Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 248
Filter
1.
Gastroenterology ; 162(2): 415-430, 2022 02.
Article in English | MEDLINE | ID: mdl-34728185

ABSTRACT

The mucosa of the body of the stomach (ie, the gastric corpus) uses 2 overlapping, depth-dependent mechanisms to respond to injury. Superficial injury heals via surface cells with histopathologic changes like foveolar hyperplasia. Deeper, usually chronic, injury/inflammation, most frequently induced by the carcinogenic bacteria Helicobacter pylori, elicits glandular histopathologic alterations, initially manifesting as pyloric (also known as pseudopyloric) metaplasia. In this pyloric metaplasia, corpus glands become antrum (pylorus)-like with loss of acid-secreting parietal cells (atrophic gastritis), expansion of foveolar cells, and reprogramming of digestive enzyme-secreting chief cells into deep antral gland-like mucous cells. After acute parietal cell loss, chief cells can reprogram through an orderly stepwise progression (paligenosis) initiated by interleukin-13-secreting innate lymphoid cells (ILC2s). First, massive lysosomal activation helps mitigate reactive oxygen species and remove damaged organelles. Second, mucus and wound-healing proteins (eg, TFF2) and other transcriptional alterations are induced, at which point the reprogrammed chief cells are recognized as mucus-secreting spasmolytic polypeptide-expressing metaplasia cells. In chronic severe injury, glands with pyloric metaplasia can harbor both actively proliferating spasmolytic polypeptide-expressing metaplasia cells and eventually intestine-like cells. Gastric glands with such lineage confusion (mixed incomplete intestinal metaplasia and proliferative spasmolytic polypeptide-expressing metaplasia) may be at particular risk for progression to dysplasia and cancer. A pyloric-like pattern of metaplasia after injury also occurs in other gastrointestinal organs including esophagus, pancreas, and intestines, and the paligenosis program itself seems broadly conserved across tissues and species. Here we discuss aspects of metaplasia in stomach, incorporating data derived from animal models and work on human cells and tissues in correlation with diagnostic and clinical implications.


Subject(s)
Cell Plasticity/physiology , Cellular Reprogramming/physiology , Gastric Mucosa/physiology , Regeneration/physiology , Stomach/physiology , Animals , Gastric Mucosa/cytology , Gastric Mucosa/pathology , Helicobacter Infections/physiopathology , Humans , Hyperplasia , Metaplasia , Parietal Cells, Gastric/physiology , Stomach/cytology , Stomach/pathology
2.
Int J Exp Pathol ; 101(6): 230-247, 2020 12.
Article in English | MEDLINE | ID: mdl-32985762

ABSTRACT

In clinical medicine, indomethacin (IND, a non-steroidal anti-inflammatory drug) is used variously in the treatment of severe osteoarthritis, rheumatoid arthritis, gouty arthritis or ankylosing spondylitis. A common complication found alongside the therapeutic characteristics is gastric mucosal damage. This complication is mediated through apoptosis and autophagy of the gastrointestinal mucosal epithelium. Apoptosis and autophagy are critical homeostatic pathways catalysed by caspases downstream of the gastrointestinal mucosal epithelial injury. Both act through molecular signalling pathways characterized by the initiation, mediation, execution and regulation of the cell regulatory cycle. In this study we hypothesized that dysregulated apoptosis and autophagy are associated with IND-induced gastric damage. We examined the spectra of in vivo experimental gastric ulcers in male Sprague-Dawley rats through gastric gavage of IND. Following an 18-hour fast, IND was administered to experimental rats. They were sacrificed at 3-, 6- and 12-hour intervals. Parietal cells (H+ , K+ -ATPase ß-subunit assay) and apoptosis (TUNEL assay) were determined. The expression of apoptosis-signalling caspase (caspases 3, 8, 9 and 12), DNA damage (anti-phospho-histone H2A.X) and autophagy (MAP-LC3, LAMP-1 and cathepsin B)-related molecules in gastric mucosal cells was examined. The administration of IND was associated with gastric mucosal erosions and ulcerations mainly involving the gastric parietal cells (PCs) of the isthmic and upper neck regions and a time-dependent gradual increase in the number of apoptotic PCs with the induction of both apoptotic (upregulation of caspases 3 and 8) cell death and autophagic (MAP-LC3-II, LAMP-1 and cathepsin B) cell death. Autophagy induced by fasting and IND 3 hours initially prompted the degradation of caspase 8. After 6 and 12 hours, damping down of autophagic activity occurred, resulting in the upregulation of active caspase 8 and its nuclear translocation. In conclusion we report that IND can induce time-dependent apoptotic and autophagic cell death of PCs. Our study provides the first indication of the interactions between these two homeostatic pathways in this context.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Indomethacin/pharmacology , Signal Transduction/drug effects , Animals , Autophagy/drug effects , Cell Death/drug effects , DNA Damage/drug effects , Gastric Mucosa/physiology , Male , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/physiology , Rats , Rats, Sprague-Dawley
3.
Microsc Microanal ; 26(4): 846-854, 2020 08.
Article in English | MEDLINE | ID: mdl-32458772

