Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 248
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Gastroenterology ; 162(2): 415-430, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34728185

RESUMO

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.


Assuntos
Plasticidade Celular/fisiologia , Reprogramação Celular/fisiologia , Mucosa Gástrica/fisiologia , Regeneração/fisiologia , Estômago/fisiologia , Animais , Mucosa Gástrica/citologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/fisiopatologia , Humanos , Hiperplasia , Metaplasia , Células Parietais Gástricas/fisiologia , Estômago/citologia , Estômago/patologia
2.
Int J Exp Pathol ; 101(6): 230-247, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32985762

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Indometacina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Autofagia/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Mucosa Gástrica/fisiologia , Masculino , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/fisiologia , Ratos , Ratos Sprague-Dawley
3.
Proc Natl Acad Sci U S A ; 114(30): E6260-E6269, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28696284

RESUMO

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.


Assuntos
Cafeína/farmacologia , Ácido Gástrico/metabolismo , Células Parietais Gástricas/fisiologia , Flavonas/farmacologia , Humanos , Células Parietais Gástricas/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Paladar
4.
Microsc Microanal ; 26(4): 846-854, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32458772

RESUMO

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.


Assuntos
Comportamento Animal/fisiologia , Isópteros/fisiologia , Glândulas Salivares/fisiologia , Animais , Células Parietais Gástricas/fisiologia
5.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G36-G42, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29517927

RESUMO

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.


Assuntos
Cálcio , Ativação Enzimática , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Células Parietais Gástricas , Inibidores da Bomba de Prótons/farmacologia , Bombas de Próton , Receptores de Detecção de Cálcio/metabolismo , Animais , Cálcio/sangue , Cálcio/metabolismo , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Ácido Gástrico/metabolismo , Histamina/metabolismo , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/fisiologia , Bombas de Próton/efeitos dos fármacos , Bombas de Próton/metabolismo , Ratos , Ratos Sprague-Dawley , Via Secretória/efeitos dos fármacos , Via Secretória/fisiologia
6.
Gastroenterology ; 152(4): 762-766.e7, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27932312

RESUMO

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.


Assuntos
Apoptose , Celulas Principais Gástricas/patologia , Células Parietais Gástricas/patologia , Células Parietais Gástricas/fisiologia , Estômago/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Atrofia/induzido quimicamente , Azetidinas , Proliferação de Células , Reprogramação Celular , Celulas Principais Gástricas/metabolismo , Toxina Diftérica/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Peptídeos e Proteínas de Sinalização Intercelular , Fator Intrínseco/metabolismo , Metaplasia/induzido quimicamente , Metaplasia/genética , Metaplasia/metabolismo , Camundongos , Células Parietais Gástricas/efeitos dos fármacos , Peptídeos/metabolismo , Piperazinas , Lectinas de Plantas/metabolismo , Tamoxifeno
7.
Vet Res ; 47(1): 101, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27756386

RESUMO

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.


Assuntos
Infecções por Helicobacter/veterinária , Helicobacter heilmannii , Células Parietais Gástricas/fisiologia , Doenças dos Suínos/microbiologia , Animais , Células Cultivadas , Técnica Indireta de Fluorescência para Anticorpo/veterinária , Ácido Gástrico/metabolismo , Infecções por Helicobacter/patologia , Infecções por Helicobacter/fisiopatologia , Células Parietais Gástricas/patologia , Células Parietais Gástricas/virologia , Reação em Cadeia da Polimerase/veterinária , Suínos , Doenças dos Suínos/fisiopatologia
8.
Biochem Biophys Res Commun ; 447(1): 38-43, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24680825

RESUMO

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.


Assuntos
Comunicação Celular/fisiologia , Mucosa Gástrica/fisiologia , Células Parietais Gástricas/fisiologia , Transdução de Sinais , Laranja de Acridina , Animais , Cálcio/fisiologia , Comunicação Celular/efeitos dos fármacos , Cobaias , Histamina/farmacologia , Isoquinolinas/farmacologia , Masculino , Microscopia de Fluorescência , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Sulfonamidas/farmacologia
9.
J Pharmacol Sci ; 121(4): 305-11, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23545479

RESUMO

Atrophic gastritis caused by infection with Helicobacter pylori is characterized by parietal cell loss, which is a main risk factor for gastric cancer. Parietal cells play a crucial role in the regulation of cell lineage maturation and proliferation in the gastric units. Among the classical cadherins, E-cadherin plays an important role not only in epithelial cell-cell connections, but also in the maintenance of epithelial polarity and gastric glandular architecture and regulation of cell proliferation. The aim of this study is to elucidate how parietal cells and E-cadherin are altered in gastritis with Helicobacter pylori infection. We studied the effects of Helicobacter pylori on gastric mucosal E-cadherin 2 weeks after inoculation and investigated the relationship between parietal cell loss and the amount of E-cadherin on parietal cells in Mongolian gerbils. The number of parietal cells and amount of staining of E-cadherin below the isthmus were investigated by immunohistochemistry. It was shown that a reduction in intercellular E-cadherin preceded the disappearance of parietal cells. The gastric glands where parietal cells were lost were replaced by mucus secreting cells without E-cadherin. These results suggest that Helicobacter pylori damaged E-cadherin on parietal cells and caused massive parietal cell loss, leading to the deregulation of gastric morphology.


Assuntos
Caderinas/fisiologia , Gastrite Atrófica/microbiologia , Gastrite Atrófica/patologia , Infecções por Helicobacter , Helicobacter pylori , Células Parietais Gástricas/patologia , Animais , Caderinas/metabolismo , Contagem de Células , Proliferação de Células , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gastrite Atrófica/metabolismo , Gerbillinae , Imuno-Histoquímica , Masculino , Células Parietais Gástricas/fisiologia
10.
Proc Natl Acad Sci U S A ; 107(50): 21511-6, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21115825

RESUMO

Epsin is a ubiquitin-binding endocytic adaptor, which is highly concentrated at clathrin-coated pits and coordinates acquisition of bilayer curvature with coat recruitment and cargo selection. Epsin is encoded by three distinct genes in mammals. Epsin 1 and 2 have broad tissue distribution with high-level expression in the brain. In contrast, epsin 3 was reported to be expressed primarily in immature keratinocytes. Here, we show that epsin 3 is selectively expressed at high levels in the stomach (including the majority of gastric cancers), where it is concentrated in parietal cells. In these cells, epsin 3 is enriched and colocalized with clathrin around apical canaliculi, the sites that control acidification of the stomach lumen via the exo-endocytosis of vesicles containing the H/K ATPase. Deletion of the epsin 3 gene in mice did not result in obvious pathological phenotypes in either the stomach or other organs, possibly because of overlapping functions of the other two epsins. However, levels of EHD1 and EHD2, two membrane tubulating proteins with a role in endocytic recycling, were elevated in epsin 3 knock-out stomachs, pointing to a functional interplay of epsin 3 with EHD proteins in the endocytic pathway of parietal cells. We suggest that epsin 3 cooperates with other bilayer binding proteins with curvature sensing/generating properties in the specialized traffic and membrane remodeling processes typical of gastric parietal cells.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Polaridade Celular , Endocitose/fisiologia , Mucosa Gástrica/metabolismo , Células Parietais Gástricas/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Humanos , Camundongos , Camundongos Knockout , Células Parietais Gástricas/ultraestrutura , Estômago/anatomia & histologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Distribuição Tecidual , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
11.
J Clin Invest ; 118(7): 2459-70, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18535670

RESUMO

Huntingtin interacting protein 1 related (Hip1r) is an F-actin- and clathrin-binding protein involved in vesicular trafficking. In this study, we demonstrate that Hip1r is abundantly expressed in the gastric parietal cell, predominantly localizing with F-actin to canalicular membranes. Hip1r may provide a critical function in vivo, as demonstrated by extensive changes to parietal cells and the gastric epithelium in Hip1r-deficient mice. Electron microscopy revealed abnormal apical canalicular membranes and loss of tubulovesicles in mutant parietal cells, suggesting that Hip1r is necessary for the normal trafficking of these secretory membranes. Accordingly, acid secretory dynamics were altered in mutant parietal cells, with enhanced activation and acid trapping, as measured in isolated gastric glands. At the whole-organ level, gastric acidity was reduced in Hip1r-deficient mice, and the gastric mucosa was grossly transformed, with fewer parietal cells due to enhanced apoptotic cell death and glandular hypertrophy associated with cellular transformation. Hip1r-deficient mice had increased expression of the gastric growth factor gastrin, and mice mutant for both gastrin and Hip1r exhibited normalization of both proliferation and gland height. Taken together, these studies demonstrate that Hip1r plays a significant role in gastric physiology, mucosal architecture, and secretory membrane dynamics in parietal cells.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Células Parietais Gástricas/fisiologia , Vesículas Secretórias/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Celulas Principais Gástricas/metabolismo , Celulas Principais Gástricas/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ácido Gástrico/metabolismo , Determinação da Acidez Gástrica , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gastrinas/sangue , Gastrinas/genética , Expressão Gênica/efeitos dos fármacos , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Histamina/farmacologia , Fator Intrínseco/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Proteínas dos Microfilamentos , Microscopia Eletrônica , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Células Parietais Gástricas/efeitos dos fármacos , Coelhos , Ranitidina/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vesículas Secretórias/ultraestrutura
13.
Gastroenterology ; 139(6): 2018-2027.e2, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20708616

