Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Gastroenterology ; 156(8): 2158-2173, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30831083

RESUMO

Acidification of the gastric lumen poses a barrier to transit of potentially pathogenic bacteria and enables activation of pepsin to complement nutrient proteolysis initiated by salivary proteases. Histamine-induced activation of the PKA signaling pathway in gastric corpus parietal cells causes insertion of proton pumps into their apical plasma membranes. Parietal cell secretion and homeostasis are regulated by signaling pathways that control cytoskeletal changes required for apical membrane remodeling and organelle and proton pump activities. Helicobacter pylori colonization of human gastric mucosa affects gastric epithelial cell plasticity and homeostasis, promoting epithelial progression to neoplasia. By intervening in proton pump expression, H pylori regulates the abundance and diversity of microbiota that populate the intestinal lumen. We review stimulation-secretion coupling and renewal mechanisms in parietal cells and the mechanisms by which H pylori toxins and effectors alter cell secretory pathways (constitutive and regulated) and organelles to establish and maintain their inter- and intracellular niches. Studies of bacterial toxins and their effector proteins have provided insights into parietal cell physiology and the mechanisms by which pathogens gain control of cell activities, increasing our understanding of gastrointestinal physiology, microbial infectious disease, and immunology.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Células Parietais Gástricas/patologia , Lesões Pré-Cancerosas/patologia , Neoplasias Gástricas/patologia , Antibacterianos/uso terapêutico , Feminino , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Microbioma Gastrointestinal/fisiologia , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/tratamento farmacológico , Humanos , Masculino , Lesões Pré-Cancerosas/tratamento farmacológico , Prognóstico , Medição de Risco , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/cirurgia , Resultado do Tratamento
2.
Curr Top Microbiol Immunol ; 400: 227-252, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28124156

RESUMO

Appropriate management of Helicobacter pylori infection of the human stomach is evolving and remains a significant clinical challenge. Acute infection results in hypochlorhydria, whereas chronic infection results in either hypo- or hyperchlorhydria, depending upon the anatomic site of infection. Acute hypochlorhydria facilitates survival of the bacterium and its infection of the stomach. Interestingly, most patients chronically infected with H. pylori manifest a pangastritis with reduced acid secretion due to bacterial virulence factors, inflammatory cytokines, and various degrees of gastric atrophy. While these patients are predisposed to develop gastric adenocarcinoma (~1%), there is increasing evidence from population studies that they are also protected from gastroesophageal reflux disease (GERD), Barrett's esophagus (BE), and esophageal adenocarcinoma (EAC). Eradication of H. pylori, in these patients, may provoke GERD in predisposed individuals and may be a contributory factor for the rising incidence of refractory GERD, BE, and EAC observed in Westernized societies. Only ~10% of chronically infected patients, mainly the young, manifest an antral predominant gastritis with increased acid secretion due to a decrease in somatostatin and increase in gastrin secretion; these patients are predisposed to develop peptic ulcer disease. H. pylori-induced changes in acid secretion, in particular hypochlorhydria, may allow ingested microorganisms to survive transit through the stomach and colonize the distal intestine and colon. Such perturbation of gut microbiota, i.e. dysbiosis, may influence human health and disease.


Assuntos
Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Helicobacter pylori/genética , Humanos , Estômago/microbiologia , Estômago/patologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
3.
PLoS One ; 11(10): e0163696, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27783698

RESUMO

The stomach, which is characterized by acid peptic digestion in vertebrates, has been lost secondarily multiple times in the evolution of the teleost fishes. The Cypriniformes are largely seen as an agastric order; however, within the superfamily Cobitoidea, the closely related sister groups Nemacheilidae and Balitoridae have been identified as gastric families. The presence of these most recently diverged gastric families in an otherwise agastric clade indicates that either multiple (>2-3) loss events occurred with the Cyprinidae, Catostomidae and Cobitidae, or that gastric reinvention arose in a recent ancestor of the Nemacheilidae/Balitoridae sister clade. In the present study, the foregut regions of Cobitidae, Nemacheilidae/Balitoridae and the ancestral Botiidae family members were examined for the presence of gastric glands and gastric proton pump (Atp4a) α subunit expression by histology and immunohistochemistry respectively. Atp4a gene expression was assessed by reverse transcriptase-polymerase chain reaction (RT-PCR). Gastric glands expressing apical H+/K+-ATPase α subunit and isolated partial sequences of atp4a, identified using degenerate primers showing clear orthology to other vertebrate atp4a sequences, were detected in representative species from Nemacheilidae/ Balitoridae and Botiidae, but not Cobitidae (Misgurnus anguillicaudatus). In summary, we provide evidence for an uninterrupted gastric evolutionary lineage in the Cobitoidea, making it highly improbable that the stomach was reinvented in the Nemacheilidae/Balitoridae clade consistent with Dollo's principle. These results also indicate that the gastric trait may be present elsewhere in the Cobitoidea.


