Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Clin J Gastroenterol ; 17(4): 607-616, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38635098

RESUMEN

A 47-year-old woman presented with multiple gastric tumors, each up to 10 mm in diameter, in the gastric body and fundus without mucosal atrophy. White spots and numerous transparent, light-brownish, small, and rounded spots were observed in the background gastric mucosa. Biopsy specimens obtained from the tumors revealed gastric neuroendocrine tumors. The patient exhibited hypergastrinemia and achlorhydria and tested negative for serum parietal cell antibody, intrinsic factor antibody, and Helicobacter pylori infection. Moreover, no additional lesions were detected on imaging. These findings were inconsistent with Rindi's classification. The tumor was resected via endoscopic submucosal resection. Histopathological examination revealed gastric neuroendocrine tumors G2 infiltrating the submucosa with no atrophy of the gastric mucosa, dilated fundic glands, parietal cell protrusions, and hyperplasia of enterochromaffin-like cells. Immunohistochemically, the parietal cells were negative for both α- and ß-subunits of H+/K+ ATPase, suggesting parietal cell dysfunction. A genomic variant was identified in adenosine triphosphatase H+/K+ transporting subunit alpha. After 7 years of treatment, there was no evidence of residual or metastatic lesions. Modification of adenosine triphosphatase H+/K+ transporting subunit alpha may be a significant factor in the pathogenesis of multiple gastric neuroendocrine tumors in the context of gastric parietal cell dysfunction.


Asunto(s)
Tumores Neuroendocrinos , Células Parietales Gástricas , Neoplasias Gástricas , Humanos , Femenino , Persona de Mediana Edad , Células Parietales Gástricas/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Aclorhidria/genética , Neoplasias Primarias Múltiples/genética , Neoplasias Primarias Múltiples/patología , Mucosa Gástrica/patología , Resección Endoscópica de la Mucosa
2.
FASEB J ; 34(8): 10699-10719, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32584506

RESUMEN

Heart failure (HF) is an increasing global health crisis, affecting 40 million people and causing 50% mortality within 5 years of diagnosis. A fuller understanding of the genetic and environmental factors underlying HF, and novel therapeutic approaches to address it, are urgently warranted. Here, we discovered that cardiac-specific germline deletion in mice of potassium channel ß subunit-encoding Kcne2 (Kcne2CS-/- ) causes dilated cardiomyopathy and terminal HF (median longevity, 28 weeks). Mice with global Kcne2 deletion (Kcne2Glo-/- ) exhibit multiple HF risk factors, yet, paradoxically survived over twice as long as Kcne2CS-/- mice. Global Kcne2 deletion, which inhibits gastric acid secretion, reduced the relative abundance of species within Bacteroidales, a bacterial order that positively correlates with increased lifetime risk of human cardiovascular disease. Strikingly, the proton-pump inhibitor omeprazole similarly altered the microbiome and delayed terminal HF in Kcne2CS-/- mice, increasing survival 10-fold at 44 weeks. Thus, genetic or pharmacologic induction of hypochlorhydria and decreased gut Bacteroidales species are associated with lifespan extension in a novel HF model.


Asunto(s)
Aclorhidria/genética , Aclorhidria/mortalidad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/mortalidad , Canales de Potasio con Entrada de Voltaje/genética , Animales , Bacteroides/crecimiento & desarrollo , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/mortalidad , Femenino , Ácido Gástrico/metabolismo , Microbioma Gastrointestinal/genética , Eliminación de Gen , Corazón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Riesgo
3.
Biosci Rep ; 40(2)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31904088

RESUMEN

Parietal cells of the gastric mucosa contain a complex and extensive secretory membrane system that harbors gastric H+, K+-adenosine triphosphatase (ATPase), the enzyme primarily responsible for gastric lumen acidification. Here, we describe the characterization of mice deficient in the H+, K+-ATPase α subunit (Atp4a-/-) to determine the role of this protein in the biosynthesis of this membrane system and the biology of the gastric mucosa. Atp4a-/- mice were produced by gene targeting. Wild-type (WT) and Atp4a-/- mice, paired for age, were examined at 10, 12, 14 and 16 weeks for histopathology, and the expression of mucin 2 (MUC2), α-methylacyl-CoA racemase (AMACR), Ki-67 and p53 proteins was analyzed by immunohistochemistry. For further information, phosphoinositide 3-kinase (PI3K), phosphorylated-protein kinase B (p-AKT), mechanistic target of rapamycin (mTOR), hypoxia-inducible factor 1α (HIF-1α), lactate dehydrogenase A (LDHA) and sirtuin 6 (SIRT6) were detected by Western blotting. Compared with the WT mice, hypochlorhydric Atp4a-/- mice developed parietal cell atrophy and significant antral inflammation (lymphocyte infiltration) and intestinal metaplasia (IM) with elevated MUC2 expression. Areas of dysplasia in the Atp4a-/- mouse stomach showed increased AMACR and Ki-67 expression. Consistent with elevated antral proliferation, tissue isolated from Atp4a-/- mice showed elevated p53 expression. Next, we examined the mechanism by which the deficiency of the H+, K+-ATPase α subunit has an effect on the gastric mucosa. We found that the expression of phosphorylated-PI3K, p-AKT, phosphorylated-mTOR, HIF-1α, LDHA and SIRT6 was significantly higher in tissue from the Atp4a-/- mice compared with the WT mice (P<0.05). The H+, K+-ATPase α subunit is required for acid-secretory activity of parietal cells in vivo, the normal development and cellular homeostasis of the gastric mucosa, and attainment of the normal structure of the secretory membranes. Chronic achlorhydria and hypergastrinemia in aged Atp4a-/- mice produced progressive hyperplasia and mucolytic and IM, and activated the Warburg effect via PI3K/AKT/mTOR signaling.