RESUMO

BACKGROUND & AIMS: Gastric stem cells are located in the isthmus of the gastric glands and give rise to epithelial progenitors that undergo bipolar migration and differentiation into pit and oxyntic lineages. Although gastric mucus neck cells located below the isthmus express trefoil factor family 2 (TFF2) protein, TFF2 messenger RNA transcripts are concentrated in cells above the neck region in normal corpus mucosa, suggesting that TFF2 transcription is a marker of gastric progenitor cells. METHODS: Using a BAC strategy, we generated a transgenic mouse with a tamoxifen-inducible Cre under the control of the TFF2 promoter (TFF2-BAC-Cre(ERT2)) and analyzed the lineage derivation from TFF2 mRNA transcript-expressing (TTE) cells. RESULTS: TTE cells were localized to the isthmus, above and distinct from TFF2 protein-expressing mucus neck cells. Lineage tracing revealed that these cells migrated toward the bottom of the gland within 20 days, giving rise to parietal, mucous neck, and chief cells, but not to enterochromaffin-like-cell. Surface mucus cells were not derived from TTE cells and the progeny of the TTE lineage did not survive beyond 200 days. TTE cells were localized in the isthmus adjacent to doublecortin CaM kinase-like-1(+) putative progenitor cells. Induction of spasmolytic polypeptide-expressing metaplasia with DMP-777-induced acute parietal cell loss revealed that this metaplastic phenotype might arise in part through transdifferentiation of chief cells as opposed to expansion of mucus neck or progenitor cells. CONCLUSIONS: TFF2 transcript-expressing cells are progenitors for mucus neck, parietal and zymogenic, but not for pit or enterochromaffin-like cell lineages in the oxyntic gastric mucosa.


Assuntos
Mucosa Gástrica/citologia , Mucosa Gástrica/fisiologia , Mucinas/genética , Proteínas Musculares/genética , Peptídeos/genética , Células-Tronco/citologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Celulas Principais Gástricas/citologia , Celulas Principais Gástricas/fisiologia , Duodeno/citologia , Duodeno/fisiologia , Células Enterocromafins/citologia , Células Enterocromafins/fisiologia , Integrases/genética , Rim/citologia , Rim/fisiologia , Pulmão/citologia , Pulmão/fisiologia , Camundongos , Camundongos Transgênicos , Células Parietais Gástricas/citologia , Células Parietais Gástricas/fisiologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Transcrição Gênica/fisiologia , Fator Trefoil-2
14.
Gastroenterology ; 139(6): 2028-2037.e9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20854822

RESUMO

BACKGROUND & AIMS: Gastric cancer evolves in the setting of a pathologic mucosal milieu characterized by both loss of acid-secreting parietal cells and mucous cell metaplasias. Indeed, mucous cell metaplasia is considered the critical preneoplastic lesion for gastric cancer. Previous investigations have shown that infection of mice with Helicobacter felis or induction of acute parietal cell loss with the drug DMP-777 leads to the emergence of a type of metaplasia designated spasmolytic polypeptide-expressing metaplasia (SPEM). We have hypothesized that SPEM arises from proliferating cells in gland bases, either from a cryptic progenitor cell or by transdifferentiation of mature chief cells. METHODS: Taking advantage of the chief cell-restricted expression of Mist1-Cre-ER(T2), we used lineage mapping to examine whether SPEM lineages were derived from chief cells in 3 independent models of induction by DMP-777 treatment, L-635 treatment, or H felis infection. RESULTS: Treatment of mice with L-635 for 3 days led to rapid parietal cell loss, induction of a prominent inflammatory infiltrate, and emergence of SPEM. In all 3 models, SPEM developed, at least in part, from transdifferentiation of chief cells. We further found that acute parietal cell loss in the setting of inflammation (L-635 treatment) led to more rapid induction and expansion of SPEM derived from transdifferentiation of chief cells. CONCLUSIONS: These studies provide direct evidence by lineage tracing that SPEM evolves from differentiated chief cells. Thus, mature gastric chief cells have the ability to act as cryptic progenitors and reacquire proliferative capacity within the context of mucosal injury and inflammation.


Assuntos
Celulas Principais Gástricas/patologia , Gastrite/patologia , Lesões Pré-Cancerosas/patologia , Células-Tronco/patologia , Neoplasias Gástricas/patologia , Doença Aguda , Animais , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Linhagem da Célula/fisiologia , Celulas Principais Gástricas/fisiologia , Doença Crônica , Modelos Animais de Doenças , Gastrite/microbiologia , Gastrite/fisiopatologia , Infecções por Helicobacter/patologia , Infecções por Helicobacter/fisiopatologia , Helicobacter felis , Peptídeos e Proteínas de Sinalização Intercelular , Óperon Lac/genética , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Parietais Gástricas/patologia , Células Parietais Gástricas/fisiologia , Peptídeos/genética , Peptídeos/metabolismo , Lesões Pré-Cancerosas/microbiologia , Lesões Pré-Cancerosas/fisiopatologia , Células-Tronco/fisiologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/fisiopatologia
15.
Cell Physiol Biochem ; 27(5): 597-604, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21691077

RESUMO

The Ca(2+) activated K(+) channel K(ca)3.1 is expressed in a variety of tissues. In the gastric gland it is expressed in the basolateral cell membrane. To determine the functional significance of K(ca)3.1 activity for gastric acid secretion, gastric acid secretion was determined in isolated glands from gene targeted mice lacking functional K(ca)3.1 (K(ca)3.1(-/-)) and from their wild type littermates (K(ca)3.1(+/+)). According to BCECF-fluorescence cytosolic pH in isolated gastric glands was similar in K(ca)3.1(-/-) and K(ca)3.1(+/+) mice. Na(ca)-independent pH recovery (ΔpH/min) following an ammonium pulse, a measure of H(ca)/K(ca) ATPase activity, was, however, significantly faster in K(ca)3.1(-/-) than in K(ca)3.1(+/+) mice. Accordingly, the luminal pH was significantly lower and the acid content significantly higher in K(ca)3.1(-/-) than in K(ca)3.1(+/+) mice. The abundance of mRNA encoding H(ca)/K(ca) ATPase and KCNQ1 was similar in both genotypes. Increase of extracellular K(ca) concentrations to 35 mM (replacing Na(ca)/NMDG) and treatment with histamine (100 µM) significantly increased ΔpH/min to a larger extent in K(ca)3.1(+/+) than in K(ca)3.1(-/-) mice and dissipated the differences between the genotypes. Carbachol (100 µM) increased ΔpH/min in both genotypes but did not abolish the difference between K(ca)3.1(-/-) and K(ca)3.1(+/+) mice. In K(ca)3.1(+/+) mice the K(ca)3.1 opener DCEBIO (100 µM) did not significantly alter basal ΔpH/min but significantly blunted ΔpH/min in the presence of carbachol. In conclusion, K(ca)3.1 activity suppresses carbachol stimulated gastric acid secretion.


Assuntos
Ácido Gástrico/enzimologia , Mucosa Gástrica/fisiologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Canal de Potássio KCNQ1/metabolismo , Células Parietais Gástricas/fisiologia , Amônia/farmacologia , Animais , Transporte Biológico , Cálcio/metabolismo , Carbacol/farmacologia , Fluoresceínas/análise , Ácido Gástrico/metabolismo , Determinação da Acidez Gástrica , Mucosa Gástrica/efeitos dos fármacos , ATPase Trocadora de Hidrogênio-Potássio/genética , Histamina/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Canal de Potássio KCNQ1/genética , Camundongos , Camundongos Knockout , Omeprazol/farmacologia , Técnicas de Cultura de Órgãos , Células Parietais Gástricas/efeitos dos fármacos , Potássio/metabolismo , Prótons , RNA Mensageiro/análise
16.
Scand J Gastroenterol ; 46(5): 531-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21342027