Assuntos
Cipriniformes/metabolismo , Mucosa Gástrica/metabolismo , Sequência de Aminoácidos , Animais , Cipriniformes/classificação , Evolução Molecular , Expressão Gênica , ATPase Trocadora de Hidrogênio-Potássio/química , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Intestinos/patologia , Dados de Sequência Molecular , Fenótipo , Filogenia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA/isolamento & purificação , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Estômago/patologia
4.
Kardiol Pol ; 74(8): 726-732, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26898969

RESUMO

BACKGROUND: Commonly used cardiopulmonary bypass systems with cardiotomy reservoir, oxygenator, and roller pump require preoperative crystalloid filling. Radical reduction of the filling fluid volume and replacing it with the patient's own blood has a fundamental impact on the outcome. AIM: A comparison of cardiopulmonary bypass filled with the patient's blood, applied in Poland for the first time, and the classical system filled with crystalloids. METHODS: Non-randomised trial in which patients undergoing coronary artery bypass grafting were divided into two groups: first operated on with use of cardiopulmonary bypass system with the patient's own blood priming, and a control group operated on with standard technique. Levels of haemoglobin (HGB), haematocrit (HCT), platelets, leukocytes, creatinine, protein, C-reactive protein, procalcitonin, volume of transfused blood products, postoperative drain output, time to extubation, and consumption of catecholamines were compared. RESULTS: The results of a study comparing the classical system with the blood-filled system (n = 60) showed a significantly smaller decrease in HGB and HCT levels (p = 0.001), resulting in reduction of blood product transfusions by 75% (p = 0.03). The new type of extracorporeal circulation reduced the total postoperative drain output by approximately 28% (p = 0.003). The systemic inflammatory response syndrome (SIRS) was less pronounced and the tissue perfusion was better due to smaller degree of haemodilution leading to better organ and heart protection. The patients required shorter mechanical ventilation times in the perioperative period. CONCLUSIONS: The use of a new system of cardiopulmonary bypass filled with the patient's blood reduces the postoperative decrease in HGB and HCT, the amount of transfused blood products, and total postoperative drain output. It also shortens the time spent on mechanical ventilatory support.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Ponte de Artéria Coronária , Soluções Isotônicas/efeitos adversos , Idoso , Proteína C-Reativa , Soluções Cristaloides , Feminino , Hemodiluição , Humanos , Masculino , Pessoa de Meia-Idade , Polônia , Síndrome de Resposta Inflamatória Sistêmica/etiologia , Resultado do Tratamento
5.
Am J Physiol Gastrointest Liver Physiol ; 309(3): G193-201, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26045613

RESUMO

Acute Helicobacter pylori infection of gastric epithelial cells and human gastric biopsies represses H,K-ATPase α subunit (HKα) gene expression and inhibits acid secretion, causing transient hypochlorhydria and supporting gastric H. pylori colonization. Infection by H. pylori strains deficient in the cag pathogenicity island (cag PAI) genes cagL, cagE, or cagM, which do not transfer CagA into host cells or induce interleukin-8 secretion, does not inhibit HKα expression, nor does a cagA-deficient strain that induces IL-8. To test the hypothesis that virulence factors other than those mediating CagA translocation or IL-8 induction participate in HKα repression by activating NF-κB, AGS cells transfected with HKα promoter-Luc reporter constructs containing an intact or mutated NF-κB binding site were infected with wild-type H. pylori strain 7.13, isogenic mutants lacking cag PAI genes responsible for CagA translocation and/or IL-8 induction (cagA, cagζ, cagε, cagZ, and cagß), or deficient in genes encoding two peptidoglycan hydrolases (slt and cagγ). H. pylori-induced AGS cell HKα promoter activities, translocated CagA, and IL-8 secretion were measured by luminometry, immunoblotting, and ELISA, respectively. Human gastric biopsy acid secretion was measured by microphysiometry. Taken together, the data showed that HKα repression is independent of IL-8 expression, and that CagA translocation together with H. pylori transglycosylases encoded by slt and cagγ participate in NF-κB-dependent HKα repression and acid inhibition. The findings are significant because H. pylori factors other than CagA and IL-8 secretion are now implicated in transient hypochlorhydria which facilitates gastric colonization and potential triggering of epithelial progression to neoplasia.


Assuntos
Mucosa Gástrica/metabolismo , Helicobacter pylori , NF-kappa B/metabolismo , Bombas de Próton/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Acloridria/etiologia , Acloridria/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Células Cultivadas , Células Epiteliais/metabolismo , Ácido Gástrico/metabolismo , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/metabolismo , Helicobacter pylori/patogenicidade , Helicobacter pylori/fisiologia , Humanos , Interleucina-8/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais , Fatores de Virulência/metabolismo
6.
CBE Life Sci Educ ; 14(1): ar3, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25673355

RESUMO

Academic medical centers nationwide face numerous fiscal challenges resulting from implementation of restructured healthcare delivery models, contracting state support for higher education, and increased competition for federal and other sources of biomedical research funding. In pursuing greater accountability and transparency in its fiscal operations, the Medical University of South Carolina (MUSC) has implemented a responsibility centers management budgetary model, which requires all MUSC colleges to be eventually self-sustaining financially. Graduate schools in the biomedical sciences are particularly vulnerable in the face of these challenges, depending traditionally as they do on financial support from training grant tuition, occasional medical school tuition and medical practice plan revenues, graduate college-based revenue-generating programs, and faculty payment of PhD tuition. The revenue streams are often insufficient to support PhD training programs, and supplemental financial support is required from the institution. In the context of a college of graduate studies, estimates of the cost of educating a graduate student become a significant necessity. This study presents a readily applicable model of empirically estimating the faculty salary costs that may provide a basis for budgetary planning that will help to sustain a biomedical sciences graduate school's commitment to its teaching, research, and service mission goals.


Assuntos
Biologia/economia , Biologia/educação , Educação de Pós-Graduação/economia , Docentes , Faculdades de Medicina/economia , Centros Médicos Acadêmicos/economia , Pesquisa Biomédica/economia , Pesquisa Biomédica/educação , Coleta de Dados , Docentes de Medicina , Humanos , Salários e Benefícios , Ciência/economia , South Carolina , Estudantes , Apoio ao Desenvolvimento de Recursos Humanos , Universidades/economia
7.
Dig Dis Sci ; 60(6): 1645-54, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25577268

RESUMO

BACKGROUND: Most patients presenting with symptoms of esophageal cancer (EC) have advanced disease. Even with resection, the cure rate is extremely low due to local recurrence and metastatic disease. Early detection and effective therapeutic intervention are essential to improve survival. AIMS: This study tested the hypothesis that the presence of EC modulates concentrations of specific plasma proteins and peptides, potentially allowing discrimination between EC and controls based on mass spectrometric analysis of the respective plasma proteomes. METHODS: Blood samples from 79 esophageal cancer patients and 40 age-matched normal subjects were processed to plasma, and protein/peptide sub-fractions were isolated using HIC8 or WAX-derivatized superparamagnetic beads. Triplicate matrix-assisted laser desorption time-of-flight mass spectra were acquired for specific plasma fractions from each subject. RESULTS: HIC8 and WAX-derivatized plasma eluates yielded 79 and 77 candidate features, respectively, and a Random Forest algorithm identified a subset of features whose peak intensities allowed discrimination between cancer patients and controls. Areas under the curve in receiver operating characteristic curves for HIC8 spectra were 0.88 and 0.83 for WAX spectra. The combined feature set discriminated EC from control plasma with 79 % sensitivity and 79 % specificity, with positive and negative test likelihood ratios of >14 and 0.17, respectively. CONCLUSIONS: These data lay the foundation for the development of a clinically useful test for esophageal cancer based on statistical analysis of proteomic spectra of patient plasma samples. This approach will be validated by analysis of larger patient cohorts, development of cancer-specific classifiers, and assessment of racial origin imbalances.


Assuntos
Adenocarcinoma/sangue , Biomarcadores Tumorais/sangue , Carcinoma de Células Escamosas/sangue , Neoplasias Esofágicas/sangue , Proteômica/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade
8.
Am J Physiol Gastrointest Liver Physiol ; 306(7): G606-13, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24503769

RESUMO

Acute Helicobacter pylori infection of gastric epithelial cells induces CagA oncoprotein- and peptidoglycan (SLT)-dependent mobilization of NF-κB p50 homodimers that bind to H-K-ATPase α-subunit (HKα) promoter and repress HKα gene transcription. This process may facilitate gastric H. pylori colonization by induction of transient hypochlorhydria. We hypothesized that H. pylori also regulates HKα expression posttranscriptionally by miRNA interaction with HKα mRNA. In silico analysis of the HKα 3' untranslated region (UTR) identified miR-1289 as a highly conserved putative HKα-regulatory miRNA. H. pylori infection of AGS cells transfected with HKα 3' UTR-Luc reporter construct repressed luciferase activity by 70%, whereas ΔcagA or Δslt H. pylori infections partially abrogated repression. Transfection of AGS cells expressing HKα 3' UTR-Luc construct with an oligoribonucleotide mimetic of miR-1289 induced maximal repression (54%) of UTR activity within 30 min; UTR activity was unchanged by nontargeting siRNA transfection. Gastric biopsies from patients infected with cagA(+) H. pylori showed a significant increase in miR-1289 expression compared with uninfected patients or those infected with cagA(-) H. pylori. Finally, miR-1289 expression was necessary and sufficient to attenuate biopsy HKα protein expression in the absence of infection. Taken together, these data indicate that miR-1289 is upregulated by H. pylori in a CagA- and SLT-dependent manner and targets HKα 3' UTR, affecting HKα mRNA translation. The sensitivity of HKα mRNA 3' UTR to binding of miR-1289 identifies a novel regulatory mechanism of gastric acid secretion and offers new insights into mechanisms underlying transient H. pylori-induced hypochlorhydria.


Assuntos
Mucosa Gástrica/enzimologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Infecções por Helicobacter/enzimologia , Helicobacter pylori/metabolismo , MicroRNAs/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Células Parietais Gástricas/enzimologia , Processamento Pós-Transcricional do RNA , Regiões 3' não Traduzidas , Acloridria/enzimologia , Acloridria/microbiologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Linhagem Celular , Mucosa Gástrica/microbiologia , Regulação Enzimológica da Expressão Gênica , Genes Reporter , ATPase Trocadora de Hidrogênio-Potássio/genética , Infecções por Helicobacter/complicações , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Subunidade p50 de NF-kappa B/genética , Células Parietais Gástricas/microbiologia , Peptidoglicano/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção , Virulência
9.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G157-66, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23154976

RESUMO

Gastric acid secretion by the H(+)-K(+)-ATPase at the apical surface of activated parietal cells requires luminal K(+) provided by the KCNQ1/KCNE2 K(+) channel. However, little is known about the trafficking and relative spatial distribution of KCNQ1 and H(+)-K(+)-ATPase in resting and activated parietal cells and the capacity of KCNQ1 to control acid secretion. Here we show that inhibition of KCNQ1 activity quickly curtails gastric acid secretion in vivo, even when the H(+)-K(+)-ATPase is permanently anchored in the apical membrane, demonstrating a key role of the K(+) channel in controlling acid secretion. Three-dimensional imaging analysis of isolated mouse gastric units revealed that the majority of KCNQ1 resides in an intracytoplasmic, Rab11-positive compartment in resting parietal cells, distinct from H(+)-K(+)-ATPase-enriched tubulovesicles. Upon activation, there was a significant redistribution of H(+)-K(+)-ATPase and KCNQ1 from intracytoplasmic compartments to the apical secretory canaliculi. Significantly, high Förster resonance energy transfer was detected between H(+)-K(+)-ATPase and KCNQ1 in activated, but not resting, parietal cells. These findings demonstrate that H(+)-K(+)-ATPase and KCNQ1 reside in independent intracytoplasmic membrane compartments, or membrane domains, and upon activation of parietal cells, both membrane proteins are transported, possibly via Rab11-positive recycling endosomes, to apical membranes, where the two molecules are closely physically opposed. In addition, these studies indicate that acid secretion is regulated by independent trafficking of KCNQ1 and H(+)-K(+)-ATPase.


Assuntos
Ácido Gástrico/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Canal de Potássio KCNQ1/metabolismo , Células Parietais Gástricas/enzimologia , Animais , Membrana Celular/enzimologia , Cromanos/farmacologia , Citoplasma/enzimologia , Endossomos/enzimologia , Transferência Ressonante de Energia de Fluorescência , Histamina/metabolismo , Canal de Potássio KCNQ1/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Transporte Proteico , Sulfonamidas/farmacologia , Fatores de Tempo , Proteínas rab de Ligação ao GTP/metabolismo
10.
J Gastroenterol ; 47(6): 609-18, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22565637

RESUMO

Infection of the human stomach mucosa by Helicobacter pylori induces strong inflammatory responses and a transitory hypochlorhydria which can progress in ~2 % of patients to atrophic gastritis, dysplasia, or gastric adenocarcinoma. H. pylori infection of gastric biopsies or cultured gastric epithelial cells in vitro represses the activity of endogenous or transfected promoter of the alpha-subunit (HKα) of gastric H,K-adenosine triphosphatase (H,K-ATPase), the parietal cell enzyme mediating acid secretion. Some mechanistic details of H. pylori-mediated repression of HKα and ensuing hypochlorhydria have been recently elucidated. H. pylori strains expressing a type IV secretion system (T4SS) encoded by the cag pathogenicity island are known to upregulate the transcription factor nuclear factor (NF)-κB. The NF-κB-binding regions in the HKα promoter were identified and shown to repress its transcriptional activity. Interaction studies have indicated that although active phosphorylated NF-κB p65 is present in infected cells, an NF-κB p50/p65 heterodimeric complex fails to bind to the HKα promoter. Point mutations at -159 and -161 bp in the HKα promoter NF-κB binding sequence prevent the binding of NF-κB p50 and prevent H. pylori repression of point-mutated HKα promoter activity. The T4SS factors CagL, CagE, CagM, and possibly CagA and the lytic transglycosylase Slt, are mechanistically involved in NF-κB activation and repression of HKα transcription. CagL, a T4SS pilus component, binds to the integrin α(5)ß(1) to mediate translocation of virulence factors into the host cell and initiate signaling. During acute H. pylori infection, CagL dissociates ADAM 17 (a disintegrin and a metalloprotease 17) from the integrin α(5)ß(1) complex and stimulates ADAM17-dependent release of heparin-binding epidermal growth factor (HB-EGF), EGF receptor (EGFR) stimulation, ERK1/2 kinase activation, and NF-κB-mediated repression of HKα. These studies suggest that H. pylori inhibits HKα gene expression by an integrin α(5)ß(1) → ADAM17 → HB-EGF → EGFR → ERK1/2 → NF-κB pathway mediating NF-κB p50 homodimer binding to the HKα promoter. Here we review the molecular basis and recent progress of this novel pathogen-dependent mechanism of H,K-ATPase inhibition, which contributes significantly to our current understanding of H. pylori pathophysiology.


Assuntos
Ácido Gástrico/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Acloridria/microbiologia , Animais , Modelos Animais de Doenças , Medicina Baseada em Evidências/métodos , ATPase Trocadora de Hidrogênio-Potássio/genética , Infecções por Helicobacter/complicações , Infecções por Helicobacter/genética , Humanos , Bombas de Próton/fisiologia , Transcrição Gênica
11.
J Clin Invest ; 122(4): 1553-66, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22378042

RESUMO

Many bacterial pathogens inject into host cells effector proteins that are substrates for host tyrosine kinases such as Src and Abl family kinases. Phosphorylated effectors eventually subvert host cell signaling, aiding disease development. In the case of the gastric pathogen Helicobacter pylori, which is a major risk factor for the development of gastric cancer, the only known effector protein injected into host cells is the oncoprotein CagA. Here, we followed the hierarchic tyrosine phosphorylation of H. pylori CagA as a model system to study early effector phosphorylation processes. Translocated CagA is phosphorylated on Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs EPIYA-A, EPIYA-B, and EPIYA-C in Western strains of H. pylori and EPIYA-A, EPIYA-B, and EPIYA-D in East Asian strains. We found that c-Src only phosphorylated EPIYA-C and EPIYA-D, whereas c-Abl phosphorylated EPIYA-A, EPIYA-B, EPIYA-C, and EPIYA-D. Further analysis revealed that CagA molecules were phosphorylated on 1 or 2 EPIYA motifs, but never simultaneously on 3 motifs. Furthermore, none of the phosphorylated EPIYA motifs alone was sufficient for inducing AGS cell scattering and elongation. The preferred combination of phosphorylated EPIYA motifs in Western strains was EPIYA-A and EPIYA-C, either across 2 CagA molecules or simultaneously on 1. Our study thus identifies a tightly regulated hierarchic phosphorylation model for CagA starting at EPIYA-C/D, followed by phosphorylation of EPIYA-A or EPIYA-B. These results provide insight for clinical H. pylori typing and clarify the role of phosphorylated bacterial effector proteins in pathogenesis.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Helicobacter pylori/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteína Tirosina Quinase CSK , Linhagem Celular/microbiologia , Europa (Continente)/epidemiologia , Ásia Oriental/epidemiologia , Infecções por Helicobacter/etnologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/classificação , Helicobacter pylori/genética , Helicobacter pylori/isolamento & purificação , Interações Hospedeiro-Patógeno , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Fosfotirosina/metabolismo , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Quinases da Família src
12.
J Biol Chem ; 286(16): 14120-8, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21367857

RESUMO

Kir4.1 channels were found to colocalize with the H(+)/K(+)-ATPase throughout the parietal cell (PC) acid secretory cycle. This study was undertaken to explore their functional role. Acid secretory rates, electrophysiological parameters, PC ultrastructure, and gene and protein expression were determined in gastric mucosae of 7-8-day-old Kir4.1-deficient mice and WT littermates. Kir4.1(-/-) mucosa secreted significantly more acid and initiated secretion significantly faster than WT mucosa. No change in PC number but a relative up-regulation of H(+)/K(+)-ATPase gene and protein expression (but not of other PC ion transporters) was observed. Electron microscopy revealed fully fused canalicular membranes and a lack of tubulovesicles in resting state Kir4.1(-/-) PCs, suggesting that Kir4.1 ablation may also interfere with tubulovesicle endocytosis. The role of this inward rectifier in the PC apical membrane may therefore be to balance between K(+) loss via KCNQ1/KCNE2 and K(+) reabsorption by the slow turnover of the H(+)/K(+)-ATPase, with consequences for K(+) reabsorption, inhibition of acid secretion, and membrane recycling. Our results demonstrate that Kir4.1 channels are involved in the control of acid secretion and suggest that they may also affect secretory membrane recycling.


Assuntos
Ácido Gástrico/metabolismo , Regulação da Expressão Gênica , Células Parietais Gástricas/citologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Animais , Transporte Biológico , Eletrofisiologia/métodos , Endocitose , Mucosa Gástrica/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/química , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão/métodos , Microscopia de Fluorescência/métodos , Potássio/química , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese
13.
Gut ; 59(7): 874-81, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20581234

RESUMO

BACKGROUND AND AIMS: Helicobacter pylori infection of gastric mucosa causes gastritis and transient hypochlorhydria, which may provoke emergence of a mucosal precancer phenotype; H pylori strains containing a cag pathogenicity island (PAI) augment cancer risk. Acid secretion is mediated by the catalytic alpha subunit of parietal cell H,K-ATPase (HKalpha). In AGS gastric epithelial cells, H pylori induces nuclear factor-kappaB (NF-kappaB) binding to and repression of transfected HKalpha promoter activity. This study sought to identify bacterial genes involved in HKalpha repression and to assess their impact on acid secretion. METHODS AND RESULTS: AGS cells transfected with an HKalpha promoter construct or human gastric body biopsies were infected with wild-type (wt) or isogenic mutant (IM) H pylori strains. AGS cell HKalpha promoter activity, and biopsy HKalpha mRNA, protein and H(+) secretory activity were measured by luminometry, reverse transcription-PCR, immunoblotting and extracellular acidification, respectively. Wt H pylori and DeltavacA, DeltaureA, Deltaslt and DeltaflaA IM strains repressed HKalpha promoter activity by approximately 50%, a DeltacagA IM strain repressed HKalpha by approximately 33%, and DeltacagE, DeltacagM and DeltacagL IM strains elicited no HKalpha repression. Wt H pylori-infected biopsies had markedly reduced HKalpha mRNA and protein compared with IM strain infections or mock-infected controls. Histamine-stimulated, SCH28080-sensitive biopsy acid secretion was significantly inhibited by wt but not by DeltacagL IM H pylori infection compared with vehicle-only controls. CONCLUSIONS: It is concluded that H pylori cag PAI gene products CagE, CagM, CagL and, possibly, CagA are mechanistically involved in repression of HKalpha transcription. Further, acute H pylori infection of human gastric mucosa downregulates parietal cell H,K-ATPase expression, significantly inhibiting acid secretion.


Assuntos
Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/genética , Infecções por Helicobacter/metabolismo , Helicobacter pylori/patogenicidade , Bombas de Próton/metabolismo , Adulto , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Mucosa Gástrica/microbiologia , Genes Bacterianos , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Humanos , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Técnicas de Cultura de Tecidos , Células Tumorais Cultivadas , Virulência/genética , Adulto Jovem
14.
J Biol Chem ; 285(30): 23515-26, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20507990

RESUMO

Fibronectin, a 250-kDa eukaryotic extracellular matrix protein containing an RGD motif plays crucial roles in cell-cell communication, development, tissue homeostasis, and disease development. The highly complex fibrillar fibronectin meshwork orchestrates the functions of other extracellular matrix proteins, promoting cell adhesion, migration, and intracellular signaling. Here, we demonstrate that CagL, a 26-kDa protein of the gastric pathogen and type I carcinogen Helicobacter pylori, mimics fibronectin in various cellular functions. Like fibronectin, CagL contains a RGD motif and is located on the surface of the bacterial type IV secretion pili as previously shown. CagL binds to the integrin receptor alpha(5)beta(1) and mediates the injection of virulence factors into host target cells. We show that purified CagL alone can directly trigger intracellular signaling pathways upon contact with mammalian cells and can complement the spreading defect of fibronectin(-/-) knock-out cells in vitro. During interaction with various human and mouse cell lines, CagL mimics fibronectin in triggering cell spreading, focal adhesion formation, and activation of several tyrosine kinases in an RGD-dependent manner. Among the activated factors are the nonreceptor tyrosine kinases focal adhesion kinase and Src but also the epidermal growth factor receptor and epidermal growth factor receptor family member Her3/ErbB3. Interestingly, fibronectin activates a similar range of tyrosine kinases but not Her3/ErbB3. These findings suggest that the bacterial protein CagL not only exhibits functional mimicry with fibronectin but is also capable of activating fibronectin-independent signaling events. We thus postulate that CagL may contribute directly to H. pylori pathogenesis by promoting aberrant signaling cross-talk within host cells.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia , Forma Celular/efeitos dos fármacos , Fibronectinas/metabolismo , Helicobacter pylori , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibronectinas/química , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/metabolismo , Proteínas Imobilizadas/farmacologia , Integrinas/metabolismo , Camundongos , Oligopeptídeos/metabolismo , Fosfotirosina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Gastroenterology ; 139(1): 239-48, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20303353

RESUMO

BACKGROUND & AIMS: Infection with Helicobacter pylori represses expression of the gastric H, K-adenosine triphosphatase alpha-subunit (HKalpha), which could contribute to transient hypochlorhydria. CagL, a pilus protein component of the H pylori type IV secretion system, binds to the integrin alpha(5)beta1 to mediate translocation of virulence factors into the host cell and initiate signaling. alpha(5)beta1 binds a disintegrin and metalloprotease (ADAM) 17, a metalloenzyme that catalyzes ectodomain shedding of receptor tyrosine kinase ligands. We investigated whether H pylori-induced repression of HKalpha is mediated by CagL activation of ADAM17 and release of heparin-binding epidermal growth factor (HB-EGF). METHODS: HKalpha promoter and ADAM17 activity were measured in AGS gastric epithelial cells transfected with HKalpha promoter-reporter constructs or ADAM17-specific small interfering RNAs and infected with H pylori. HB-EGF secretion was measured by enzyme-linked immunosorbent assay analysis, and ADAM17 interaction with integrins was investigated by coimmunoprecipitation analyses. RESULTS: Infection of AGS cells with wild-type H pylori or an H pylori cagL-deficient isogenic mutant that also contained a wild-type version of cagL (P12DeltacagL/cagL) repressed HKalpha promoter-Luc reporter activity and stimulated ADAM17 activity. Both responses were inhibited by point mutations in the nuclear factor-kappaB binding site of HKalpha or by infection with P12DeltacagL. Small interfering RNA-mediated silencing of ADAM17 in AGS cells inhibited the repression of wild-type HKalpha promoter and reduced ADAM17 activity and HB-EGF production, compared to controls. Coimmunoprecipitation studies of AGS lysates showed that wild-type H pylori disrupted ADAM17-alpha5beta1 complexes. CONCLUSIONS: During acute H pylori infection, CagL dissociates ADAM17 from the integrin alpha(5)beta1 and activates ADAM17-dependent, nuclear factor-kappaB-mediated repression of HKalpha. This might contribute to transient hypochlorhydria in patients with H pylori infection.


Assuntos
Proteínas ADAM/fisiologia , Helicobacter pylori/fisiologia , ATPase Trocadora de Sódio-Potássio/genética , Estômago/enzimologia , Proteína ADAM17 , Repressão Enzimática , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Integrina beta1/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Oligopeptídeos/fisiologia , Regiões Promotoras Genéticas , ATPase Trocadora de Sódio-Potássio/biossíntese
16.
J Physiol ; 587(Pt 15): 3955-65, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19491250

RESUMO

Parietal cell (PC) proton secretion via H(+)/K(+)-ATPase requires apical K(+) recycling. A variety of K(+) channels and transporters are expressed in the PC and the molecular nature of the apical K(+) recycling channel is under debate. This study was undertaken to delineate the exact function of KCNQ1 channels in gastric acid secretion. Acid secretory rates and electrophysiological parameters were determined in gastric mucosae of 7- to 8-day-old KCNQ1(+/+), (+/-) and (-/-) mice. Parietal cell ultrastructure, abundance and gene expression levels were quantified. Glandular structure and PC abundance, and housekeeping gene expression did not differ between the KCNQ1(-/-) and (+/+) mucosae. Microvillar secretory membranes were intact, but basal acid secretion was absent and forskolin-stimulated acid output reduced by approximately 90% in KCNQ1(-/-) gastric mucosa. Application of a high K(+) concentration to the luminal membrane restored normal acid secretory rates in the KCNQ1(-/-) mucosa. The study demonstrates that the KCNQ1 channel provides K(+) to the extracellular K(+) binding site of the H(+)/K(+)-ATPase during acid secretion, and no other gastric K(+) channel can substitute for this function.


Assuntos
Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Canal de Potássio KCNQ1/metabolismo , Animais , Proteínas de Transporte de Ânions/metabolismo , Antiporters/metabolismo , Peso Corporal/fisiologia , Colforsina/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Canal de Potássio KCNQ1/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteínas SLC4A , Canal Kir5.1
17.
Am J Physiol Gastrointest Liver Physiol ; 295(5): G977-86, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18772363

RESUMO

Helicobacter pylori infection of the gastric body induces transient hypochlorhydria and contributes to mucosal progression toward gastric carcinoma. Acid secretion is mediated by parietal cell H,K-ATPase, in which the catalytic alpha-subunit (HKalpha) promoter activity in transfected gastric epithelial [gastric adenocarcinoma (AGS)] cells is repressed by H. pylori through NF-kappaB p50 homodimer binding to the promoter. IL-1beta, an acid secretory inhibitor whose mucosal level is increased by H. pylori, upregulates HKalpha promoter activity in AGS cells. Because IL-1beta also activates NF-kappaB signaling, we investigated disparate HKalpha regulation by H. pylori and IL-1beta, testing the hypothesis that IL-1beta-induced HKalpha promoter activation is mediated by the transcription factor Sp1. DNase I footprinting revealed Sp1 binding to the HKalpha promoter at -56 to -39 bp. IL-1beta stimulated the activity of three HKalpha promoter constructs containing NF-kappaB and Sp1 sites transfected into AGS cells and also stimulated a construct containing only an Sp1 site. This stimulation was abrogated by mutating the HKalpha promoter Sp1 binding site. Gelshift assays showed that IL-1beta increased Sp1 but not p50 binding to cognate HKalpha probes and that Sp1 also interacts with an HKalpha NF-kappaB site when bound to its cognate HKalpha cis-response element. H. pylori did not augment Sp1 binding to an HKalpha Sp1 probe, and small interfering RNA-mediated knockdown of Sp1 expression abrogated IL-1beta-induced HKalpha promoter stimulation. We conclude that IL-1beta upregulates HKalpha gene transcription by inducing Sp1 binding to HKalpha Sp1 and NF-kappaB sites and that the H. pylori perturbation of HKalpha gene expression is independent of Sp1-mediated basal HKalpha transcription.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Helicobacter pylori/fisiologia , Interleucina-1beta/metabolismo , Fator de Transcrição Sp1/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/genética , Humanos , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Ligação Proteica , Transdução de Sinais
18.
Am J Physiol Gastrointest Liver Physiol ; 294(3): G795-807, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18202112

RESUMO

Infection of human gastric body mucosa by the gram-negative, microaerophilic bacterium Helicobacter pylori induces an inflammatory response and a transitory hypochlorhydria that progresses in approximately 2% of patients to atrophic gastritis, dysplasia, and gastric adenocarcinoma. We have previously shown that H. pylori infection of cultured gastric epithelial cells (AGS) represses the activity of the transfected alpha-subunit (HKalpha) promoter of H,K-ATPase, the parietal cell enzyme mediating acid secretion. However, the mechanistic details of H. pylori-mediated repression of HKalpha and ensuing hypochlorhydria are unknown. H. pylori is known to upregulate the transcription factor NF-kappaB through the ERK1/2 MAPK pathway. We identified NF-kappaB-binding regions in the HKalpha promoter and found that H. pylori inoculation of AGS cells increased NF-kappaB p50 binding to the transfected HKalpha promoter and repressed its transcriptional activity. Immunoblot and DNA-protein interaction studies showed that although active phosphorylated NF-kappaB p65 is present in H. pylori-infected AGS cells, an NF-kappaB p50/p65 heterodimeric complex fails to bind to the HKalpha promoter. Point mutations at -159 and -161 bp in the HKalpha promoter NF-kappaB binding sequence prevented binding of NF-kappaB p50 and prevented H. pylori repression of point-mutated HKalpha promoter activity in transfected AGS cells. Small interfering RNA-mediated knockdown of NF-kappaB p50 in H. pylori-infected AGS cells also abrogated H. pylori-induced HKalpha repression, whereas NF-kappaB p65 knockdown did not. We conclude that H. pylori inhibits HKalpha gene expression by ERK1/2-mediated NF-kappaB p50 homodimer binding to the HKalpha promoter. This study identifies a novel pathogen-dependent mechanism of H,K-ATPase inhibition and contributes to understanding of H. pylori pathophysiology.


Assuntos
Infecções por Helicobacter/enzimologia , Infecções por Helicobacter/genética , Helicobacter pylori , Subunidade p50 de NF-kappa B/genética , ATPase Trocadora de Sódio-Potássio/genética , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Desoxirribonuclease I/genética , Desoxirribonuclease I/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Indicadores e Reagentes , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Dados de Sequência Molecular , Plasmídeos/genética , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Neoplasias Gástricas/patologia , Transcrição Gênica , Transfecção
19.
Am J Physiol Gastrointest Liver Physiol ; 292(4): G1055-61, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17204545

RESUMO

Helicobacter pylori infection of the human gastric body induces hypochlorhydria by perturbing acid secretion. H. pylori inhibits parietal cell H,K-ATPase alpha-subunit (HKalpha) gene and protein expression, providing a mechanistic basis for clinical hypochlorhydria. Given that H. pylori infection increases gastric mucosal IL-1beta, an acid secretory inhibitor, we investigated the role of IL-1beta in H. pylori-mediated inhibition of HKalpha transcription. Human gastric adenocarcinoma (AGS) cells were transfected with promoter-reporter constructs containing human HKalpha 5'-flanking sequence deletions. IL-1beta (10 ng/ml) had no effect on the transcriptional activity of six progressively shorter deletion constructs of the HKalpha promoter (HKalpha2179-HKalpha340) and significantly stimulated the activity of HKalpha206, HKalpha177, HKalpha165, and HKalpha102 deletion constructs (80%, 100%, 46%, and 35%, respectively). H. pylori inhibited the transcriptional activity of HKalpha2179, HKalpha206, HKalpha177, and HKalpha165; IL-1beta relieved the H. pylori inhibition of HKalpha2179 and HKalpha206 activity but not HKalpha177 and HKalpha165 activity. AGS cell pretreatment with a MEK1/2 inhibitor prevented the IL-1beta-mediated stimulation, but p38 and JNK pathway inhibitors did not. IL-1beta mRNA levels in AGS cells were low and unaffected by H. pylori, and ELISAs of H. pylori-conditioned AGS culture media showed no measurable IL-1beta secretion. These data indicate that an IL-1beta-dependent cis-response element lies downstream of -206 nt in the HKalpha promoter and that IL-1beta-mediated upregulation of HKalpha transcription is affected by an ERK1/2 kinase signal pathway. We conclude that an IL-1beta-responsive HKalpha cis element positively regulates HKalpha gene transcription in shortened deletion constructs and that H. pylori-induced inhibition of HKalpha transcription is not mediated by IL-1beta.


Assuntos
Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/biossíntese , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Interleucina-1beta/metabolismo , Transcrição Gênica , Região 5'-Flanqueadora , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Genes Reporter , ATPase Trocadora de Hidrogênio-Potássio/genética , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Humanos , Interleucina-1beta/farmacologia , Luciferases , Sistema de Sinalização das MAP Quinases , Inibidores de Proteínas Quinases/farmacologia , Subunidades Proteicas , RNA Mensageiro/biossíntese , Neoplasias Gástricas/patologia , Fatores de Tempo , Transfecção , Regulação para Cima
20.
Am J Physiol Gastrointest Liver Physiol ; 292(5): G1249-62, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17255364

RESUMO

Gastric parietal cells possess an amplified apical membrane recycling system dedicated to regulated apical recycling of H-K-ATPase. While amplified in parietal cells, apical recycling is critical to polarized secretory processes in most epithelial cells. To clarify putative regulators of apical recycling, we prepared immunoisolated parietal cell H-K-ATPase-containing recycling membranes from human stomachs and analyzed protein contents by tryptic digestion and mass spectrometry. We identified and validated by Western blots many of the proteins previously identified on immunoisolated rabbit tubulovesicles, including Rab11, Rab25, syntaxin 3, secretory carrier membrane proteins (SCAMPs), and vesicle-associated membrane protein (VAMP)2. In addition, we detected several previously unrecognized proteins, including Rab10, VAMP8, syntaxin 7, and syntaxin 12/13. We also identified the K(+) channel component KCNQ1. Immunostaining of human gastric mucosal sections confirmed the presence of each of these proteins in parietal cells and their colocalization with H-K-ATPase on tubulovesicles. To investigate the role of the identified soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins in apical recycling, we transfected them as DsRed2 fusions into an enhanced green fluorescent protein (EGFP)-Rab11a-expressing Madin-Darby canine kidney (MDCK) cell line. Syntaxin 12/13 and VAMP8 caused a collapse of the EGFP-Rab11a compartment, whereas a less dramatic effect was observed in cells transfected with syntaxin 3, syntaxin 7, or VAMP2. The five DsRed2-SNARE chimeras were also transfected into a MDCK cell line overexpressing Rab11-FIP2(129-512). All five of the chimeras were drawn into the collapsed apical recycling system. This study, which represents the first proteomic analysis of an immunoisolated vesicle population from native human tissue, demonstrates the diversity of putative regulators of the apical recycling system.


Assuntos
ATPase Trocadora de Hidrogênio-Potássio/fisiologia , Células Parietais Gástricas/fisiologia , Adulto , Animais , Western Blotting , Células Cultivadas , Cromatografia Líquida , Cães , Feminino , ATPase Trocadora de Hidrogênio-Potássio/análise , Humanos , Masculino , Pessoa de Meia-Idade , Células Parietais Gástricas/ultraestrutura , Proteoma/análise , Proteínas SNARE/fisiologia , Sistemas do Segundo Mensageiro , Espectrometria de Massas em Tandem , Proteínas rab de Ligação ao GTP/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...