Asunto(s)
Aclorhidria/enzimología , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , Células Parietales Gástricas/enzimología , Lesiones Precancerosas/enzimología , Neoplasias Gástricas/enzimología , Aclorhidria/genética , Aclorhidria/patología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Enfermedad Crónica , Metabolismo Energético , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Metaplasia , Ratones Endogámicos C57BL , Ratones Noqueados , Mucina 2/metabolismo , Células Parietales Gástricas/patología , Fosfatidilinositol 3-Quinasa/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo
4.
Gastric Cancer ; 23(1): 52-63, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31250150

RESUMEN

BACKGROUND: Type I gastric neuroendocrine tumors (gNETs) arise from hypergastrinemia in patients with autoimmune chronic atrophic gastritis. According to the classical model, the gastric H+/K+ ATPase was the causative autoantigen recognized by CD4+ T cells in chronic autoimmune scenario that secretes IL-17 and correlates with parietal cell (PC) atrophy, which drives to gastric achlorhydria and increases the risk for gastric neoplasms. However, the mechanism by which the inflammatory response correlates with PC atrophy is not clearly defined. METHODS: Recently, we found that the ATP4Ap.R703C mutation impaired PC function and gastric acidification, which drove familial gNET. Our group constructed a knock-in mouse model for the ATP4A mutation, which has served us to better understand the relation between impaired capability to export protons across the plasma membrane of PCs and tumor progression. RESULTS: The ATP4Ap.R703C mutation drives gastric achlorhydria, but also deregulates the acid-base balance within PCs, affecting mitochondrial biogenesis. Mitochondrial malfunction activates ROS signaling, which triggers caspase-3-mediated apoptosis of parietal cells. In addition, when gastric euchlorhydria was restored, mitochondrial function is recovered. Infection by H. pylori promotes destabilization of the mitochondria of the PCs by a mechanism similar to that described for APT4Ap.R703C carriers. CONCLUSIONS: A genetic origin that drives mitochondria alteration would initiate the gastric chronic inflammation instead of the classical IL-17 secretion-mediated mechanism explanation. Gastric euchlorhydria restoration is suggested to be indicated for mitochondrial recover. Our results open a new window to understand gastric neoplasms formation but also the inflammatory mechanisms and autoimmune disorders conducted by genetic origin that composes a premalignant scenario.


Asunto(s)
Equilibrio Ácido-Base/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Tumores Neuroendocrinos/inmunología , Neoplasias Gástricas/inmunología , Aclorhidria/genética , Animales , Apoptosis/fisiología , Autoinmunidad/genética , Técnicas de Sustitución del Gen , Infecciones por Helicobacter/patología , Humanos , Ratones Mutantes , Mitocondrias/patología , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Estrés Oxidativo , Células Parietales Gástricas/inmunología , Células Parietales Gástricas/patología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
5.
Tsitol Genet ; 50(3): 57-65, 2016.
Artículo en Inglés, Ruso | MEDLINE | ID: mdl-30480410

RESUMEN

Expression of Tlr2, Muc2, Defa genes in rat duodenal upon long-term gastric hypacidity (hypochlorhydria) and with multiprobiotic correction was investigated. Increasing of Tlr2, Muc2, Defa expression on the background of the intensification of lipid peroxidation in rat duodenal villus and crypt epithelial cells upon gastric hypoacidity was shown. The mRNAs patterns of genes mentioned above were shown to decrease or approach to control values as well as the level of lipid peroxidation products under the treatment of hypoacidic rats with the multiprobiotic. The data obtained may indicate involvement of genes Tlr2, Muc2, Defa in development of inflammation in the duodenum due to dysbiotic changes under conditions of prolonged hypochlorhydria.


Asunto(s)
Aclorhidria/terapia , Disbiosis/terapia , Mucina 2/genética , Probióticos/farmacología , Receptor Toll-Like 2/genética , alfa-Defensinas/genética , Aclorhidria/inducido químicamente , Aclorhidria/genética , Aclorhidria/microbiología , Animales , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Duodeno/microbiología , Disbiosis/inducido químicamente , Disbiosis/genética , Disbiosis/microbiología , Regulación de la Expresión Génica , Concentración de Iones de Hidrógeno , Masculino , Mucina 2/metabolismo , Omeprazol/administración & dosificación , Inhibidores de la Bomba de Protones/administración & dosificación , Ratas , Estómago/efectos de los fármacos , Estómago/microbiología , Receptor Toll-Like 2/metabolismo , alfa-Defensinas/metabolismo
6.
PLoS One ; 8(8): e72735, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24009701

RESUMEN

BACKGROUND/AIMS: H. pylori CagL amino acid polymorphisms such as Y58/E59 can increase integrin α5ß1 expression and gastric cancer risk. Hypochlorhydria during chronic H. pylori infection promotes gastric carcinogenesis. The study test whether CagL-Y58/E59 isolates may regulate integrin α5ß1 to translocate CagA via the type IV secretory system even under adverse pH conditions, and whether the integrin α5ß1 expression primed by H. pylori is a pH-dependent process involving hypochlorhydria in a vicious cycle to promote gastric carcinogenesis. METHODS: The expressions of integrin α5 and ß1, CagA phosphorylation, IL-8, FAK, EGFR, and AKT activation of AGS cells exposed to CagL-Y58/E59 H. pylori, isogenic mutants, and different H. pylori CagL amino acid replacement mutants under different pH values were determined. Differences in the pepsinogen I/II ratio (indirectly indicating gastric acidity) and gastric integrin α5ß1 expression were compared among the 172 H. pylori-infected patients with different cancer risks. RESULTS: Even under adversely low pH condition, H. pylori CagL-Y58/E59 still keep active integrin ß1 with stronger binding affinity, CagA translocation, IL-8, FAK, EGFR, and AKT activation than the other mutants (p<0.05). The in vitro assay revealed higher priming of integrin α5ß1 by H. pylori under elevated pH as hypochlorhydria (p<0.05). In the H. pylori-infected patients, the gastric integrin α5ß1 expressions were higher in those with pepsinogen I/II ratio <6 than in those without (p<0.05). CONCLUSIONS: H. pylori CagL-Y58/E59 prime higher integrin under adverse pH and may involve to enhance hypochlorhydria vicious cycle for gastric carcinogenesis, and thus require an early eradication.


Asunto(s)
Aclorhidria/metabolismo , Proteínas Bacterianas/metabolismo , Transformación Celular Neoplásica/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Integrina alfa5beta1/metabolismo , Neoplasias Gástricas/etiología , Aclorhidria/genética , Adulto , Anciano , Proteínas Bacterianas/genética , Transformación Celular Neoplásica/genética , Receptores ErbB/metabolismo , Femenino , Helicobacter pylori/genética , Humanos , Concentración de Iones de Hidrógeno , Integrina alfa5beta1/genética , Interleucina-8/biosíntesis , Masculino , Persona de Mediana Edad , Modelos Biológicos , Mutación , Pepsinógeno A/metabolismo , Pepsinógeno C/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
7.
Am J Physiol Gastrointest Liver Physiol ; 302(1): G105-15, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21921286

RESUMEN

SAMP1/YitFcs mice serve as a model of Crohn's disease, and we have used them to assess gastritis. Gastritis was compared in SAMP1/YitFcs, AKR, and C57BL/6 mice by histology, immunohistochemistry, and flow cytometry. Gastric acid secretion was measured in ligated stomachs, while anti-parietal cell antibodies were assayed by immunofluorescence and enzyme-linked immunosorbent spot assay. SAMP1/YitFcs mice display a corpus-dominant, chronic gastritis with multifocal aggregates of mononuclear cells consisting of T and B lymphocytes. Relatively few aggregates were observed elsewhere in the stomach. The infiltrates in the oxyntic mucosa were associated with the loss of parietal cell mass. AKR mice, the founder strain of the SAMP1/YitFcs, also have gastritis, although they do not develop ileitis. Genetic studies using SAMP1/YitFcs-C57BL/6 congenic mice showed that the genetic regions regulating ileitis had comparable effects on gastritis. The majority of the cells in the aggregates expressed the T cell marker CD3 or the B cell marker B220. Adoptive transfer of SAMP1/YitFcs CD4(+) T helper cells, with or without B cells, into immunodeficient recipients induced a pangastritis and duodenitis. SAMP1/YitFcs and AKR mice manifest hypochlorhydria and anti-parietal cell antibodies. These data suggest that common genetic factors controlling gastroenteric disease in SAMP1/YitFcs mice regulate distinct pathogenic mechanisms causing inflammation in separate sites within the digestive tract.


Asunto(s)
Aclorhidria/inmunología , Enfermedades Autoinmunes/inmunología , Gastritis/inmunología , Ileítis/inmunología , Aclorhidria/genética , Aclorhidria/patología , Traslado Adoptivo , Animales , Autoanticuerpos/análisis , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Linfocitos B/inmunología , Linfocitos B/patología , Complejo CD3/análisis , Complejo CD3/inmunología , Femenino , Ácido Gástrico/metabolismo , Gastritis/genética , Gastritis/patología , Ileítis/genética , Ileítis/patología , Antígenos Comunes de Leucocito/análisis , Antígenos Comunes de Leucocito/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Linfocitos T/patología , Linfocitos T Colaboradores-Inductores/inmunología
8.
Gastroenterology ; 140(3): 857-67, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21111738

RESUMEN

BACKGROUND & AIMS: Mutations in TRPML1, a lysosomal Ca(2+)-permeable TRP channel, lead to mucolipidosis type IV, a neurodegenerative lysosomal storage disease. An unusual feature of mucolipidosis type IV is constitutive achlorhydria. We produced Trpml1(-/-) (null) mice to investigate the requirement for this protein in gastric acid secretion. METHODS: Trpml1-null mice were generated by gene targeting. The expression of Trpml1 and its role in acid secretion by gastric parietal cells were analyzed using biochemical, histologic, and ultrastructural approaches. RESULTS: Trpml1 is expressed by parietal cells and localizes predominantly to the lysosomes; it was dynamically palmitoylated and dephosphorylated in vivo following histamine stimulation of acid secretion. Trpml1-null mice had significant impairments in basal and histamine-stimulated gastric acid secretion and markedly reduced levels of the gastric proton pump. Histologic and ultrastructural analyses revealed that Trpml1(-/-) parietal cells were enlarged, had multivesicular and multi-lamellated lysosomes, and maintained an abnormal intracellular canalicular membrane. The intralysosomal Ca(2+) content and receptor-mediated Ca(2+) signaling were, however, unaffected in Trpml1(-/-) gastric glands, indicating that Trpml1 does not function in the regulation of lysosomal Ca(2+). CONCLUSIONS: Loss of Trpml1 causes reduced levels and mislocalization of the gastric proton pump and alters the secretory canaliculi, causing hypochlorhydria and hypergastrinemia. The lysosomal enlargement and defective intracellular canaliculi formation observed in Trpml1(-/-) parietal cells indicate that Trpml1 functions in the formation and trafficking of the tubulovesicles. This study provides direct evidence for the regulation of gastric acid secretion by a TRP channel; TRPML1 is an important protein in parietal cell apical membrane trafficking.


Asunto(s)
Ácido Gástrico/metabolismo , Mucolipidosis/metabolismo , Células Parietales Gástricas/metabolismo , Canales Catiónicos TRPM/deficiencia , Aclorhidria/genética , Aclorhidria/metabolismo , Aclorhidria/fisiopatología , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Gastrinas/sangre , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Histamina/metabolismo , Hipertrofia , Lipoilación , Lisosomas/metabolismo , Lisosomas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucolipidosis/genética , Mucolipidosis/patología , Mucolipidosis/fisiopatología , Células Parietales Gástricas/ultraestructura , Fosforilación , Transporte de Proteínas , Canales Catiónicos TRPM/genética , Factores de Tiempo , Canales de Potencial de Receptor Transitorio
9.
Clin Cancer Res ; 14(7): 2227-35, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381965

RESUMEN

PURPOSE: Helicobacter pylori infection by virulent strains is associated with gastric adenocarcinoma. We aimed to determine whether infection with virulent H. pylori preceded precancerous gastric hypochlorhydria and atrophy in gastric cancer relatives and quantify the extent of virulence factor evolution. EXPERIMENTAL DESIGN: H. pylori strains from 51 Scottish gastric cancer relatives were characterized by genetic fingerprinting and typing the vacuolating cytotoxin gene (vacA), the cytotoxin-associated gene (cagA), and housekeeping genes. We phenotyped strains by coculture with gastric epithelial cells and assessing vacuolation (microscopy), CagA tyrosine phosphorylation (immunoblot), and interleukin-8 secretion (ELISA). RESULTS: Toxigenic (vacA type s1/m1) H. pylori was associated with precancerous gastric hypochlorhydria (P<0.01). Adult family members with this type of H. pylori had the same strain as currently noncohabiting adult family members in 68% cases, implying acquisition during childhood from each other or a common source. We analyzed different isolates of the same strain within families and showed that H. pylori commonly microevolved to change virulence: this occurred in 22% individuals and a striking 44% cases where the strain was shared within families. Microevolution in vacA occurred by extragenomic recombination and in cagA by this or duplication/deletion. Microevolution led to phenotypic changes in virulence. Passage of microevolved strains could be tracked within families. CONCLUSIONS: Toxigenic H. pylori infection precedes and so likely causes gastric hypochlorhydria, suggesting that virulent H. pylori increases cancer risk by causing this condition. Microevolution of virulence genes is common within families of gastric cancer patients and changes H. pylori virulence.


Asunto(s)
Aclorhidria/virología , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/patogenicidad , Lesiones Precancerosas/virología , Neoplasias Gástricas/virología , Aclorhidria/genética , Adulto , Anciano , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Dermatoglifia del ADN , Familia , Femenino , Helicobacter pylori/genética , Humanos , Masculino , Persona de Mediana Edad , Linaje , Lesiones Precancerosas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/genética , Virulencia
10.
Exp Anim ; 56(4): 295-300, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17660684

RESUMEN

The KCNQ1 gene encodes a voltage-dependent potassium ion channel, and mutations in this gene are the most common cause of congenital long QT syndrome (LQTS). In the present study, we investigated the various phenotypic characteristics of vertigo 2 Jackson (C3H/HeJCrl-Kcnq1(vtg-2J)/J) mice with a Kcnq1 mutation. Both heterozygotes (vtg-2J/+) and homozygotes (vtg-2J/vtg-2J) showed prolonged QT intervals in electrocardiograms (ECGs) compared to C3H/HeJ control (+/+) mice. Furthermore, vtg-2J/vtg-2J mice showed gastric achlorhydria associated with elevation of their serum gastrin levels. The serum corticosterone levels were also significantly increased in vtg-2J/vtg-2J mice. In addition, vtg-2J/vtg-2J mice exhibited significantly higher blood pressure. These findings indicate that the Kcnq1 mutation in vtg-2J mice alters various physiological functions in the cardiac, gastric and adrenocortical systems, and suggest that vtg-2J mice may represent a useful model for studying Kcnq1 functions.


Asunto(s)
Aclorhidria/genética , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Aclorhidria/sangre , Aclorhidria/patología , Animales , Corticosterona/sangre , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Ácido Gástrico/metabolismo , Determinación de la Acidez Gástrica , Mucosa Gástrica/metabolismo , Gastrinas/sangre , Genotipo , Hipertensión/genética , Hipertensión/fisiopatología , Síndrome de QT Prolongado/patología , Síndrome de QT Prolongado/fisiopatología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Mutantes , Mutación , Fenotipo , Estómago/patología
11.
Gastroenterology ; 132(3): 905-12, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17324405

RESUMEN

BACKGROUND AND AIMS: TLR4 is a cell-surface signaling receptor involved in the recognition and host response to Helicobacter pylori. The TLR4+896A>G polymorphism linked with impaired reactivity to bacterial lipopolysaccharide may play a role in gastric carcinogenesis. METHODS: We assessed associations with premalignant gastric changes in 149 relatives of gastric cancer patients, including 45 with hypochlorhydria and gastric atrophy. We also genotyped 2 independent Caucasian population-based case-control studies of upper gastrointestinal tract cancer, initially in 312 noncardia gastric carcinoma cases and 419 controls and then in 184 noncardia gastric carcinomas, 123 cardia carcinomas, 159 esophageal cancers, and 211 frequency-matched controls. Odds ratios were computed from logistic models and adjusted for potential confounding factors. RESULTS: TLR4+896G carriers had an 11-fold (95% confidence interval [CI], 2.5-48) increased odds ratio (OR) for hypochlorhydria; the polymorphism was unassociated with gastric acid output in the absence of H pylori infection. Carriers also had significantly more severe gastric atrophy and inflammation. Seventeen percent of gastric carcinoma patients in the initial study and 15% of the noncardia gastric carcinoma patients in the replication study had 1 or 2 TLR4 variant alleles vs 8% of both control populations (combined OR = 2.3; 95% CI = 1.6-3.4). In contrast, prevalence of TLR4+896G was not significantly increased in esophageal squamous cell (2%, OR = 0.2) or adenocarcinoma (9%, OR = 1.4) or gastric cardia carcinoma (11%, OR = 1.4). CONCLUSIONS: Our data suggest that the TLR4+896A>G polymorphism is a risk factor for noncardia gastric carcinoma and its precursors. The findings underscore the role of the host innate immune response in outcome of H pylori infection.


Asunto(s)
Carcinoma/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori , Polimorfismo Genético , Lesiones Precancerosas/genética , Neoplasias Gástricas/genética , Receptor Toll-Like 4/genética , Aclorhidria/genética , Aclorhidria/microbiología , Carcinoma/microbiología , Carcinoma/patología , Estudios de Casos y Controles , Estudios de Cohortes , Europa (Continente) , Femenino , Gastritis Atrófica/genética , Gastritis Atrófica/microbiología , Regulación Neoplásica de la Expresión Génica , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Humanos , Modelos Logísticos , Masculino , Oportunidad Relativa , Fenotipo , Vigilancia de la Población , Lesiones Precancerosas/microbiología , Lesiones Precancerosas/patología , Sistema de Registros , Medición de Riesgo , Factores de Riesgo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Estados Unidos
12.
J Biol Chem ; 281(33): 23740-7, 2006 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-16754665

RESUMEN

Genes in the KCNE family encode single transmembrane domain ancillary subunits that co-assemble with voltage-gated potassium (Kv) channel alpha subunits to alter their function. KCNE2 (also known as MiRP1) is expressed in the heart, is associated with human cardiac arrhythmia, and modulates cardiac Kv alpha subunits hERG and KCNQ1 in vitro. KCNE2 and KCNQ1 are also expressed in parietal cells, leading to speculation they form a native channel complex there. Here, we disrupted the murine kcne2 gene and found that kcne2 (-/-) mice have a severe gastric phenotype with profoundly reduced parietal cell proton secretion, abnormal parietal cell morphology, achlorhydria, hypergastrinemia, and striking gastric glandular hyperplasia arising from an increase in the number of non-acid secretory cells. KCNQ1 exhibited abnormal distribution in gastric glands from kcne2 (-/-) mice, with increased expression in non-acid secretory cells. Parietal cells from kcne2 (+/-) mice exhibited normal architecture but reduced proton secretion, and kcne2 (+/-) mice were hypochlorhydric, indicating a gene-dose effect and a primary defect in gastric acid secretion. These data demonstrate that KCNE2 is essential for gastric acid secretion, the first genetic evidence that a member of the KCNE gene family is required for normal gastrointestinal function.


Asunto(s)
Ácido Gástrico/metabolismo , Canales de Potasio con Entrada de Voltaje/fisiología , Subunidades de Proteína/fisiología , Aclorhidria/genética , Animales , Células Cultivadas , Femenino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Mucosa Gástrica/ultraestructura , Gastrinas/antagonistas & inhibidores , Gastrinas/biosíntesis , Marcación de Gen , Hiperplasia/genética , Hipertrofia/genética , Canal de Potasio KCNQ1/biosíntesis , Canal de Potasio KCNQ1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Parietales Gástricas/metabolismo , Células Parietales Gástricas/patología , Células Parietales Gástricas/ultraestructura , Canales de Potasio con Entrada de Voltaje/deficiencia , Canales de Potasio con Entrada de Voltaje/genética , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Estómago/patología , Estómago/ultraestructura , Regulación hacia Arriba/genética
13.
J Cell Biol ; 169(1): 21-8, 2005 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-15809309

RESUMEN

Loss of gastric acid secretion is pathologically known as achlorhydria. Acid-secreting parietal cells are characterized by abundant expression of ezrin (Vil2), one of ezrin/radixin/moesin proteins, which generally cross-link actin filaments with plasma membrane proteins. Here, we show the direct in vivo involvement of ezrin in gastric acid secretion. Ezrin knockout (Vil2(-/-)) mice did not survive >1.5 wk after birth, making difficult to examine gastric acid secretion. We then generated ezrin knockdown (Vil2(kd/kd)) mice by introducing a neomycin resistance cassette between exons 2 and 3. Vil2(kd/kd) mice born at the expected Mendelian ratio exhibited growth retardation and a high mortality. Approximately 7% of Vil2(kd/kd) mice survived to adulthood. Ezrin protein levels in Vil2(kd/kd) stomachs decreased to <5% of the wild-type levels without compensatory up-regulation of radixin or moesin. Adult Vil2(kd/kd) mice suffered from severe achlorhydria. Immunofluorescence and electron microscopy revealed that this achlorhydria was caused by defects in the formation/expansion of canalicular apical membranes in gastric parietal cells.


Asunto(s)
Aclorhidria/metabolismo , Mucosa Gástrica/metabolismo , Células Parietales Gástricas/metabolismo , Fosfoproteínas/metabolismo , Aclorhidria/genética , Ácidos/metabolismo , Animales , Proteínas Sanguíneas/metabolismo , Polaridad Celular/efectos de los fármacos , Polaridad Celular/genética , Polaridad Celular/fisiología , Clonación Molecular , Proteínas del Citoesqueleto/metabolismo , Histamina/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica de Transmisión , Microvellosidades/metabolismo , Microvellosidades/ultraestructura , Células Parietales Gástricas/efectos de los fármacos , Células Parietales Gástricas/ultraestructura , Fosfoproteínas/genética , Estómago/efectos de los fármacos , Estómago/ultraestructura , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Hum Mol Genet ; 13(22): 2813-21, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15385447

RESUMEN

Increased susceptibility to gastric cancer has been associated with a wide range of host genetic and environmental factors, including Helicobacter pylori infection. Helicobacter pylori infection is postulated to initiate a progression through atrophic gastritis, metaplasia and dysplasia to cancer, and has been associated with reduction of acid output and dysregulation of stomach mucins. Here, we present the characterization of two mouse lines carrying mutant alleles of the gene encoding the Kcnq1 potassium channel, which very rapidly establish chronic gastritis in a pathogen-exposed environment. These mice develop gastric hyperplasia, hypochlorhydria and mucin dysregulation independent of infection. Metaplasia, dysplasia and pre-malignant adenomatous hyperplasia of the stomach have been observed in these Kcnq1 mutant mice, also independent of infection. The data presented here suggest that Kcnq1 mutant mice can be used both as an efficient model for the development of atrophic gastritis after infection and to determine the processes during the later stages of progression to gastric cancer independent of infection. Thus, Kcnq1 mutant mice are a powerful new tool for investigating the connection between acid balance, Helicobacter infection and mucin disruption in the progression to gastric cancer.


Asunto(s)
Adenoma/genética , Gastritis/genética , Canales de Potasio KCNQ/genética , Lesiones Precancerosas/genética , Neoplasias Gástricas/genética , Aclorhidria/etiología , Aclorhidria/genética , Aclorhidria/patología , Adenoma/patología , Alelos , Animales , Enfermedad Crónica , Femenino , Mucinas Gástricas/metabolismo , Gastritis/etiología , Gastritis/patología , Predisposición Genética a la Enfermedad , Infecciones por Helicobacter/complicaciones , Helicobacter pylori , Hiperplasia , Masculino , Metaplasia , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Miocardio/patología , Lesiones Precancerosas/patología , Neoplasias Gástricas/etiología , Neoplasias Gástricas/patología , Estría Vascular/patología , Translocación Genética
15.
J Biol Chem ; 279(29): 30531-9, 2004 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-15123620

RESUMEN

The AE2 Cl-/HCO3- exchanger is expressed in numerous cell types, including epithelial cells of the kidney, respiratory tract, and alimentary tract. In gastric epithelia, AE2 is particularly abundant in parietal cells, where it may be the predominant mechanism for HCO3- efflux and Cl- influx across the basolateral membrane that is needed for acid secretion. To investigate the hypothesis that AE2 is critical for parietal cell function and to assess its importance in other tissues, homozygous null mutant (AE2(-/-)) mice were prepared by targeted disruption of the AE2 (Slc4a2) gene. AE2(-/-) mice were emaciated, edentulous (toothless), and exhibited severe growth retardation, and most of them died around the time of weaning. AE2(-/-) mice exhibited achlorhydria, and histological studies revealed abnormalities of the gastric epithelium, including moderate dilation of the gastric gland lumens and a reduction in the number of parietal cells. There was little evidence, however, that parietal cell viability was impaired. Ultrastructural analysis of AE2(-/-) gastric mucosa revealed abnormal parietal cell structure, with severely impaired development of secretory canaliculi and few tubulovesicles but normal apical microvilli. These results demonstrate that AE2 is essential for gastric acid secretion and for normal development of secretory canalicular and tubulovesicular membranes in mouse parietal cells.


Asunto(s)
Proteínas de Transporte de Anión , Antiportadores , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Aclorhidria/genética , Alelos , Animales , Northern Blotting , Western Blotting , Supervivencia Celular , Antiportadores de Cloruro-Bicarbonato , Células Epiteliales/metabolismo , Epitelio/metabolismo , Mucosa Gástrica/metabolismo , Vectores Genéticos , Concentración de Iones de Hidrógeno , Membranas Intracelulares/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Mutación , Células Parietales Gástricas/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Proteínas SLC4A , Transgenes
16.
Br J Nutr ; 88(3): 265-71, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12207836

RESUMEN

Free radicals and reactive species produced in vivo can trigger cell damage and DNA modifications resulting in carcinogenesis. Dietary antioxidants trap these species limiting their damage. The present study evaluated the role of vitamins C and E in the prevention of potentially premalignant modifications to DNA in the human stomach by supplementing patients who, because of hypochlorhydria and possible depletion of gastric antioxidants, could be at increased risk of gastric cancer. Patients undergoing surveillance for Barrett's oesophagus (n 100), on long-term proton pump inhibitors were randomized into two groups: vitamin C (500 mg twice/d) and vitamin E (100 mg twice/d) for 12 weeks (the supplemented group) or placebo. Those attending for subsequent endoscopy had gastric juice, plasma and mucosal measurements of vitamin levels and markers of DNA damage. Seventy-two patients completed the study. Plasma ascorbic acid, total vitamin C and vitamin E were elevated in the supplemented group consistent with compliance. Gastric juice ascorbic acid and total vitamin C levels were raised significantly in the supplemented group (P=0.01) but supplementation had no effect on the mucosal level of this vitamin. However, gastric juice ascorbic acid and total vitamin C were within normal ranges in the unsupplemented group. Mucosal malondialdehyde, chemiluminescence and DNA damage levels in the comet assay were unaffected by vitamin supplementation. In conclusion, supplementation does not affect DNA damage in this group of patients. This is probably because long-term inhibition of the gastric proton pump alone does not affect gastric juice ascorbate and therefore does not increase the theoretical risk of gastric cancer because of antioxidant depletion.


Asunto(s)
Aclorhidria/genética , Antiácidos/efectos adversos , Antioxidantes/uso terapéutico , Transformación Celular Neoplásica/efectos de los fármacos , Daño del ADN , Suplementos Dietéticos , Aclorhidria/metabolismo , Adulto , Anciano , Antioxidantes/farmacocinética , Ácido Ascórbico/farmacocinética , Ácido Ascórbico/uso terapéutico , Esófago de Barrett/genética , Esófago de Barrett/metabolismo , Femenino , Determinación de la Acidez Gástrica , Jugo Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Inhibidores de la Bomba de Protones , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Vitamina E/farmacocinética , Vitamina E/uso terapéutico
17.
Gastroenterology ; 123(1): 92-105, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12105837

RESUMEN

BACKGROUND & AIMS: Interleukin-1 beta (IL-1beta) polymorphisms are associated with increased risk of gastric cancer in whites. This study aimed to examine effects of these polymorphisms on gastric acid secretion, atrophic gastritis, and risk of peptic ulcer in Japan. METHODS: We determined IL-1B-511/-31 and IL-1RN genotypes and measured gastric juice pH, serum pepsinogen (PG) I and II levels, and gastritis and atrophy scores in Helicobacter pylori-positive patients with gastritis only, gastric ulcers, or duodenal ulcers (DUs), and H. pylori-negative controls. RESULTS: In the H. pylori-positive group, subjects with the proinflammatory IL-1B-511 T/T genotype had the highest atrophy and gastritis scores, the highest median gastric juice pH, and the lowest median serum PG I/PG II ratios. Although gastric juice pH significantly increased and serum PG I and PG I/PG II ratios significantly decreased in the IL-1B-511 T/T genotype group with age, no such age-dependent changes were observed in the C/C genotype group. Changes in the C/T genotype group were intermediate. In the H. pylori-negative group, the IL-1 loci had no effect on any of the physiologic or morphologic parameters. Carriage of IL-1RN allele 2 significantly protected against DU disease while the IL-1B-511 T/T genotype significantly protected against DU recurrence in patients older than 60 years. CONCLUSIONS: Proinflammatory IL-1beta polymorphisms are associated with hypochlorhydria and atrophic gastritis in Japan. The effects are dependent on H. pylori infection and become more significant with advancing age. This may explain the high incidence of gastric cancer in Japan and also the age-dependent decrease in DU recurrence in infected subjects.


Asunto(s)
Aclorhidria/genética , Pueblo Asiatico/genética , Úlcera Duodenal/genética , Gastritis Atrófica/genética , Predisposición Genética a la Enfermedad/genética , Interleucina-1/genética , Polimorfismo Genético , Alelos , Femenino , Jugo Gástrico/química , Gastritis Atrófica/metabolismo , Gastritis Atrófica/microbiología , Gastritis Atrófica/patología , Frecuencia de los Genes , Genotipo , Helicobacter pylori/aislamiento & purificación , Humanos , Concentración de Iones de Hidrógeno , Proteína Antagonista del Receptor de Interleucina 1 , Japón , Masculino , Persona de Mediana Edad , Pepsinógeno A/sangre , Pepsinógeno C/sangre , Úlcera Péptica/genética , Recurrencia , Sialoglicoproteínas/genética
18.
J Biol Chem ; 275(28): 21555-65, 2000 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-10764766

RESUMEN

The H,K-ATPase of the gastric parietal cell is the most critical component of the ion transport system mediating acid secretion in the stomach. To study the requirement of this enzyme in the development, maintenance, and function of the gastric mucosa, we used gene targeting to prepare mice lacking the alpha-subunit. Homozygous mutant (Atp4a(-/-)) mice appeared healthy and exhibited normal systemic electrolyte and acid-base status but were achlorhydric and hypergastrinemic. Immunocytochemical, histological, and ultrastructural analyses of Atp4a(-/-) stomachs revealed the presence of chief cells, demonstrating that the lack of acid secretion does not interfere with their differentiation. Parietal cells were also present in normal numbers, and despite the absence of alpha-subunit mRNA and protein, the beta-subunit was expressed. However, Atp4a(-/-) parietal cells had dilated canaliculi and lacked typical canalicular microvilli and tubulovesicles, and subsets of these cells contained abnormal mitochondria and/or massive glycogen stores. Stomachs of adult Atp4a(-/-) mice exhibited metaplasia, which included the presence of ciliated cells. We conclude that ablation of the H,K-ATPase alpha-subunit causes achlorhydria and hypergastrinemia, severe perturbations in the secretory membranes of the parietal cell, and metaplasia of the gastric mucosa; however, the absence of the pump appears not to perturb parietal cell viability or chief cell differentiation.


Asunto(s)
Aclorhidria/genética , Cilios/patología , Mucosa Gástrica/patología , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , Células Parietales Gástricas/patología , Aclorhidria/sangre , Aclorhidria/patología , Animales , Electrólitos/sangre , Ácido Gástrico/metabolismo , Mucosa Gástrica/ultraestructura , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Metaplasia , Ratones , Ratones Noqueados , Células Parietales Gástricas/ultraestructura , Pepsinógeno A/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...