ABSTRACT

Suicidal behavior in termite workers is an extreme defensive strategy, probably a consequence of having a low number of soldiers available in the colony and there being high predation from enemies. We investigated the suicidal mechanism in workers of the Neotropical termite Neocapritermes opacus, which involves salivary gland autothysis followed by body cuticle rupture and the release of a defensive secretion. Autothysis was triggered by a physical stimulus such as a soldier bite that causes the protrusion of the salivary acini, burst reservoirs, and foregut. Histochemical and ultrastructural analyses showed salivary acini composed of peripheral parietal cells and two types of central cells, types I and II. Type I cells are filled with large electron-lucent secretory vesicles, which reacted positively to bromophenol blue and xylidine-Ponceau tests, indicating the occurrence of proteins. Type II cells are elongated and display smaller apical secretory vesicles. Parietal cells present an intracellular canaliculus with dense microvilli and cytoplasm rich in mitochondria and large electron-dense vesicles, which may participate in the self-destructive mechanism. Worker suicidal behavior was previously reported for N. taracua and N. braziliensis. N. opacus is a new species in which a salivary weapon has been developed and factors contributing to this altruistic response are discussed.


Subject(s)
Behavior, Animal/physiology , Isoptera/physiology , Salivary Glands/physiology , Animals , Parietal Cells, Gastric/physiology
4.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G36-G42, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29517927

ABSTRACT

The H+,K+-ATPase was identified as the primary proton secretory pathway in the gastric parietal cell and is the pharmacological target of agents suppressing acid secretion. Recently, we identified a second acid secretory protein expressed in the parietal cell, the vacuolar H+-ATPase (V-type ATPase). The aim of the present study was to further characterize H+-ATPase activation by modulations in extracellular calcium via the calcium sensing receptor (CaSR). Isolated gastric glands were loaded with the pH indicator dye BCECF-AM [2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein acetoxymethyl ester] to measure intracellular pH. Experiments were conducted in the absence of sodium and potassium to monitor H+-ATPase-specific transport activity. CaSR was activated with the calcimimetic R568 (400 nM) and/or by modulations in extracellular Ca2+. Elevation in calcium concentrations increased proton extrusion from the gastric parietal cell. Allosteric modification of the CaSR via R568 and calcium increased vacuolar H+-ATPase activity significantly (ΔpH/minlowCa2+(0.1mM) = 0.001 ± 0.001, ΔpH/minnormalCa2+(1.0mM) = 0.033 ± 0.004, ΔpH/minhighCa2+(5.0mM) = 0.051 ± 0.005). Carbachol significantly suppressed calcium-induced gastric acid secretion via the H+-ATPase under sodium- and potassium-free conditions. We conclude that the V-type H+-ATPase is tightly linked to CaSR activation. We observed that proton pump inhibitor (PPI) exposure does not modulate H+-ATPase activity. This elevated blood calcium activation of the H+-ATPase could provide an explanation for recurrent reflux symptoms while taking a PPI therapy. NEW & NOTEWORTHY This study emphasizes the role of the H+-ATPase in acid secretion. We further demonstrate the modification of this proton excretion pathway by extracellular calcium and the activation of the calcium sensing receptor CaSR. The novelty of this paper is based on the modulation of the H+-ATPase via both extracellular Ca (activation) and the classical secretagogues histamine and carbachol (inactivation). Both activation and inactivation of this proton pump are independent of PPI modulation.


Subject(s)
Calcium , Enzyme Activation , H(+)-K(+)-Exchanging ATPase/metabolism , Parietal Cells, Gastric , Proton Pump Inhibitors/pharmacology , Proton Pumps , Receptors, Calcium-Sensing/metabolism , Animals , Calcium/blood , Calcium/metabolism , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Gastric Acid/metabolism , Histamine/metabolism , Ion Transport/drug effects , Ion Transport/physiology , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/physiology , Proton Pumps/drug effects , Proton Pumps/metabolism , Rats , Rats, Sprague-Dawley , Secretory Pathway/drug effects , Secretory Pathway/physiology
5.
Proc Natl Acad Sci U S A ; 114(30): E6260-E6269, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696284

ABSTRACT

Caffeine, generally known as a stimulant of gastric acid secretion (GAS), is a bitter-tasting compound that activates several taste type 2 bitter receptors (TAS2Rs). TAS2Rs are expressed in the mouth and in several extraoral sites, e.g., in the gastrointestinal tract, in which their functional role still needs to be clarified. We hypothesized that caffeine evokes effects on GAS by activation of oral and gastric TAS2Rs and demonstrate that caffeine, when administered encapsulated, stimulates GAS, whereas oral administration of a caffeine solution delays GAS in healthy human subjects. Correlation analysis of data obtained from ingestion of the caffeine solution revealed an association between the magnitude of the GAS response and the perceived bitterness, suggesting a functional role of oral TAS2Rs in GAS. Expression of TAS2Rs, including cognate TAS2Rs for caffeine, was shown in human gastric epithelial cells of the corpus/fundus and in HGT-1 cells, a model for the study of GAS. In HGT-1 cells, various bitter compounds as well as caffeine stimulated proton secretion, whereby the caffeine-evoked effect was (i) shown to depend on one of its cognate receptor, TAS2R43, and adenylyl cyclase; and (ii) reduced by homoeriodictyol (HED), a known inhibitor of caffeine's bitter taste. This inhibitory effect of HED on caffeine-induced GAS was verified in healthy human subjects. These findings (i) demonstrate that bitter taste receptors in the stomach and the oral cavity are involved in the regulation of GAS and (ii) suggest that bitter tastants and bitter-masking compounds could be potentially useful therapeutics to regulate gastric pH.


Subject(s)
Caffeine/pharmacology , Gastric Acid/metabolism , Parietal Cells, Gastric/physiology , Flavones/pharmacology , Humans , Parietal Cells, Gastric/metabolism , Receptors, G-Protein-Coupled/physiology , Taste
6.
Gastroenterology ; 152(4): 762-766.e7, 2017 03.
Article in English | MEDLINE | ID: mdl-27932312

ABSTRACT

Parietal cell atrophy is considered to cause metaplasia in the stomach. We developed mice that express the diphtheria toxin receptor specifically in parietal cells to induce their death, and found this to increase proliferation in the normal stem cell zone and neck but not to cause metaplastic reprogramming of chief cells. Furthermore, the metaplasia-inducing agents tamoxifen or DMP-777 still induced metaplasia even after previous destruction of parietal cells by diphtheria toxin. Atrophy of parietal cells alone therefore is not sufficient to induce metaplasia: completion of metaplastic reprogramming of chief cells requires mechanisms beyond parietal cell injury or death.


Subject(s)
Apoptosis , Chief Cells, Gastric/pathology , Parietal Cells, Gastric/pathology , Parietal Cells, Gastric/physiology , Stomach/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Atrophy/chemically induced , Azetidines , Cell Proliferation , Cellular Reprogramming , Chief Cells, Gastric/metabolism , Diphtheria Toxin/pharmacology , Heparin-binding EGF-like Growth Factor/genetics , Intercellular Signaling Peptides and Proteins , Intrinsic Factor/metabolism , Metaplasia/chemically induced , Metaplasia/genetics , Metaplasia/metabolism , Mice , Parietal Cells, Gastric/drug effects , Peptides/metabolism , Piperazines , Plant Lectins/metabolism , Tamoxifen
7.
Vet Res ; 47(1): 101, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27756386

ABSTRACT

The stomach of pigs at slaughter age is often colonized by Helicobacter (H.) suis, which is also the most prevalent gastric non-H. pylori Helicobacter (NHPH) species in humans. It is associated with chronic gastritis, gastric ulceration and other gastric pathological changes in both hosts. Parietal cells are highly specialized, terminally differentiated epithelial cells responsible for gastric acid secretion and regulation. Dysfunction of these cells is closely associated with gastric pathology and disease. Here we describe a method for isolation and culture of viable and responsive parietal cells from slaughterhouse pigs. In addition, we investigated the interactions between H. suis and gastric parietal cells both in H. suis-infected six-month-old slaughter pigs, as well as in our in vitro parietal cell model. A close interaction of H. suis and parietal cells was observed in the fundic region of stomachs from H. suis positive pigs. The bacterium was shown to be able to directly interfere with cultured porcine parietal cells, causing a significant impairment of cell viability. Transcriptional levels of Atp4a, essential for gastric acid secretion, showed a trend towards an up-regulation in H. suis positive pigs compared to H. suis-negative pigs. In addition, sonic hedgehog, an important factor involved in gastric epithelial differentiation, gastric mucosal repair, and stomach homeostasis, was also significantly up-regulated in H. suis positive pigs. In conclusion, this study describes a successful approach for the isolation and culture of porcine gastric parietal cells. The results indicate that H. suis affects the viability and function of this cell type.


Subject(s)
Helicobacter Infections/veterinary , Helicobacter heilmannii , Parietal Cells, Gastric/physiology , Swine Diseases/microbiology , Animals , Cells, Cultured , Fluorescent Antibody Technique, Indirect/veterinary , Gastric Acid/metabolism , Helicobacter Infections/pathology , Helicobacter Infections/physiopathology , Parietal Cells, Gastric/pathology , Parietal Cells, Gastric/virology , Polymerase Chain Reaction/veterinary , Swine , Swine Diseases/physiopathology
9.
J Clin Endocrinol Metab ; 99(9): E1691-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24878048

ABSTRACT

BACKGROUND: The incretin effect is reduced in type 2 diabetes mellitus (T2DM) patients. Whether the impaired function of the enteropancreatic axis in these patients is due to defective GLP-1 receptor (GLP-1R) expression in extrapancreatic target organs is not known. AIMS AND METHODS: To compare the GLP-1R expression and distribution in gastric mucosa biopsies of patients with (n =22) and without (n =22) T2DM referred for routine esophagogastroduodenoscopies. GLP-1R mRNA levels were estimated by real-time PCR. The intensity of GLP-1R immunostaining, frequency, and types of glandular cells bearing GLP-1R and their glandular distribution in different stomach mucosa regions were evaluated by immunohistochemical morphological semiquantitative and quantitative analysis. RESULTS: Mean mRNA GLP-1R levels were significantly reduced in patients with T2DM compared with nondiabetic patients (P < .02). Immunohistochemical analysis revealed that the reduced GLP-1R expression in T2DM patients was due to a decreased intensity of immunostaining (P < .01). The number of glandular GLP-1R-bearing cells in both body and antrum mucosa was decreased in T2DM patients. Most notably, the frequency of GLP-1R immunoreactive acid-secreting parietal cells was reduced in the neck area of the gastric principal glands of T2DM patients (P < .01). No correlation was found between the reduced GLP-1R expression and clinical parameters including body mass index, age, glycosylated hemoglobin, and disease duration. CONCLUSION: This is the first evidence of reduced GLP-1R expression in gastric glands of T2DM patients. These data demonstrate that the defective function of the incretin axis in T2DM may also result from decreased GLP-1R expression in its extrapancreatic target organs.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gastric Mucosa/physiology , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Adult , Aged , Biopsy , Endoscopy, Digestive System , Enteroendocrine Cells/cytology , Enteroendocrine Cells/physiology , Female , Gastric Mucosa/cytology , Gene Expression Regulation , Glucagon-Like Peptide-1 Receptor , Humans , Male , Middle Aged , Parietal Cells, Gastric/cytology , Parietal Cells, Gastric/physiology , RNA, Messenger/metabolism
10.
Biochem Biophys Res Commun ; 447(1): 38-43, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24680825

ABSTRACT

Secretion from the gastric gland involves the activation of various types of cells in a coordinated manner. In order to elucidate the mechanisms underlying the coordination of secretion, we studied live fluorescence images of guinea pig gastric glands stained with acridine orange (AO). On 2 µM AO staining, individual cells were characterized by metachromatic colors and various intensities of fluorescence. When the gland was stimulated with 100 µM of histamine, green fluorescence was transiently increased in parietal cells and intermediate cells and propagated along the gland for a long distance over many cells. Local stimulation in a couple of cells with histamine in the presence of suramin also induced propagation. However, the fluorescence response was suppressed by the addition of H-89, a protein kinase A inhibitor. These findings suggest that a cAMP-dependent signal propagates intercellularly through a variety of cells to induce coordinated secretion in the entire gastric gland.


Subject(s)
Cell Communication/physiology , Gastric Mucosa/physiology , Parietal Cells, Gastric/physiology , Signal Transduction , Acridine Orange , Animals , Calcium/physiology , Cell Communication/drug effects , Guinea Pigs , Histamine/pharmacology , Isoquinolines/pharmacology , Male , Microscopy, Fluorescence , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/metabolism , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL