated the direct effects of atorvastatin on gastric smooth muscle tone. Atorvastatin effectively relaxed isolated rat gastric fundus strips precontracted with acetylcholine, potassium chloride, and serotonin. Incubation of the strips with nitric oxide synthase inhibitor, l-NOARG (10-4 M, 20 min), l-type voltage-operated Ca2+ channel (VOCC) blocker, nifedipine (10-6 M, 30 min), KATP channel blocker, glibenclamide (10-5 M, 30 min), or precursor of cholesterol, mevalonate (10-2 M, 45 min) did not change the relaxations to atorvastatin. However, pretreatment of fundus strips with atorvastatin (3×10-5-3×10-4 M, 30 min) inhibited the contractions to calcium chloride (10-4-10-1 M), acetylcholine (10-4 M), and caffeine (20 mM) in the calcium-free medium. Moreover, atorvastatin reduced the contractions induced by sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitor, cyclopiazonic acid (10-7-3×10-5 M). The current study demonstrated that atorvastatin produces an acute relaxant effect on gastric fundus strips, which appears to be mediated by several Ca2+-signalling mechanisms such as the blockade of l-type VOCC-independent Ca2+ entry, decrease in smooth muscle Ca2+ sensitivity, inhibition of IP3- and ryanodine-sensitive intracellular stores to mediate Ca2+ release, as well as the activation of SERCA. This acute relaxing effect seems unlikely to be related with nitric oxide, KATP channels, and the mevalonate pathway.


Subject(s)
Atorvastatin/pharmacology , Calcium Signaling/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Animals , Gastric Fundus/drug effects , Gastric Fundus/physiology , Male , Muscle, Smooth/cytology , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
9.
Neurogastroenterol Motil ; 31(4): e13540, 2019 04.
Article in English | MEDLINE | ID: mdl-30663175

ABSTRACT

BACKGROUND: Acotiamide, a prokinetic agent was shown to be efficacious in the treatment of functional dyspepsia (FD). The exact mechanism of action is incompletely elucidated. METHODS: This randomized, placebo-controlled, cross-over study aimed to examine the effect of acotiamide on gastric motility, measured as intragastric pressure, gastric emptying (GE) rate and gastrointestinal (GI) symptom perception in healthy volunteers (HVs). Participants were treated with acotiamide (100 mg tid) and placebo for 3 weeks, separated by a 1-week washout period. A daily symptom diary was collected during both treatments. At the end of each treatment period, GE rate and gastric motility were assessed with a 13 C-octanoic acid breath test and high-resolution manometry during nutrient infusion, respectively. GI symptom levels were scored during high-resolution manometry. Data were analyzed using mixed models. The study was registered as NCT03402984. KEY RESULTS: Twenty HVs (10 female, 25 ± 4.1 years, 22.58 ± 2.73 kg/m2 ) participated in the study. There was no difference in GE half time between both treatments (P = 0.92). Acotiamide had no effect on fundic pressures before and after nutrient infusion (P = 0.91). However, postprandial antral pressures remained significantly lower compared to placebo (P = 0.015). There was no significant difference in hunger, satiation and GI symptoms scores assessed during IGP measurement and by the daily diary (P > 0.12 for all). CONCLUSION: Acotiamide is associated with lower antral pressures after nutrient intake, whereas it has no effect on fundic pressures, GE rate and symptom perceptions in HVs. Studies in FD need to elucidate whether lower antral pressures induced by acotiamide underlie postprandial symptom improvement in FD.


Subject(s)
Benzamides/pharmacology , Gastric Emptying/drug effects , Gastric Fundus/drug effects , Gastrointestinal Agents/pharmacology , Gastrointestinal Motility/drug effects , Pyloric Antrum/drug effects , Thiazoles/pharmacology , Adult , Cross-Over Studies , Female , Healthy Volunteers , Humans , Male , Manometry , Single-Blind Method , Young Adult
10.
Clin Exp Pharmacol Physiol ; 46(1): 40-47, 2019 01.
Article in English | MEDLINE | ID: mdl-30229988

ABSTRACT

ß-Phenylethylamine (ß-PEA) is a trace amine with chemical proximity to biogenic amines and amphetamines. It is an endogenous agonist of trace amine-associated receptors (TAARs) that acts as a neuromodulator of classic neurotransmitters in the central nervous system. At high concentrations, ß-PEA contracts smooth muscle, and a role for TAARs in these responses has been postulated. The high dietary intake of trace amines has been associated with such symptoms as hypertension and migraine, especially after the intake of foods containing such compounds. In gastrointestinal tissues, TAAR expression was reported, although the effect of ß-PEA on gastric contractile behaviour is unknown. Here, isolated strips that were obtained from the rat gastric fundus were stimulated with high micromolar concentrations of ß-PEA. Under resting tonus, ß-PEA induced contractions. In contrast, when the strips were previously contracted with KCl, a relaxant response to ß-PEA was observed. The contractile effect of ß-PEA was inhibited by 5-hydroxytryptamine (5-HT) receptor antagonists (i.e., cyproheptadine and ketanserin) but not by the TAAR1 antagonist EPPTB. In gastric fundus strips that were previously contracted with 80 mmol/L KCl, the relaxant effect of ß-PEA intensified in the presence of 5-HT receptor antagonists, which was inhibited by EPPTB and the adenylyl cyclase inhibitor MDL-12,330A. The guanylyl cyclase inhibitor ODQ did not alter the relaxant effects of ß-PEA. In conclusion, ß-PEA exerted dual contractile and relaxant effects on rat gastric fundus. The contractile effect appeared to involve the recruitment of 5-HT receptors, and the relaxant effect of ß-PEA on KCl-elicited contractions likely involved TAAR1 .


Subject(s)
Gastric Fundus/drug effects , Gastric Fundus/physiology , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Phenethylamines/pharmacology , Animals , Gastric Fundus/metabolism , Muscle Contraction/drug effects , Potassium Chloride/pharmacology , Rats , Receptors, G-Protein-Coupled/metabolism , Receptors, Serotonin/metabolism
11.
Acta Biol Hung ; 69(2): 135-143, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29888673

ABSTRACT

Aqueous extract of the spines of the brown bullhead catfish (Ameiurus nebulosus Lesueur, 1819) caused contraction of the guinea-pig small intestine in vitro, a widely-used preparation in pharmacology. The action is dependent on extracellular Ca2+, and probably takes place on the smooth muscle cells. Mouse gastrointestinal preparations were also contracted by the extract. Stings by the spines of this fish species causes a painful sensation in man. We tested the effect of an extract of spines in isolated organ experiments on innervated smooth muscle preparations. In the guinea-pig ileum, the response to the extract was abolished by the Ca2+-channel blocker nifedipine, but only slightly reduced by atropine (a muscarine receptor antagonist) or tetrodotoxin (TTX; a blocker axonal conduction) or antagonists for P2X purinoceptors. Blocking of serotonin or histamine H1 receptors, tachykinin NK1 receptors, functional impairment of capsaicin-sensitive sensory nerve endings or inhibition of cyclo-oxygenases failed to influence the contractile effect of the extract. No inhibitory action of the extract was detected on the ileum subject to electrical motor nerve stimulation.


Subject(s)
Fish Venoms/pharmacology , Gastric Fundus/drug effects , Ileum/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Animals , Atropine/pharmacology , Calcium Channel Blockers/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Female , Guinea Pigs , Histamine H1 Antagonists/pharmacology , Ictaluridae , In Vitro Techniques , Intestine, Small/drug effects , Male , Mice , Muscarinic Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/pharmacology , Nifedipine/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Serotonin Antagonists/pharmacology , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
12.
Phytomedicine ; 43: 55-59, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29747754

ABSTRACT

BACKGROUND: Resveratrol is a polyphenolic compound that can be isolated from plants and also is a constituent of red wine. Resveratrol induces relaxation of vascular smooth muscle and may prevent cardiovascular diseases. PURPOSE: Impaired gastric accommodation plays an important role in functional dyspepsia and fundic relaxation and is a therapeutic target of functional dyspepsia. Although drugs for fundic relaxation have been developed, these types of drugs are still rare. The purpose of this study was to investigate the relaxant effects of resveratrol in the guinea pig fundus. STUDY DESIGN: We studied the relaxant effects of resveratrol in the guinea pig fundus. In addition, we investigated the mechanism of resveratrol-induced relaxation on the guinea pig fundus by using tetraethylammonium (a non-selective potassium channel blocker), apamine (a selective inhibitor of the small conductance calcium-activated potassium channel), iberiotoxin (an inhibitor of large conductance calcium-activated potassium channels), glibenclamide (an ATP-sensitive potassium channel blocker), KT 5720 (a cAMP-dependent protein kinase A inhibitor), KT 5823 (a cGMP-dependent protein kinase G inhibitor), NG-nitro-L-arginine (a competitive inhibitor of nitric oxide synthase), tetrodotoxin (a selective neuronal Na+ channel blocker), ω-conotoxin GVIA (a selective neuronal Ca2+ channel blocker) and G-15 (a G-protein coupled estrogen receptor antagonist). RESULTS: The results of this study showed that resveratrol has potent and dose-dependent relaxant effects on the guinea pig fundic muscle. In addition, the results showed that resveratrol-induced relaxation of the guinea pig fundus occurs through nitric oxide and ATP-sensitive potassium channels. CONCLUSION: This study provides the first evidence concerning the relaxant effects of resveratrol in the guinea pig fundic muscle strips. Furthermore, resveratrol may be a potential drug to relieve gastrointestinal dyspepsia.


Subject(s)
Gastric Fundus/drug effects , Muscle Relaxation/drug effects , Stilbenes/pharmacology , Animals , Dose-Response Relationship, Drug , Gastric Fundus/physiology , Guinea Pigs , KATP Channels/metabolism , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Organ Culture Techniques , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Resveratrol , Stilbenes/administration & dosage , Tetraethylammonium/pharmacology
13.
Neurogastroenterol Motil ; 30(10): e13372, 2018 10.
Article in English | MEDLINE | ID: mdl-29740911

ABSTRACT

BACKGROUND: Sphingosine and its metabolite sphingosine phosphate (S1P) regulate a multitude of biological functions, including the contractile state of smooth. Gastrointestinal side effects have been reported in patients treated with FTY720, a sphingosine analog that is approved for the treatment of multiple sclerosis. The aim of this study was to characterize the effects of FTY720 on rat gastric fundus smooth muscle under basal conditions and during activation induced by high-K+ solution. METHODS: Isometric contractions of isolated circular strips of gastric fundus smooth muscle were recorded using the organ bath method. The effects of FTY720 or vehicle were recorded under control conditions and in the presence of indomethacin, L-NAME, HA-1100, nifedipine, JTE-013, and suramin. Tone and contractions recorded in the presence of FTY720 or vehicle are reported as % of the amplitude of an initial high-K+ contraction obtained under control conditions. KEY RESULTS: From a concentration of 10 µmol L-1 onwards, FTY720 increased the tone, reaching 8.9% ± 7.5% at 100 µmol L-1 (P < .05). With indomethacin in the solution, the effects of FTY720 were enhanced (32.1% ± 7.7%; P < .001). The FTY720-induced increase in tone was abolished in the absence of extracellular Ca2+ and reduced by nifedipine, HA-1100, JTE-013, and suramin. Furthermore, FTY720 increased high-K+ contractions in the presence of indomethacin. CONCLUSIONS & INFERENCES: FTY720 increases tone and contractile responses to depolarization in gastric fundus smooth muscle by triggering calcium entry and calcium sensitization in a S1P receptor-dependent manner. Taken together, the experimental results presented in this work suggest that FTY720 may increase gastric tone and contractility in patients.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Gastric Fundus/drug effects , Immunosuppressive Agents/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Animals , Female , Male , Rats , Rats, Wistar
14.
Exp Physiol ; 102(12): 1607-1618, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28929535

ABSTRACT

NEW FINDINGS: What is the central question of this study? Acute acidosis that results from short-term exercise is involved in delayed gastric emptying in rats and the lower responsiveness of gastric fundus strips to carbachol. Does extracellular acidosis decrease responsiveness to carbachol in tissues of sedentary rats? How? What is the main finding and its importance? Extracellular acidosis inhibits cholinergic signalling in the rat gastric fundus by selectively influencing the Gq/11 protein signalling pathway. Acute acidosis that results from short-term exercise delays gastric emptying in rats and decreases the responsiveness to carbachol in gastric fundus strips. The regulation of cytosolic Ca2+ concentrations appears to be a mechanism of action of acidosis. The present study investigated the way in which acidosis interferes with gastric smooth muscle contractions. Rat gastric fundus isolated strips at pH 6.0 presented a lower magnitude of carbachol-induced contractions compared with preparations at pH 7.4. This lower magnitude was absent in carbachol-stimulated duodenum and KCl-stimulated gastric fundus strips. In Ca2+ -free conditions, repeated contractions that were induced by carbachol progressively decreased, with no influence of extracellular pH. In fundus strips, CaCl2 -induced contractions were lower at pH 6.0 than at pH 7.4 but only when stimulated in the combined presence of carbachol and verapamil. In contrast, verapamil-sensitive contractions that were induced by CaCl2 in the presence of KCl did not change with pH acidification. In Ca2+ store-depleted preparations that were treated with thapsigargin, the contractions that were induced by extracellular Ca2+ restoration were smaller at pH 6.0 than at pH 7.4, but relaxation that was induced by SKF-96365 (an inhibitor of store-operated Ca2+ entry) was unaltered by extracellular acidification. At pH 6.0, the phospholipase C inhibitor U-73122 relaxed carbachol-induced contractions less than at pH 7.4, and this phenomenon was absent in tissue that was treated with the RhoA kinase blocker Y-27632. Thus, extracellular acidosis inhibited pharmacomechanical coupling in gastric fundus by selectively inhibiting the Gq/11 protein signalling pathway, whereas electromechanical coupling remained functionally preserved.


Subject(s)
Acidosis/metabolism , Calcium Signaling/drug effects , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gastric Emptying/drug effects , Gastric Fundus/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Animals , Dose-Response Relationship, Drug , Gastric Fundus/metabolism , Hydrogen-Ion Concentration , In Vitro Techniques , Male , Muscle, Smooth/metabolism , Rats, Wistar
15.
Indian J Pharmacol ; 49(1): 42-48, 2017.
Article in English | MEDLINE | ID: mdl-28458421

ABSTRACT

OBJECTIVES: The aim of this study was to evaluate anticholinergic, antihistaminic, and antiserotonergic activity of the n-hexane extract of the seeds of Zanthoxylum alatum (ZAHE) on isolated ileum of rat and guinea pig and fundus of rat. MATERIALS AND METHODS: ZAHE was prepared using soxhlet extraction and cumulative concentration response curves were constructed using various doses on the tissues for acetylcholine (ACh), 5-hydroxytryptamine (5-HT), and histamine with or without n-hexane extract. Atropine, ketanserin, and pheniramine maleate were used as antagonists for ACh, serotonin, and histamine, respectively. RESULTS: ZAHE-induced concentration-dependent inhibition of isolated ileum and fundus in rat and ileum of guinea pig. The half maximal effective concentration (EC50) of ACh in the presence of atropine (10-6 M; P < 0.05) and ZAHE (1000 µg/ml; P < 0.01) was significantly higher than EC50of ACh alone. The EC50of 5-HT in the presence of ketanserin (10-5 M; P < 0.01) and ZAHE (1000 µg/ml; P < 0.05) was higher than EC50of 5-HT alone. Similarly, the EC50of histamine in the presence of pheniramine maleate (10-6 M; P < 0.01) and ZAHE (300 µg/ml; P < 0.01 and 1000 µg/ml; P < 0.05) was also significantly higher than EC50of histamine alone. CONCLUSION: From the study, it was observed that ZAHE shows significant anticholinergic, antiserotonergic, and antihistaminic activity. The study provides sufficient evidence that the seeds can be used in gastric disorders, cough, chest infection, etc., as per folklore claims.


Subject(s)
Cholinergic Antagonists/pharmacology , Histamine Antagonists/pharmacology , Plant Extracts/pharmacology , Serotonin Antagonists/pharmacology , Zanthoxylum/chemistry , Acetylcholine/metabolism , Animals , Cholinergic Antagonists/administration & dosage , Cholinergic Antagonists/isolation & purification , Dose-Response Relationship, Drug , Gastric Fundus/drug effects , Gastric Fundus/metabolism , Guinea Pigs , Hexanes/chemistry , Histamine/metabolism , Histamine Antagonists/administration & dosage , Histamine Antagonists/isolation & purification , Ileum/drug effects , Ileum/metabolism , Male , Plant Extracts/administration & dosage , Rats , Rats, Wistar , Seeds , Serotonin/metabolism , Serotonin Antagonists/administration & dosage , Serotonin Antagonists/isolation & purification
16.
Exp Physiol ; 102(7): 779-790, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28383821

ABSTRACT

NEW FINDINGS: What is the central question of this study? The present study investigated the relationship between H2 S and NO in regulation of gastric fundus tension. What is the main finding and its importance? Endogenous or exogenous H2 S and NO have opposite effects on fundus tension, and H2 S-induced gastric fundus tension enhancements are mediated by inhibition of NO generation through the phosphoinositide 3-kinase/Akt pathway. These results are very important in exploring the mechanism of physiological accommodation and accommodation disorder. Hydrogen sulphide (H2 S) is considered a new gasotransmitter, along with NO and CO. It was recently confirmed that H2 S and NO play important roles in the regulation of gastrointestinal smooth muscle tension. The present study was designed to elucidate the interactions between H2 S and NO with respect to the regulation of gastric fundus smooth muscle tension using Western blotting, physiological and electrochemical techniques. Real-time H2 S and NO generation was detected in gastric smooth muscle tissue. NaHS, an H2 S donor, enhanced fundus smooth muscle tension, whereas SNP, an NO donor, decreased fundus smooth muscle tension in a dose-dependent manner. NaHS-induced increases in fundus smooth muscle tension were suppressed by l-NAME, an NO synthase inhibitor. Aminooxyacetic acid (AOAA), a cystathionine ß-synthase inhibitor, exerted inhibitory effects on fundus smooth muscle tension; these effects were also suppressed by l-NAME. Real-time NO generation was significantly potentiated by AOAA. Endothelial nitric oxide synthase (eNOS) phosphorylation at serine 1177 and Akt phosphorylation at serine 308 and threonine 473 were significantly inhibited by NaHS. LY294002, a phosphoinositide 3-kinase inhibitor, blocked these NaHS-mediated effects. However, eNOS phosphorylation at serine 1177 and Akt phosphorylation at serine 308 and threonine 473 were significantly potentiated by AOAA. Cystathionine ß-synthase siRNA interference significantly increased eNOS phosphorylation at serine 1177 and Akt phosphorylation at serine 308 and threonine 473. Cystathionine ß-synthase siRNA interference also increased total eNOS protein expression levels but did not significantly change total Akt kinase protein expression levels. These results suggest that H2 S-induced enhancement of gastric fundus tension is mediated by inhibition of NO generation through the phosphoinositide 3-kinase/Akt pathway.


Subject(s)
Gastric Fundus/drug effects , Hydrogen Sulfide/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Signal Transduction , Animals , Male , Mice , Muscle Tonus/drug effects , Muscle, Smooth/physiology , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
17.
Article in English | MEDLINE | ID: mdl-28332745

ABSTRACT

BACKGROUND: In the gastrointestinal tract of several species, facilitating 5-HT4 receptors were proposed on myenteric cholinergic neurons innervating smooth muscle by in vitro study of the effect of the selective 5-HT4 receptor agonist prucalopride on submaximal cholinergic contractions. This was not yet established in the murine gastrointestinal tract. METHODS: In circular smooth muscle strips from murine fundus, jejunum and colon, contractions were induced by electrical field stimulation in the presence of guanethidine, L-NAME and for colon also MRS 2500. Submaximal contractions were induced to study the influence of prucalopride. KEY RESULTS: Electrical field stimulation at reduced voltage induced reproducible submaximal neurogenic and cholinergic contractions as the contractions were abolished by tetrodotoxin and atropine. Hexamethonium had no systematic inhibitory effect but mecamylamine reduced the responses, suggesting that part of the cholinergic response is due to activation of preganglionic neurons. Prucalopride concentration-dependently increased the submaximal cholinergic contractions in the three tissue types, reaching maximum from 0.03 µmol/L onwards. The facilitation in the different series with 0.03 µmol/L prucalopride ranged from 41% to 104%, 30% to 76% and 24% to 74% in fundus, jejunum, and colon, respectively. The effect of 0.03 µmol/L prucalopride was concentration-dependently inhibited by GR 113808. CONCLUSIONS & INFERENCES: In the murine gastrointestinal tract, activation of 5-HT4 receptors with prucalopride enhances cholinergic contractions, illustrating facilitation of myenteric cholinergic neurotransmission. The degree of enhancement with prucalopride is of similar magnitude as previously reported in other species, but the effective concentrations are lower than those needed in the gastrointestinal tract of other species.


Subject(s)
Acetylcholine/physiology , Gastrointestinal Tract/physiology , Receptors, Serotonin, 5-HT4/physiology , Synaptic Transmission , Animals , Benzofurans/administration & dosage , Colon/drug effects , Colon/physiology , Deoxyadenine Nucleotides/administration & dosage , Electric Stimulation , Gastric Fundus/drug effects , Gastric Fundus/physiology , Gastrointestinal Tract/drug effects , Guanethidine/administration & dosage , Hexamethonium/administration & dosage , Jejunum/drug effects , Jejunum/physiology , Male , Mecamylamine/administration & dosage , Mice, Inbred C57BL , Muscle Contraction , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , NG-Nitroarginine Methyl Ester/administration & dosage , Serotonin 5-HT4 Receptor Agonists/administration & dosage
18.
Dig Dis Sci ; 62(1): 93-104, 2017 01.
Article in English | MEDLINE | ID: mdl-27864656

ABSTRACT

AIM: The objective was to evaluate the effects of nitric oxide (NO) and hydrogen sulfide (H2S) donors and possible interactions between these two systems in modulating gastric function. METHODS: Mice received saline, sodium nitroprusside (SNP), or sodium hydrosulfite (NaHS), and after 1 h, the animals were killed for immunofluorescence analysis of CSE or eNOS expressions, respectively. Other groups received saline, SNP, NaHS, Lawesson's reagent (H2S donor), PAG + SNP, L-NAME, L-NAME + NaHS, or L-NAME + Lawesson's reagent. Then, the gastric secretions (mucous and acid), gastric blood flow, gastric defense against ethanol, and gastric motility (gastric emptying and gastric contractility) were evaluated. RESULTS: SNP and NaHS increased the expression of CSE or eNOS, respectively. SNP or Lawesson's reagent did not alter gastric acid secretion but increased mucus production, and these effects reverted with PAG and L-NAME treatment, respectively. SNP or NaHS increased gastric blood flow and protected the gastric mucosa against ethanol injury, and these effects reverted with PAG and L-NAME treatments, respectively. SNP delayed gastric emptying when compared with saline, and PAG partially reversed this effect. NaHS accelerate gastric emptying, and L-NAME partially reversed this effect. SNP and NaHS alone induced gastric fundus and pylorus relaxation. However, pretreatment with PAG or L-NAME reversed these relaxant effects only in the pylorus but not in the gastric fundus. CONCLUSION: NO and H2S interact in gastric physiological functions, and this "cross-talk" is important in the control of mucus secretion, gastric blood flow, gastric mucosal defense, and gastric motility, but not in the control of basal gastric acid secretion.


Subject(s)
Cystathionine gamma-Lyase/drug effects , Gastric Emptying/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/drug effects , Nitroprusside/pharmacology , Stomach/drug effects , Sulfides/pharmacology , Alkynes/pharmacology , Animals , Central Nervous System Depressants/pharmacology , Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/metabolism , Drug Interactions , Enzyme Inhibitors/pharmacology , Ethanol/pharmacology , Fluorescent Antibody Technique , Gastric Acid/metabolism , Gastric Fundus/drug effects , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Glutathione/drug effects , Glutathione/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Laser-Doppler Flowmetry , Male , Malondialdehyde/metabolism , Mice , Mucus/drug effects , Mucus/metabolism , Muscle Contraction/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Pylorus/drug effects , Rats , Rats, Wistar , Regional Blood Flow , Stomach/blood supply
19.
Folia Morphol (Warsz) ; 76(2): 252-261, 2017.
Article in English | MEDLINE | ID: mdl-27813628

ABSTRACT

BACKGROUND: Peptic ulcer diseases are common and are induced by many factors, including stress, smoking, and ingestion of non-steroidal anti-inflammatory drugs. Quercetin is considered to be an anti-oxidant with healing effects on many experimental toxic injuries. The present study aimed to explore the possible effect of quercetin on acute gastric ulcer induced by indomethacin in rats. MATERIALS AND METHODS: Three groups received indomethacin (30 mg/kg body weight) orally by orogastric gavage on two consecutive days. The rats received famotidine (50 mg/kg body weight), quercetin (50 mg/kg body weight), or vehicle alone for 15 consecutive days by oral gavage. The control group received no indomethacin but received vehicle for 15 days by oral gavage. The ulcer index, volume, and pH of gastric juice were measured, and the stomachs were examined by routine light microscopy. RESULTS: Compared with the control group, the indomethacin-treated rats showed a marked damage of the gastric mucosal surface and a high ulcer index. In the famotidine- and quercetin-treated groups, significantly increased antioxidant enzyme activities were observed. The congestion, erosions, and necrosis were reduced with mild inflammatory cell infiltration while no major damage of endothelial cells was observed in the treated rats. CONCLUSIONS: The findings of the study show that quercetin had antioxidant effect and can protect gastric mucosa against indomethacin-induced gastric ulceration than famotidine.


Subject(s)
Quercetin/therapeutic use , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Acute Disease , Animals , Gastric Fundus/drug effects , Gastric Fundus/pathology , Gastric Mucosa/drug effects , Glutathione/metabolism , Hydrogen-Ion Concentration , Indomethacin , Male , Quercetin/pharmacology , Rats
20.
J Vasc Interv Radiol ; 27(12): 1923-1928, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27717647

ABSTRACT

PURPOSE: To assess whether the number of fundal arteries embolized and use of gastroprotective agents have an impact on ghrelin suppression and gastric ulceration rates. MATERIALS AND METHODS: Twenty-two healthy, growing swine (mean, 38.4 kg; range, 30.3-47.0 kg) were evaluated. Six control swine underwent a sham procedure. Gastric embolization was performed by the infusion of 40-µm microspheres selectively into some or all gastric arteries supplying the gastric fundus. In group 1, 6 swine underwent embolization of all 4 arteries to the gastric fundus. In group 2, 5 swine underwent embolization of 2 gastric fundal arteries. In group 3, 5 swine underwent embolization of 1 gastric fundal artery. Animals in groups 2 and 3 were treated with gastroprotective agents (sucralfate and omeprazole). Weight and fasting plasma ghrelin levels were analyzed at baseline and at week 4. Upon animal euthanasia, gross analysis was performed for identification of ulcers. RESULTS: Only group 1 animals exhibited changes in serum ghrelin levels that rendered them significantly lower than those in control animals (P = .049). Group 3 animals exhibited marked elevations in serum ghrelin levels compared with control animals (P = .001). Gross pathologic evaluation revealed 0 ulcers in the control animals, 3 ulcers (50%) in group 1, 2 ulcers (40%) in group 2, and 2 ulcers (40%) in group 3. CONCLUSIONS: Administration of gastroprotective agents and embolization of fewer arteries to the gastric fundus did not prevent gastric ulceration in treated animals. Only animals that underwent embolization of all gastric arteries exhibited significant decreases in serum ghrelin levels.


Subject(s)
Embolization, Therapeutic/methods , Gastric Fundus/blood supply , Gastric Fundus/drug effects , Gastric Mucosa/drug effects , Omeprazole/pharmacology , Proton Pump Inhibitors/pharmacology , Stomach Ulcer/prevention & control , Sucralfate/pharmacology , Angiography , Animals , Anti-Ulcer Agents , Arteries/diagnostic imaging , Biomarkers/blood , Cytoprotection , Down-Regulation , Embolization, Therapeutic/adverse effects , Gastric Fundus/metabolism , Gastric Fundus/pathology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Ghrelin/blood , Models, Animal , Pilot Projects , Stomach Ulcer/blood , Stomach Ulcer/etiology , Stomach Ulcer/pathology , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...