RESUMO

BACKGROUND AND AIMS: Enterochromaffin-like (ECL) cells are central in the regulation of acid secretion. G cells release gastrin and activate ECL cell histamine secretion which stimulates parietal cell H(2) receptors initiating acid secretion. It is unclear whether histamine-mediated parietal cell activation is via a vascular or paracrine pathway. To assess this, we utilized immunohistochemistry (IHC) and electron microscopy to examine gastric tissue and used visualization of formalin fixed dispersed gastric cells and glands to investigate and define the anatomical relationship between ECL and parietal cells. MATERIAL AND METHODS: Sprague-Dawley rat stomachs were instilled with formalin. Thereafter fixed mucosal cells and whole gastric glands were dispersed by mechanical and chemical dissolution and enzymatic digestion. Smears with fixed isolated cells and whole glands were stained by IHC with histidine decarboxylase (HDC) and H+/K+-ATPase antibodies. Whole tissue samples of Sprague-Dawley and cotton rat oxyntic mucosa were investigated with IHC using HDC, VMAT2 and H+/K+-ATPase antibodies, and electron microscopy was performed to further delineate the precise anatomic relationship between ECL cells and parietal cells. RESULTS: Each ECL cell generated a network of HDC- and VMAT2-positive dendritic-like elongations that were in direct contact with several parietal cells. Thus, ECL cells at the base of the gland were in communication with parietal cells in the middle of the gland. Electron microscopy confirmed that the cytoplasmic ECL cell elongations containing secretory vesicles were in direct juxtaposition to parietal cells. CONCLUSIONS: These findings indicate that ECL cells directly regulate parietal cell function in a neurocrine manner via slender neuron-like elongations.


Assuntos
Celulas Tipo Enterocromafim/citologia , Celulas Tipo Enterocromafim/metabolismo , Ácido Gástrico/metabolismo , Mucosa Gástrica/anatomia & histologia , Células Parietais Gástricas/citologia , Células Parietais Gástricas/metabolismo , Animais , Comunicação Celular/fisiologia , Celulas Tipo Enterocromafim/fisiologia , Feminino , Mucosa Gástrica/metabolismo , Liberação de Histamina/fisiologia , Junções Intercelulares/fisiologia , Células Parietais Gástricas/fisiologia , Ratos , Ratos Sprague-Dawley , Sigmodontinae
17.
Am J Physiol Cell Physiol ; 298(1): C1-C10, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19889964

RESUMO

The parietal cell is responsible for secreting concentrated hydrochloric acid into the gastric lumen. To fulfill this task, it is equipped with a broad variety of functionally coupled apical and basolateral ion transport proteins. The concerted scientific effort over the last years by a variety of researchers has provided us with the molecular identity of many of these transport mechanisms, thereby contributing to the clarification of persistent controversies in the field. This article will briefly review the current model of parietal cell physiology and ion transport in particular and will update the existing models of apical and basolateral transport in the parietal cell.


Assuntos
Células Parietais Gástricas/fisiologia , Estômago/fisiologia , Adenilato Quinase/metabolismo , Antiporters/genética , Transporte Biológico , Proteínas de Transporte de Cátions/metabolismo , Digestão , Ácido Gástrico/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Íons/metabolismo , Modelos Biológicos , Células Parietais Gástricas/enzimologia , Transportadores de Sulfato
18.
Am J Physiol Cell Physiol ; 299(2): C431-43, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20505040

RESUMO

Ezrin is an important membrane/actin cytoskeleton linker protein, especially in epithelia. Ezrin has two important binding domains: an NH(2)-terminal region that binds to plasma membrane and a COOH-terminal region that binds to F-actin only after a conformational activation by phosphorylation at Thr567 of ezrin. The present experiments were undertaken to investigate the detailed cellular changes in the time course of expression of ezrin-T567 mutants (nonphosphorylatable T567A and permanent phospho-mimic T567D) in parietal cells and to assess ezrin distribution and its influence on the elaborate membrane recruitment processes of these cells. T567A mutant and wild-type (WT) ezrin were consistently localized to the apical plasma membrane, even with overexpression. On the other hand, T567D went first to apical membrane at early times and low expression levels, then accumulated mainly at the basal surface after 24 h. Overexpression of WT or T567A led to incorporation of internal membranes to apical vacuoles, while overexpression of T567D led to large incorporation of apical and intracellular membranes (including H-K-ATPase) to the basal surface. Differences in polar distribution of ezrin suggest a role for the linker protein in promoting formation and plasticity of membrane surface projections, forming the basis for a novel theory for ezrin as an organizer and regulator of membrane recruitment. A model simulating the cellular distribution of ezrin and its associated membrane- and F-actin-binding forms is given to predict redistributions observed with phosphorylation and mutant overexpression, and it can easily be modified as more specific information regarding binding constants and specific sites becomes available.


Assuntos
Comunicação Celular/fisiologia , Polaridade Celular/fisiologia , Proteínas do Citoesqueleto/fisiologia , Actinas/metabolismo , Actinas/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/fisiologia , Humanos , Células Parietais Gástricas/citologia , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/fisiologia , Coelhos , Fatores de Tempo
19.
J Biomed Biotechnol ; 2010: 394198, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20339514

RESUMO

Acid secretion in gastric parietal cells requires highly coordinated membrane transport and vesicle trafficking. Histologically, consensus defines acid secretion as the ratio of the volume density (Vd) of canalicular and apical membranes (CAMs) to tubulovesicular (TV) membranes, a value which varies widely under normal conditions. Examination of numerous achlorhydric mice made it clear that this paradigm is discrepant when used to assess most mice with genetic mutations affecting acid secretion. Vd of organelles in parietal cells of 6 genetically engineered mouse strains was obtained to identify a stable histological phenotype of acid secretion. We confirmed that CAM to TV ratio fairly represented secretory activity in untreated and secretion-inhibited wild-type (WT) mice and in NHE2-/- mice as well, though the response was significantly attenuated in the latter. However, high CAM to TV ratios wrongly posed as active acid secretion in AE2-/-, GHKAalpha-/-, and NHE4-/- mice. Achlorhydric genotypes also had a significantly higher Vd of basolateral membrane than WT mice, and reduced Vd of mitochondria and canaliculi. The Vd of mitochondria, and ratio of the Vd of basolateral membranes/Vd of mitochondria were preferred predictors of the level of acid secretion. Alterations in acid secretion, then, cause significant changes not only in the Vd of secretory membranes but also in mitochondria and basolateral membranes.


Assuntos
Membrana Basal/ultraestrutura , Mitocôndrias/fisiologia , Células Parietais Gástricas/fisiologia , Animais , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Antiporters/genética , Antiporters/metabolismo , Membrana Celular , Tamanho Celular , Ácido Gástrico/metabolismo , Corpos de Inclusão/metabolismo , Corpos de Inclusão/ultraestrutura , Camundongos , Camundongos Transgênicos , Mitocôndrias/ultraestrutura , Células Parietais Gástricas/citologia , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/ultraestrutura , Proteínas SLC4A , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo
20.
Mol Cell Biochem ; 341(1-2): 43-50, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20237950

RESUMO

Gastric pathology is a common complication in diabetes mellitus. The aim of the study was to evaluate the functions and morphological changes of the parietal cells of the rat stomach after streptozotocin-induced diabetes. Diabetes mellitus was induced in Wistar rats by a single intraperitoneal injection of streptozotocin (60 mg/kg body weight). The rats were weighed weekly and sacrificed after 6 months. The glandular portion of the stomach was removed and processed for H(+)-K(+)-ATPase immunohistochemistry and light and electron microscopy studies. Acid secretion was measured in vivo. After 6 months of diabetes, the mean weight of the rats was significantly lower (P < 0.001) compared to control. The mean weight of the stomach to body weight percentage increased significantly (P < 0.001) compared to control. The blood glucose level in diabetic rats was significantly higher (P < 0.001) than in normal control. Diabetic rats showed significant (P < 0.001) decrease in basal and stimulated acid secretion when compared to control. Electron micrographs of the parietal cells of glandular stomach of diabetic rats revealed significant (P < 0.0002) reduction in the number of mitochondria and a small though not significant increase in the number of canaliculi in the parietal cells compared with normal. Immunohistochemistry showed reduced H(+)-K(+)-ATPase (P < 0.00001) compared to control. Long-term diabetes induces morphological as well as functional changes in gastric parietal cells. The decrease in the number of mitochondria accompanied by reduced in H(+)-K(+)-ATPase in parietal cells may explain the reduced acid secretion observed in diabetics.


Assuntos
Diabetes Mellitus Experimental/patologia , Células Parietais Gástricas/patologia , Animais , Peso Corporal , Complicações do Diabetes/patologia , Ácido Gástrico/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/análise , Mitocôndrias , Tamanho do Órgão , Células Parietais Gástricas/fisiologia , Ratos , Ratos Wistar , Estômago , Estreptozocina , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA