Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 378
Filter
1.
Nature ; 541(7636): 182-187, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28052057

ABSTRACT

Despite the global prevalence of gastric disease, there are few adequate models in which to study the fundus epithelium of the human stomach. We differentiated human pluripotent stem cells (hPSCs) into gastric organoids containing fundic epithelium by first identifying and then recapitulating key events in embryonic fundus development. We found that disruption of Wnt/ß-catenin signalling in mouse embryos led to conversion of fundic to antral epithelium, and that ß-catenin activation in hPSC-derived foregut progenitors promoted the development of human fundic-type gastric organoids (hFGOs). We then used hFGOs to identify temporally distinct roles for multiple signalling pathways in epithelial morphogenesis and differentiation of fundic cell types, including chief cells and functional parietal cells. hFGOs are a powerful model for studying the development of the human fundus and the molecular bases of human gastric physiology and pathophysiology, and also represent a new platform for drug discovery.


Subject(s)
Gastric Fundus/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Body Patterning , Cell Differentiation , Cell Lineage , Drug Discovery/methods , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/embryology , Epithelium/metabolism , Female , Gastric Fundus/cytology , Gastric Fundus/embryology , Homeodomain Proteins/metabolism , Humans , Male , Mice , Organoids/cytology , Organoids/embryology , Organoids/metabolism , Parietal Cells, Gastric/cytology , Parietal Cells, Gastric/metabolism , Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Trans-Activators/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/agonists
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Cell Physiol Biochem ; 51(2): 664-680, 2018.
Article in English | MEDLINE | ID: mdl-30463068

ABSTRACT

BACKGROUND/AIMS: Roux-en-Y Gastric Bypass, RYGB, is the most effective strategy to control body weight in morbid obesity. RYGB leads to rapid improvement of glycemic status and weight loss, which are largely attributed to the alteration of gastrointestinal hormones including ghrelin. The current study examined potential mechanisms of altered ghrelin synthesis after RYGB. METHODS: Gastric mammalian target of rapamycin (mTOR) signaling, ghrelin synthesis and secretion were determined in lean or obese male mice with or without RYGB operation, as well as in obese patients pre- and post-RYGB surgery. Ghrelin expression and mTOR signaling were investigated by western blotting and immunohistochemistry. Ghrelin mRNA levels were detected by real-time PCR. Plasma ghrelin was measured by enzyme immunoassay. RESULTS: mTOR activity in the gastric fundus was significantly lower than in the forestomachs. Both of them were decreased after 24h fasting. A significant negative correlation was found between gastric levels of phospho-S6 (phospho-S6 ribosomal protein) and proghrelin during changes of energy status. mTOR activity was activated, whereas ghrelin expression was inhibited by Roux-en-Y Gastric Bypass in both rodents and human beings. Increment of ghrelin synthesis and decline of mTOR signaling induced by rapamycin were significantly reversed by RYGB in both lean and obese mice. Administration of Ad-S6K1 (adenovirus-mediated p70 ribosomal protein subunit 6 kinase 1) from tail vein suppressed the expression of ghrelin in RYGB-operated mice relative to control animals. CONCLUSION: mTOR is therefore a gastric fuel sensor whose activity is linked to the regulation of ghrelin after Roux-en-Y Gastric Bypass.


Subject(s)
Ghrelin/blood , Mechanistic Target of Rapamycin Complex 1/metabolism , Obesity, Morbid/pathology , Adult , Animals , Body Weight , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diet, High-Fat , Energy Metabolism , Gastric Bypass , Gastric Fundus/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Male , Mice, Inbred C57BL , Middle Aged , Obesity, Morbid/complications , Obesity, Morbid/surgery , Signal Transduction/drug effects , Sirolimus/pharmacology
11.
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
12.
Chirurgia (Bucur) ; 112(1): 33-38, 2017.
Article in English | MEDLINE | ID: mdl-28266290

ABSTRACT

Introduction: Obesity is currently an important health problem that is rapidly increasing worldwide. In recent years, the number of obesity-related surgeries has increased. The most common type of obesity-related surgery is laparoscopic sleeve gastrectomy (LSG). The aim of this study was to compare the genetic expression of the hormone ghrelin in different parts of the stomach. Materials and Methods: Nineteen obese patients who underwent LSG were examined in this study. Fat tissue from two different parts of the stomach, the fundus and the upper part of the fundus, were analysed by enzyme-linked immunosorbent assay (ELISA). The ribonucleic acid (RNA) isolation, complementary DNA (cDNA) and real-time quantitative polymerase chain reaction (RQ-PCR) techniques were applied. Additionally, a human ghrelin ELISA kit was used to measure ghrelin in obese patients. The ghrelin levels of fat tissue from the fundus and upper part of the fundus were statistically compared. Results: In all 19 patients, the average ghrelin level in the fundus was greater than 30. The average ghrelin level of the fat pad, which is located in the upper part of the fundus, was greater than 30 for 4 patients; the average level was approximately 5 in the remaining patients. A statistically significant difference in the ghrelin level was found between the fundus and the fundus fat tissue. CONCLUSION: Collection of fundus fat tissue is not routinely performed during LSG. However, ghrelin hormone elevation in this tissue may require collection of fundus tissue during surgery.


Subject(s)
Gastrectomy , Gastric Fundus/metabolism , Ghrelin/metabolism , Laparoscopy , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Body Mass Index , Female , Gastrectomy/methods , Humans , Male , Middle Aged , Prospective Studies , Weight Loss
13.
Gut ; 65(1): 73-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25410165

ABSTRACT

OBJECTIVE: Impaired gastric accommodation is reported in patients with functional dyspepsia (FD). Previous findings in postinfectious patients with FD suggest that low-grade inflammation and dysfunction of nitrergic nerves play a role in impaired accommodation. To date, spontaneous animal models to study the relationship between these changes are lacking. We hypothesise that the normoglycaemic BioBreeding diabetes-prone (BB-DP) rat provides an animal model of inflammation-induced impaired gastric motor function. DESIGN: Control diabetes-resistant biobreeding, normoglycaemic and hyperglycaemic BB-DP rats were sacrificed at the age of 30, 70 and 220 days and gastric fundus tissue was harvested to study nitrergic motor control, inflammation and expression of neuronal isoform of nitric oxide synthase (nNOS) and inducible isoform of nitric oxide synthase (iNOS). Nutrient-induced changes in intragastric pressure (IGP) were measured in normoglycaemic BB-DP rats to study accommodation. RESULTS: No differences in nitrergic function and inflammation were observed between BB-DP and control rats at 30 days. The nitrergic component of the fundic muscle relaxation was reduced in BB-DP rats of 70 and 220 days. This was accompanied by a significant loss of nNOS proteins. IGP significantly increased during nutrient infusion in BB-DP rats of 220 days, indicating impaired accommodation. Infiltration of polymorphonuclear cells, increased myeloperoxidase activity and increased expression of iNOS was observed in the fundic mucosa and muscularis propria of 70-day-old and 220-day-old BB-DP rats. CONCLUSIONS: BB-DP rats of 220 days display altered fundic motor control and impaired accommodation, which is least partially explained by loss of nitrergic function. This may be related to inflammatory changes in the neuromuscular layer, suggesting that normoglycaemic BB-DP rats provide a spontaneous model for inflammation-induced impaired gastric accommodation.


Subject(s)
Disease Models, Animal , Dyspepsia/physiopathology , Nitrergic Neurons/physiology , Rats, Inbred BB/physiology , Stomach/physiopathology , Animals , Biomarkers/metabolism , Blotting, Western , Gastric Fundus/innervation , Gastric Fundus/metabolism , Gastric Fundus/physiopathology , Gastric Mucosa/metabolism , Hyperglycemia/physiopathology , Immunohistochemistry , Nitrergic Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stomach/innervation
14.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G1169-75, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27125274

ABSTRACT

Feeding intolerance is a common issue in the care of preterm neonates. The condition manifests as delayed emptying of gastric contents and represents a therapeutic challenge, since the factors accounting for its manifestations are unknown. The main goal of this study was to comparatively investigate the age-related function of rat gastric and pyloric smooth muscle and their putative regulators. We hypothesized that a reduced gastric muscle contraction potential early in life contributes to the delayed gastric emptying of the newborn. Newborn and adult rat gastric (fundus) and pyloric sphincter tissues were comparatively studied in vitro. Shortening of the tissue-specific dissociated smooth muscle cell was evaluated, and expression of the key regulatory proteins Rho-associated kinase 2 and myosin light chain kinase was determined. Gastric and pyloric smooth muscle cell shortening was significantly greater in the adult than the respective newborn counterpart. Expression of myosin light chain kinase and Rho-associated kinase 2 was developmentally regulated and increased with age. Pyloric sphincter muscle expresses a higher neuronal nitric oxide synthase and phosphorylated vasodilator-stimulated phosphoprotein content in newborn than adult tissue. Compared with later in life, the newborn rat gastropyloric muscle has a Ca(2+)-related reduced potential for contraction and the pyloric sphincter relaxation-dependent modulators are overexpressed. To the extent that these rodent data can be extrapolated to humans, the delayed gastric emptying in the newborn reflects reduced stomach muscle contraction potential, as opposed to increased pyloric sphincter tone.


Subject(s)
Gastric Emptying , Gastric Fundus/physiology , Pylorus/physiology , Animals , Gastric Fundus/growth & development , Gastric Fundus/metabolism , Muscle Contraction , Muscle, Smooth/growth & development , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Pylorus/growth & development , Pylorus/metabolism , Rats , Rats, Sprague-Dawley , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
15.
Am J Physiol Regul Integr Comp Physiol ; 310(3): R235-42, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26608656

ABSTRACT

Gastric hypersensitivity is one of the key contributors to the postprandial symptoms of epigastric pain/discomfort, satiety, and fullness in functional dyspepsia patients. Epidemiological studies found that adverse early-life experiences are risk factors for the development of gastric hypersensitivity. Preclinical studies found that neonatal colon inflammation elevates plasma norepinephrine (NE), which upregulates expression of nerve growth factor (NGF) in the muscularis externa of the gastric fundus. Our goal was to investigate the cellular mechanisms by which NE upregulates the expression of NGF in gastric hypersensitive (GHS) rats, which were subjected previously to neonatal colon inflammation. Neonatal colon inflammation upregulated NGF protein, but not mRNA, in the gastric fundus of GHS rats. Western blotting showed upregulation of p110γ of phosphatidylinositol 4,5-bisphosphate 3-kinase (PI3K), phosphoinositide-dependent kinase-1 (PDK1), pAKT(Ser473), and phosphorylated 4E-binding protein (p4E-BP1)(Thr70), suggesting AKT activation and enhanced NGF protein translation. AKT inhibitor MK-2206 blocked the upregulation of NGF in the fundus of GHS rats. Matrix metalloproteinase 9 (MMP-9), the major NGF-degrading protease, was suppressed, indicating that NGF degradation was impeded. Incubation of fundus muscularis externa with NE upregulated NGF by modulating the protein translation and degradation pathways. Yohimbine, an α2-adrenergic receptor antagonist, upregulated plasma NE and NGF expression by activating the protein translation and degradation pathways in naive rats. In contrast, a cocktail of adrenergic receptor antagonists suppressed the upregulation of NGF by blocking the activation of the protein translation and degradation pathways. Our findings provide evidence that the elevation of plasma NE induces NGF expression in the gastric fundus.


Subject(s)
Abdominal Pain/metabolism , Colitis/metabolism , Colon/metabolism , Gastric Fundus/metabolism , Nerve Growth Factor/metabolism , Norepinephrine/blood , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Abdominal Pain/etiology , Abdominal Pain/physiopathology , Adrenergic Antagonists/pharmacology , Age Factors , Animals , Animals, Newborn , Carrier Proteins/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Colitis/chemically induced , Colitis/physiopathology , Colon/drug effects , Colon/innervation , Colon/physiopathology , Disease Models, Animal , Gastric Fundus/drug effects , Gastric Fundus/innervation , Gastric Fundus/physiopathology , Intracellular Signaling Peptides and Proteins , Male , Matrix Metalloproteinase 9/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction , Trinitrobenzenesulfonic Acid , Up-Regulation
16.
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
17.
J Pharmacol Sci ; 132(3): 181-186, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27816547

ABSTRACT

In gastric smooth muscles, the released Ca2+ activates the contractile proteins and Ca2+ taken up from the cytosol cause relaxation. The Na+/Ca2+ exchanger (NCX) is an antiporter membrane protein that controls Ca2+ influx and efflux across the membrane. However, the possible relation of NCX in gastric fundus motility is largely unknown. Here, we investigated electric field stimulation (EFS)-induced relaxations in the circular muscles of the gastric fundus in smooth muscle-specific NCX1 transgenic mice (Tg). EFS caused a bi-phasic response, transient and sustained relaxation. The sustained relaxation prolonged for an extended period after the end of the stimulus. EFS-induced transient relaxation and sustained relaxation were greater in Tg than in wild-type mice (WT). Disruption of nitric oxide component by N-nitro-l-arginine, EFS-induced transient and sustained relaxations caused still marked in Tg compared to WT. Inhibition of PACAP by antagonist, EFS-induced sustained relaxation in Tg was not seen, similar to WT. Nevertheless, transient relaxation remained more pronounced in Tg than in WT. Next, we examined responses to NO and PACAP in smooth muscles. The magnitudes of NOR-1, which generates NO, and PACAP-induced relaxations were greater in Tg than in WT. In this study, we demonstrate that NCX1 regulates gastric fundus motility.


Subject(s)
Gastric Fundus/physiology , Sodium-Calcium Exchanger/biosynthesis , Animals , Electric Stimulation , Gastric Fundus/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Relaxation/physiology , Muscle, Smooth/metabolism , Muscle, Smooth/physiology
18.
Bratisl Lek Listy ; 117(6): 355-7, 2016.
Article in English | MEDLINE | ID: mdl-27546369

ABSTRACT

BACKGROUND: Ghrelin is a 28-amino acid peptide that is isolated mainly from the oxyntic glands of the stomach, especially fundus. Ghrelin administration, either centrally or peripherally, increases food intake and body weight in both rodents and humans. This study evaluates the effects of fundus resection and sclerosing agent injection on ghrelin level and weight loss. MATERIAL AND METHODS: Thirty rats were divided into three groups. In group 1, NaCl was injected into the submucosal space at the gastric fundus while in Group 2, a sclerosing agent was injected into the latter site. In group 3, gastric fundus was resected. Ghrelin levels and weight were recorded. RESULTS: In group 1, rats continued gaining weight and ghrelin levels stayed stable. In group 2, rats' weight and ghrelin levels stayed stable and in group 3, while weight stayed stable, ghrelin levels decreased significantly. CONCLUSION: In rats, the resection of fundus stabilizes weight gain and decreases ghrelin levels. However, in sclerotherapy, although weight gain was stabilized, there was no decrease in ghrelin levels. In humans, the effect of fundus resection on weight gain can usher in a new era of investigation (Tab. 2, Ref. 16).


Subject(s)
Body Weight/drug effects , Gastric Fundus/drug effects , Gastric Mucosa/drug effects , Ghrelin/drug effects , Sclerosing Solutions/pharmacology , Weight Loss/drug effects , Animals , Gastric Fundus/metabolism , Gastric Fundus/surgery , Gastric Mucosa/metabolism , Gastric Mucosa/surgery , Ghrelin/metabolism , Rats , Rats, Wistar
19.
Eksp Klin Gastroenterol ; (7): 64-6, 2014.
Article in Russian | MEDLINE | ID: mdl-25842408

ABSTRACT

The researchers are investigated in 32 white male, rats by means of electron-microscopic methods and morphometric analysis. The sharp reduction acid produceds functions parietal hutches after total colonectomy, her (its) stability at early periods after operation are certainly conditioned inflammatory-distrofic change on the part of secretory membranes, and particularly mitochondria of the device.


Subject(s)
Adaptation, Physiological , Colectomy , Gastric Fundus/physiopathology , Parietal Cells, Gastric/ultrastructure , Animals , Gastric Acid/metabolism , Gastric Fundus/metabolism , Gastric Fundus/ultrastructure , Male , Parietal Cells, Gastric/metabolism , Rats
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL