Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 378
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.
Biochem Pharmacol ; 220: 115969, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38086489

ABSTRACT

Flavonoids, ubiquitously distributed in the plant world, are regularly ingested with diets rich in fruit, vegetables, wine, and tea. During digestion, they are partially absorbed in the stomach. The present work aimed to assess the in vitro effects of quercetin and ten structurally related flavonoids on the rat gastric fundus smooth muscle, focussing on ATP-dependent K+ (Kir6.1) channels, which play a central role in the regulation of resting membrane potential, membrane excitability and, consequently, of gastric motility. Whole-cell currents through Kir6.1 channels (IKir6.1) were recorded with the patch-clamp technique and the mechanical activity of gastric fundus smooth muscle strips was studied under isometric conditions. Galangin ≈ tamarixetin > quercetin > kaempferol > isorhamnetin ≈ luteolin ≈ fisetin > (±)-taxifolin inhibited pinacidil-evoked, glibenclamide-sensitive IKir6.1 in a concentration-dependent manner. Morin, rutin, and myricetin were ineffective. The steric hindrance of the molecule and the number and position of hydroxyl groups on the B ring played an important role in the activity of the molecule. Molecular docking simulations revealed a possible binding site for flavonoids in the C-terminal domain of the Kir6.1 channel subunit SUR2B, in a flexible loop formed by residues 251 to 254 of chains C and D. Galangin and tamarixetin, but not rutin relaxed both high K+- and carbachol-induced contraction of fundus strips in a concentration-dependent manner. Furthermore, both flavonoids shifted to the right the concentration-relaxation curves to either pinacidil or L-cysteine constructed in strips pre-contracted by high K+, rutin being ineffective. In conclusion, IKir6.1 inhibition exerted by dietary flavonoids might counterbalance their myorelaxant activity, affect gastric accommodation or, at least, some stages of digestion.


Subject(s)
Gastric Fundus , Vasodilator Agents , Rats , Animals , Pinacidil/pharmacology , Vasodilator Agents/pharmacology , Gastric Fundus/metabolism , Quercetin/pharmacology , Molecular Docking Simulation , Potassium Channels/metabolism , Muscle, Smooth/metabolism , Electrophysiology , Rutin , Diet , Sulfonylurea Receptors/metabolism
3.
Hormones (Athens) ; 22(2): 151-163, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36705877

ABSTRACT

PURPOSE: Ghrelin, one of the most studied gut hormones, is mainly produced by the gastric fundus. Abundant evidence exists from preclinical and clinical studies underlining its contribution to glucose regulation. In the following narrative review, the role of the gastric fundus in glucose regulation is summarized and we investigate whether its resection enhances glycemic control. METHODS: An electronic search was conducted in the PubMed® database and in Google Scholar® using a combination of medical subject headings (MeSH). We examined types of metabolic surgery, including, in particular, gastric fundus resection, either as part of laparoscopic sleeve gastrectomy (LSG) or modified laparoscopic gastric bypass with fundus resection (LRYGBP + FR), and the contribution of ghrelin reduction to glucose regulation. RESULTS: Fourteen human studies were judged to be eligible and included in this narrative review. Reduction of ghrelin levels after fundus resection might be related to early glycemic improvement before significant weight loss is achieved. Long-term data regarding the role of ghrelin reduction in glucose homeostasis are sparse. CONCLUSION: The exact role of ghrelin in achieving glycemic control is still ambiguous. Data from human studies reveal a potential contribution of ghrelin reduction to early glycemic improvement, although further well-designed studies are needed.


Subject(s)
Gastric Bypass , Laparoscopy , Obesity, Morbid , Humans , Ghrelin/metabolism , Obesity, Morbid/surgery , Gastric Fundus/surgery , Gastric Fundus/metabolism , Glycemic Control , Glucose/metabolism , Gastrectomy
4.
Pharmacol Res Perspect ; 10(3): e00952, 2022 06.
Article in English | MEDLINE | ID: mdl-35466586

ABSTRACT

The inhibitory effects of docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and linoleic acid (LA) on the contractions induced by five prostanoids and U46619 (a TP receptor agonist) were examined in guinea pig gastric fundus smooth muscle (GFSM). Tension changes were isometrically measured, and the mRNA expression of prostanoid receptors was measured by RT-qPCR. DHA and EPA significantly inhibited contractions induced by the prostanoids and U46619, whereas LA inhibited those induced by prostaglandin D2 and U46619. The mRNA expression levels of the prostanoid receptors were TP ≈ EP3  >> FP > EP1 . The inhibition by DHA, EPA, and LA was positively correlated with that by SQ 29,548 (a TP receptor antagonist) but not with that by L-798,106 (an EP3 receptor antagonist). DHA and EPA suppressed high KCl-induced contractions by 35% and 25%, respectively, and the contractions induced by the prostanoids and U46619 were suppressed by verapamil, a voltage-dependent Ca2+ channel (VDCC) inhibitor, by 40%-85%. Although LA did not suppress high KCl-induced contractions, it suppressed U46619-induced contractions in the presence of verapamil. However, LA did not show significant inhibitory effects on U46619-induced Ca2+ increases in TP receptor-expressing cells. In contrast, LA inhibited U46619-induced contractions in the presence of verapamil, which was also suppressed by SKF-96365 (a store-operated Ca2+ channel [SOCC] inhibitor). These findings suggest that the TP receptor and VDCC are targets of DHA and EPA to inhibit prostanoid-induced contractions of guinea pig GFSM, and SOCCs play a significant role in LA-induced inhibition of U46619-induced contractions.


Subject(s)
Docosahexaenoic Acids , Eicosapentaenoic Acid , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Calcium Channels, L-Type/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Gastric Fundus/metabolism , Guinea Pigs , Muscle, Smooth , Prostaglandins/metabolism , Prostaglandins/pharmacology , RNA, Messenger/metabolism , Receptors, Thromboxane/metabolism , Verapamil/metabolism , Verapamil/pharmacology
5.
Nitric Oxide ; 119: 19-28, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34902580

ABSTRACT

In this study, we investigated the possible role of the l-cysteine/hydrogen sulfide pathway in ß3-adrenoceptors-mediated relaxation in isolated mouse gastric fundus tissue. l-cysteine (endogenous H2S; 10-6-10-2 M), sodium hydrogen sulfide (NaHS; exogenous H2S; 10-6-10-3 M), selective ß3-adrenoceptors agonist BRL 37344 (10-9-10-4 M) and non-selective ß-adrenoceptor agonist isoprenaline (10-9-10-4 M) produced concentration-dependent relaxation in mouse gastric fundus. The non-selective ß-adrenoceptors antagonist propranolol (10-6 M) inhibited the relaxant response to isoprenaline but not to BRL 37344. On the other hand, the selective ß3-adrenoceptors antagonist SR 59230A (10-5 M) inhibited the relaxant responses to BRL 37344. In addition, cystathionine-gamma-lyase (CSE) inhibitor D,L-propargylglycine (PAG, 10-2 M), cystathionine-beta-synthase inhibitor (CBS) aminooxyacetic acid (AOAA, 10-2 M), and the combination of these inhibitors significantly reduced the relaxant responses induced by l-cysteine and BRL 37344. Pre-incubation of gastric fundal strips with propranolol (10-6 M) and SR 59230A (10-5 M) did not affect relaxations to l-cysteine and NaHS. Also, the existence of CSE, CBS, 3-mercaptopurivate sulfur transferase (3-MST) enzymes and ß3-adrenoceptors were detected in gastric fundal tissue. Furthermore, basal H2S release was detected in the measurements. H2S level increased in the presence of l-cysteine, NaHS, and BRL 37344. The increase in H2S level by l-cysteine and BRL 37344 decreased significantly with PAG and AOAA enzyme inhibitors. These results suggest that endogenous H2S is synthesized from l-cysteine at least by CBS and CSE enzymes. Also, ß3-adrenoceptors are found in the mouse stomach fundus and mediate BRL 37344-induced relaxations, and l-cysteine/hydrogen sulfide pathway plays a partial role in ß3-adrenoceptors-mediated relaxation in mouse gastric fundus tissue.


Subject(s)
Cysteine/metabolism , Gastric Fundus/metabolism , Hydrogen Sulfide/metabolism , Muscle Relaxation/physiology , Receptors, Adrenergic, beta-3/metabolism , Animals , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Ethanolamines/pharmacology , Gastric Fundus/enzymology , Isoproterenol/pharmacology , Male , Mice , Propanolamines/pharmacology , Propranolol/pharmacology , Sulfurtransferases/metabolism
6.
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
7.
Neurogastroenterol Motil ; 33(10): e14162, 2021 10.
Article in English | MEDLINE | ID: mdl-33939222

ABSTRACT

BACKGROUND: The motor protein, Myosin 5a (Myo5a) is known to play a role in inhibitory neurotransmission in gastric fundus. However, there is no information regarding the relative expression of total Myo5a, or of its alternative exon splice variants, across the stomach. This study investigated the differential distribution of Myo5a variants expressed within distinct anatomical regions of murine stomach. METHODS: The distribution of Myo5a protein and mRNA in the stomach was assessed by immunofluorescence microscopy and fluorescent in situ hybridization. Quantitative PCR, restriction enzyme analysis, and electrophoresis were used to identify Myo5a splice variants and quantify their expression levels in the fundus, body, antrum, and pylorus. KEY RESULTS: Myo5a protein colocalized with ßIII-Tubulin in the myenteric plexus, and with synaptophysin in nerve fibers. Total Myo5a mRNA expression was lower in pylorus than in antrum, body, or fundus (p < 0.001), which expressed equivalent amounts of Myo5a. However, Myo5a splice variants were differentially expressed across the stomach. While the ABCE splice variant predominated in the antrum and body regions, the ACEF/ACDEF variants were enriched in fundus and pylorus. CONCLUSIONS AND INFERENCES: Myo5a splice variants varied in their relative expression across anatomically distinguishable stomach regions and might mediate distinct physiological functions in gastric neurotransmission.


Subject(s)
Myenteric Plexus , Stomach , Animals , Gastric Fundus/metabolism , In Situ Hybridization, Fluorescence , Mice , Myenteric Plexus/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach/innervation
8.
Gut ; 70(3): 464-475, 2021 03.
Article in English | MEDLINE | ID: mdl-32532891

ABSTRACT

OBJECTIVE: Tumour heterogeneity represents a major obstacle to accurate diagnosis and treatment in gastric adenocarcinoma (GA). Here, we report a systematic transcriptional atlas to delineate molecular and cellular heterogeneity in GA using single-cell RNA sequencing (scRNA-seq). DESIGN: We performed unbiased transcriptome-wide scRNA-seq analysis on 27 677 cells from 9 tumour and 3 non-tumour samples. Analysis results were validated using large-scale histological assays and bulk transcriptomic datasets. RESULTS: Our integrative analysis of tumour cells identified five cell subgroups with distinct expression profiles. A panel of differentiation-related genes reveals a high diversity of differentiation degrees within and between tumours. Low differentiation degrees can predict poor prognosis in GA. Among them, three subgroups exhibited different differentiation grade which corresponded well to histopathological features of Lauren's subtypes. Interestingly, the other two subgroups displayed unique transcriptome features. One subgroup expressing chief-cell markers (eg, LIPF and PGC) and RNF43 with Wnt/ß-catenin signalling pathway activated is consistent with the previously described entity fundic gland-type GA (chief cell-predominant, GA-FG-CCP). We further confirmed the presence of GA-FG-CCP in two public bulk datasets using transcriptomic profiles and histological images. The other subgroup specifically expressed immune-related signature genes (eg, LY6K and major histocompatibility complex class II) with the infection of Epstein-Barr virus. In addition, we also analysed non-malignant epithelium and provided molecular evidences for potential transition from gastric chief cells into MUC6+TFF2+ spasmolytic polypeptide expressing metaplasia. CONCLUSION: Altogether, our study offers valuable resource for deciphering gastric tumour heterogeneity, which will provide assistance for precision diagnosis and prognosis.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Sequence Analysis, RNA , Single-Cell Analysis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Adenocarcinoma/metabolism , Biomarkers, Tumor/genetics , Chief Cells, Gastric/metabolism , Chief Cells, Gastric/pathology , Gastric Fundus/metabolism , Gastric Fundus/pathology , Gene Expression Profiling , Humans , Stomach Neoplasms/metabolism , Transcriptome
9.
Anat Sci Int ; 96(2): 221-230, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33030698

ABSTRACT

Many studies have been conducted to determine the composition of the glycoconjugates of the mucus-secreting cells of the fundic glands of the stomach. However, the chief cells of these glands have been largely ignored because they secrete mainly zymogens with a lower glycosylation. The aim of this work was to analyze the glycoconjugates of the gastric chief cells by a battery of 17 different lectins, recognizing Fucose, N-acetylgalactosamine, Galactose, N-acetylneuraminic acid, N-acetylglucosamine and Mannose containing oligosaccharides. Histochemical techniques were performed with several lectins and also combined with two pre-treatments; ß-elimination, which removes O-linked oligosaccharides, and incubation with Peptide-N-Gycosidase F, which removes N-linked oligosaccharides. In addition, acid hydrolysis was performed before WGA histochemistry, and incubation with glucose oxidase before Con A labeling. Many lectins did not stain the chief cells. In addition, the presence of O-glycans in the apical cell membrane was demonstrated with the lectins AAL, HPA, MPA/MPL, PNA, RCA-I, and WGA. Some of these O-glycans were resistant to short-term ß-elimination pre-treatments. Mannose-binding lectins stained the basal cytoplasm of the chief cells. The level of glycosylation of the chief cells was lower than that of the mucous cells. The presence of O-glycans in the apical cell membrane is consistent with the presence of mucins such as MUC1 in the apical membrane of chief cells. Moreover, Mannose-binding lectins revealed N-glycosylation in the basal cytoplasm. The knowledge of gastric chief cell glycoconjugates is relevant because of their potential involvement not only in in physiological but also in pathological processes, such as cancer.


Subject(s)
Cell Membrane/metabolism , Chief Cells, Gastric/metabolism , Gastric Fundus/metabolism , Gastric Mucosa/metabolism , Glycoconjugates/metabolism , Animals , Lectins/metabolism , Rats
10.
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
11.
J Vasc Interv Radiol ; 31(9): 1483-1491, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32800664

ABSTRACT

PURPOSE: To examine safety and efficacy of bariatric arterial embolization (BAE) with x-ray-visible embolic microspheres (XEMs) and an antireflux catheter in swine. MATERIAL AND METHODS: BAE with selective infusion of XEMs (n = 6) or saline (n = 4, control) into gastric fundal arteries was performed under x-ray guidance. Weight and plasma hormone levels were measured at baseline and weekly for 4 weeks after embolization. Cone-beam CT images were acquired immediately after embolization and weekly for 4 weeks. Hormone-expressing cells in the stomach were assessed by immunohistochemical staining. RESULTS: BAE pigs lost weight 1 week after embolization followed by significantly impaired weight gain relative to control animals (14.3% vs 20.9% at 4 weeks, P = .03). Plasma ghrelin levels were significantly lower in BAE pigs than in control animals (1,221.6 pg/mL vs 1,706.2 pg/mL at 4 weeks, P < .01). XEMs were visible on x-ray and cone-beam CT during embolization, and radiopacity persisted over 4 weeks (165.5 HU at week 1 vs 158.5 HU at week 4, P = .9). Superficial mucosal ulcerations were noted in 1 of 6 BAE animals. Ghrelin-expressing cell counts were significantly lower in the gastric fundus (17.7 vs 36.8, P < .00001) and antrum (24.2 vs 46.3, P < .0001) of BAE pigs compared with control animals. Gastrin-expressing cell counts were markedly reduced in BAE pigs relative to control animals (98.5 vs 127.0, P < .02). Trichrome staining demonstrated significantly more fibrosis in BAE animals compared with control animals (13.8% vs 8.7%, P < .0001). CONCLUSIONS: XEMs enabled direct visualization of embolic material during and after embolization. BAE with XEMs and antireflux microcatheters was safe and effective.


Subject(s)
Appetite Regulation , Behavior, Animal , Catheters , Embolization, Therapeutic/instrumentation , Gastric Artery , Gastric Fundus/blood supply , Ghrelin/blood , Weight Loss , Animals , Cone-Beam Computed Tomography , Gastric Artery/diagnostic imaging , Gastric Fundus/metabolism , Gastric Fundus/pathology , Infusions, Intra-Arterial , Microspheres , Sus scrofa , Time Factors
12.
Arch Pharm Res ; 43(6): 666-675, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32607942

ABSTRACT

Dysfunction of gastrointestinal (GI) motility is a common complication in patients with diabetes mellitus (DM). Studies related to changes in fundus contraction induced by inhibitors in DM are not well known. Therefore, this study aimed to investigate the signaling pathways involved in the changes in the contraction of fundus smooth muscle obtained from control and DM rats. DM was induced by injecting streptozotocin (65 mg/kg) into Sprague-Dawley rats. The rats were sacrificed after 14 days. Fundus smooth muscle contraction was stimulated using electrical field stimulation (amplitude, 50 V; duration, 1 min; frequency, 2-20 Hz) and acetylcholine (0.1 mM). The inhibitor-mediated cell membrane was pre-treated with atropine, verapamil, methysergide, ketanserin, ondansetron, and GR 113808. Inhibitors related to intracellular signaling, such as U73122, chelerythrine, L-NNA, were also used. ML-9 and Y-27632 were identified as inhibitors of factors of myosin light chain (MLC). The contractility was observed to be lower in the DM group than in the control group. Further, the activities of phospholipase C (PLC), protein kinase C (PKC), and myosin light chain kinase (MLCK) were decreased in the DM group. DM reduced the activity of PLC, PKC, and MLCK, which resulted in a decrease in the contractility of the fundus smooth muscle. Therefore, our results present the mechanism of this DM-mediated GI disorder.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Gastric Fundus/metabolism , Muscle, Smooth/metabolism , Signal Transduction , Animals , Cell Membrane/metabolism , Diabetes Mellitus, Experimental/chemically induced , Male , Muscle Contraction , Rats , Rats, Sprague-Dawley , Streptozocin/administration & dosage
13.
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
14.
Tech Vasc Interv Radiol ; 23(1): 100657, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32192637

ABSTRACT

Obesity is a well-known major public health concern associated with significant morbidity and mortality. Bariatric arterial embolization (BAE) is a minimally invasive, image-guided therapy that targets hormones linked to obesity by defunctionalizing specific portions of the stomach with the delivery of embolics. The goal of BAE is to induce weight loss through a reduction of appetite-mediating hormones with transarterial embolization of the gastric fundus. This article will review the anatomical and technical considerations for successful BAE.


Subject(s)
Embolization, Therapeutic , Gastric Artery , Gastric Fundus/blood supply , Obesity/therapy , Appetite Regulation , Embolization, Therapeutic/adverse effects , Feeding Behavior , Gastric Fundus/metabolism , Ghrelin/metabolism , Humans , Obesity/metabolism , Obesity/physiopathology , Obesity/psychology , Signal Transduction , Treatment Outcome , Weight Loss
15.
Tech Vasc Interv Radiol ; 23(1): 100658, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32192638

ABSTRACT

Bariatric arterial embolization (BAE) is a novel technique that is investigated as an alternative, often supplementary, method for weight management. BAE reduces blood perfusion to the gastric fundus, and thus, reduces the production of appetite-inducing hormones. No randomized controlled trial has evaluated the efficacy of BAE to date. Available evidence from published studies include retrospective evaluations of patients undergoing left gastric artery embolization for gastrointestinal bleeding, and early prospective, single-arm clinical trials. Review of clinical data from human trials suggest an average weight loss of about 8-9 kg (ranging 7.6-22.0 kg), corresponding to 8-9% (ranging 4.8-17.2%) of the patients' baseline weight. Common complications include superficial gastric ulcers. Though uncommon, gastric perforation and splenic infarct are important major complication that may arise after left gastric artery embolization. Overall, BAE is an effective, relatively safe procedure that may be associated with clinically significant weight loss in patients with obesity.


Subject(s)
Embolization, Therapeutic , Gastric Artery , Gastric Fundus/blood supply , Obesity/therapy , Appetite Regulation , Clinical Trials as Topic , Embolization, Therapeutic/adverse effects , Evidence-Based Medicine , Feeding Behavior , Gastric Fundus/metabolism , Ghrelin/metabolism , Humans , Obesity/metabolism , Obesity/physiopathology , Obesity/psychology , Signal Transduction , Treatment Outcome , Weight Loss
16.
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
17.
Nat Commun ; 10(1): 39, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604760

ABSTRACT

The human gastric mucosa is the most active layer of the stomach wall, involved in food digestion, metabolic processes and gastric carcinogenesis. Anatomically, the human stomach is divided into seven regions, but the protein basis for cellular specialization is not well understood. Here we present a global analysis of protein profiles of 82 apparently normal mucosa samples obtained from living individuals by endoscopic stomach biopsy. We identify 6,258 high-confidence proteins and estimate the ranges of protein expression in the seven stomach regions, presenting a region-resolved proteome reference map of the near normal, human stomach. Furthermore, we measure mucosa protein profiles of tumor and tumor nearby tissues (TNT) from 58 gastric cancer patients, enabling comparisons between tumor, TNT, and normal tissue. These datasets provide a rich resource for the gastrointestinal tract research community to investigate the molecular basis for region-specific functions in mucosa physiology and pathology including gastric cancer.


Subject(s)
Gastric Mucosa/metabolism , Neoplasm Proteins/analysis , Proteome/analysis , Stomach Neoplasms/pathology , Biopsy , Carcinogenesis/pathology , Cardia/metabolism , Cardia/pathology , Datasets as Topic , Gastric Fundus/metabolism , Gastric Fundus/pathology , Gastric Mucosa/pathology , Gastroscopy , Humans , Neoplasm Proteins/metabolism , Proteome/metabolism , Proteomics/methods , Pyloric Antrum/metabolism , Pyloric Antrum/pathology , Pylorus/metabolism , Pylorus/pathology
18.
Nat Protoc ; 14(1): 28-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30470820

ABSTRACT

The human stomach contains two primary domains: the corpus, which contains the fundic epithelium, and the antrum. Each of these domains has distinct cell types and functions, and therefore each presents with unique disease pathologies. Here, we detail two protocols to differentiate human pluripotent stem cells (hPSCs) into human gastric organoids (hGOs) that recapitulate both domains. Both protocols begin with the differentiation of hPSCs into definitive endoderm (DE) using activin A, followed by the generation of free-floating 3D posterior foregut spheroids using FGF4, Wnt pathway agonist CHIR99021 (CHIR), BMP pathway antagonist Noggin, and retinoic acid. Embedding spheroids in Matrigel and continuing 3D growth in epidermal growth factor (EGF)-containing medium for 4 weeks results in antral hGOs (hAGOs). To obtain fundic hGOs (hFGOs), spheroids are additionally treated with CHIR and FGF10. Induced differentiation of acid-secreting parietal cells in hFGOs requires temporal treatment of BMP4 and the MEK inhibitor PD0325901 for 48 h on protocol day 30. In total, it takes ~34 d to generate hGOs from hPSCs. To date, this is the only approach that generates functional human differentiated gastric cells de novo from hPSCs.


Subject(s)
Cell Culture Techniques , Endoderm/cytology , Epithelial Cells/cytology , Gastric Fundus/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Pyloric Antrum/cytology , Activins/pharmacology , Benzamides/pharmacology , Carrier Proteins/pharmacology , Cell Differentiation , Collagen/chemistry , Culture Media/chemistry , Culture Media/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Drug Combinations , Endoderm/drug effects , Endoderm/metabolism , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblast Growth Factor 10/pharmacology , Fibroblast Growth Factor 4/pharmacology , Gastric Fundus/metabolism , Humans , Laminin/chemistry , Organ Specificity , Organoids/drug effects , Organoids/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proteoglycans/chemistry , Pyloric Antrum/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Tretinoin/pharmacology , Wnt Signaling Pathway/drug effects
19.
Cell Tissue Res ; 376(1): 37-49, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30467709

ABSTRACT

Gastric endocrine cell hormones contribute to the control of the stomach and to signalling to the brain. In other gut regions, enteroendocrine cells (EECs) exhibit extensive patterns of colocalisation of hormones. In the current study, we characterise EECs in the human gastric fundus and corpus. We utilise immunohistochemistry to investigate EECs with antibodies to ghrelin, serotonin (5-HT), somatostatin, peptide YY (PYY), glucagon-like peptide 1, calbindin, gastrin and pancreastatin, the latter as a marker of enterochromaffin-like (ECL) cells. EECs were mainly located in regions of the gastric glands populated by parietal cells. Gastrin cells were absent and PYY cells were very rare. Except for about 25% of 5-HT cells being a subpopulation of ECL cells marked by pancreastatin, colocalisation of hormones in gastric EECs was infrequent. Ghrelin cells were distributed throughout the fundus and corpus; most were basally located in the glands, often very close to parietal cells and were closed cells i.e., not in contact with the lumen. A small proportion had long processes located close to the base of the mucosal epithelium. The 5-HT cells were of at least three types: small, round, closed cells; cells with multiple, often very long, processes; and a subgroup of ECL cells. Processes were in contact with their surrounding cells, including parietal cells. Mast cells had very weak or no 5-HT immunoreactivity. Somatostatin cells were a closed type with long processes. In conclusion, four major chemically defined EEC types occurred in the human oxyntic mucosa. Within each group were cells with distinct morphologies and relationships to other mucosal cells.


Subject(s)
Enteroendocrine Cells , Gastric Fundus , Gastrointestinal Hormones/analysis , Enteroendocrine Cells/chemistry , Enteroendocrine Cells/cytology , Female , Gastric Fundus/cytology , Gastric Fundus/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Obesity/surgery
20.
Obes Surg ; 29(1): 76-83, 2019 01.
Article in English | MEDLINE | ID: mdl-30178156

ABSTRACT

BACKGROUND: Weight loss after laparoscopic sleeve gastrectomy (LSG) has been mainly attributed to the restriction of gastric volume; however; other factors may contribute to weight loss after LSG. This study aimed to investigate the correlation between the number of ghrelin-secreting cells in the gastric fundus and excess weight loss (EWL) at 12 months after LSG. METHODS: The surface area of the gastric fundus was measured postoperatively in square centimeter. Histopathologic examination of the gastric fundus was made to estimate the number of ghrelin-secreting cells per square centimeter then was multiplied by the surface area of the fundus to calculate the total number of ghrelin-secreting cells in the fundus. The number of ghrelin-secreting cells was correlated with EWL and BMI at 12 months postoperatively. RESULTS: The present study included 39 patients of a mean age of 33.7 years. The mean %EWL at 12 months was 59.7 ± 12.7. The mean total number of ghrelin-producing cells in the gastric fundus was 26,228.4 ± 16,995.3. The total number of ghrelin-secreting cells had a weak positive correlation with BMI at 12 months (r = 0.2891, p = 0.07), and weak negative correlation with %EWL (r = - 0.1592, p = 0.33). CONCLUSION: There was a weak correlation between the total number of ghrelin-producing cells in the gastric fundus and plasma ghrelin levels with EWL after LSG.


Subject(s)
Gastrectomy/statistics & numerical data , Gastric Fundus , Ghrelin , Obesity, Morbid , Weight Loss/physiology , Adult , Cohort Studies , Gastric Fundus/cytology , Gastric Fundus/metabolism , Gastric Fundus/surgery , Ghrelin/metabolism , Humans , Obesity, Morbid/epidemiology , Obesity, Morbid/metabolism , Obesity, Morbid/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...