Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gastroenterology ; 141(2): 553-64, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21704622

RESUMO

BACKGROUND & AIMS: Colonization of gastric mucosa by Helicobacter pylori leads to epithelial hyperproliferation, which increases the risk for gastric adenocarcinoma. One H pylori virulence locus associated with cancer risk, cag, encodes a secretion system that transports effectors into host cells and leads to aberrant activation of ß-catenin and p120-catenin (p120). Peroxisome proliferator-activated receptor (PPAR)δ is a ligand-activated transcription factor that affects oncogenesis in conjunction with ß-catenin. We used a carcinogenic H pylori strain to define the role of microbial virulence constituents and PPARδ in regulating epithelial responses that mediate development of adenocarcinoma. METHODS: Gastric epithelial cells or colonies were co-cultured with the H pylori cag(+) strain 7.13 or cagE(-), cagA(-), soluble lytic transglycosylase(-), or cagA(-)/soluble lytic transglycosylase(-) mutants. Levels of PPARδ and cyclin E1 were determined by real-time, reverse-transcription polymerase chain reaction, immunoblot analysis, or immunofluorescence microscopy; proliferation was measured in 3-dimensional culture. PPARδ and Ki67 expression were determined by immunohistochemical analysis of human biopsies and rodent gastric mucosa. RESULTS: H pylori induced ß-catenin- and p120-dependent expression and activation of PPARδ in gastric epithelial cells, which were mediated by the cag secretion system substrates CagA and peptidoglycan. H pylori stimulated proliferation in vitro, which required PPARδ-mediated activation of cyclin E1; H pylori did not induce expression of cyclin E1 in a genetic model of PPARδ deficiency. PPARδ expression and proliferation in rodent and human gastric tissue was selectively induced by cag(+) strains and PPARδ levels normalized after eradication of H pylori. CONCLUSIONS: The H pylori cag secretion system activates ß-catenin, p120, and PPARδ, which promote gastric epithelial cell proliferation via activation of cyclin E1. PPARδ might contribute to gastric adenocarcinoma development in humans.


Assuntos
Adenocarcinoma/microbiologia , Células Epiteliais/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , PPAR delta/metabolismo , Neoplasias Gástricas/microbiologia , Adenocarcinoma/patologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cateninas/metabolismo , Proliferação de Células , Transformação Celular Neoplásica , Células Cultivadas , Ciclina E/metabolismo , Células Epiteliais/microbiologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gerbillinae , Helicobacter pylori/genética , Humanos , Antígeno Ki-67/metabolismo , Proteínas Oncogênicas/metabolismo , Transdução de Sinais , Neoplasias Gástricas/patologia , beta Catenina/metabolismo , delta Catenina
3.
Gastroenterology ; 139(4): 1333-43, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20547161

RESUMO

BACKGROUND & AIMS: Infection with the gastric mucosal pathogen Helicobacter pylori is the strongest identified risk factor for distal gastric cancer. These bacteria colonize a significant part of the world's population. We investigated the molecular mechanisms of p53 regulation in H pylori-infected cells. METHODS: Mongolian gerbils were challenged with H pylori and their gastric tissues were analyzed by immunohistochemistry and immunoblotting with p53 antibodies. Gastric epithelial cells were co-cultured with H pylori and the regulation of p53 was assessed by real-time polymerase chain reaction, immunoblotting, immunofluorescence, and cell survival assays. Short hairpin RNA and dominant-negative mutants were used to inhibit activities of Human Double Minute 2 (HDM2) and AKT1 proteins. RESULTS: We found that in addition to previously reported up-regulation of p53, H pylori can also negatively regulate p53 by increasing ubiquitination and proteasomal degradation via activation of the serine/threonine kinase AKT1, which phosphorylates and activates the ubiquitin ligase HDM2. These effects were mediated by the bacterial virulence factor CagA; ectopic expression of CagA in gastric epithelial cells increased phosphorylation of HDM2 along with the ubiquitination and proteasomal degradation of p53. The decrease in p53 levels increased survival of gastric epithelial cells that had sustained DNA damage. CONCLUSIONS: H pylori is able to inhibit the tumor suppressor p53. H pylori activates AKT1, resulting in phosphorylation and activation of HDM2 and subsequent degradation of p53 in gastric epithelial cells. H pylori-induced dysregulation of p53 is a potential mechanism by which the microorganism increases the risk of gastric cancer in infected individuals.


Assuntos
Mucosa Gástrica/microbiologia , Helicobacter pylori/patogenicidade , Proteína Supressora de Tumor p53/análise , Animais , Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Linhagem Celular Tumoral , Gerbillinae , Humanos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Neoplasias Gástricas/etiologia , Virulência
4.
Cancer Res ; 70(1): 30-5, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20048070

RESUMO

Helicobacter pylori-induced gastritis is the strongest singular risk factor for gastric adenocarcinoma. Matrix metalloproteinase-7 (MMP-7) is a proteolytic enzyme that can modify the intestinal microbial replicative niche as well as affect tumorigenesis, and H. pylori stimulates expression of MMP-7 in gastric epithelial cells in vitro. Utilizing a transgenic murine model of H. pylori-mediated injury, our experiments now show that gastric inflammation is increased within the context of MMP-7 deficiency, which involves both Th1- and Th17-mediated pathways. Enhanced gastritis in H. pylori-infected mmp-7-/- mice is strongly linked to accelerated epithelial cellular turnover. However, more severe inflammation and heightened proliferation and apoptosis are not dependent on MMP-7-mediated bacterial eradication. Collectively, these studies indicate that H. pylori-mediated induction of MMP-7 may serve to protect the gastric mucosa from pathophysiologic processes that promote carcinogenesis.


Assuntos
Gastrite/enzimologia , Infecções por Helicobacter/enzimologia , Metaloproteinase 7 da Matriz/metabolismo , Lesões Pré-Cancerosas/enzimologia , Adenocarcinoma/enzimologia , Adenocarcinoma/microbiologia , Animais , Mucosa Gástrica/enzimologia , Mucosa Gástrica/microbiologia , Gastrite/microbiologia , Helicobacter pylori , Imuno-Histoquímica , Metaloproteinase 7 da Matriz/genética , Camundongos , Camundongos Transgênicos , Lesões Pré-Cancerosas/microbiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/microbiologia
5.
Mol Biol Cell ; 19(10): 4110-21, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18653469

RESUMO

Helicobacter pylori is the strongest known risk factor for gastric adenocarcinoma, yet only a fraction of infected persons develop cancer. One H. pylori constituent that augments disease risk is the cytotoxin-associated gene (cag) pathogenicity island, which encodes a secretion system that translocates bacterial effector molecules into host cells. Matrix metalloproteinase (MMP)-7, a member of a family of enzymes with tumor-initiating properties, is overexpressed in premalignant and malignant gastric lesions, and H. pylori cag(+) strains selectively increase MMP-7 protein levels in gastric epithelial cells in vitro and in vivo. We now report that H. pylori-mediated mmp-7 induction is transcriptionally regulated via aberrant activation of p120-catenin (p120), a component of adherens junctions. H. pylori increases mmp-7 mRNA levels in a cag- and p120-dependent manner and induces translocation of p120 to the nucleus in vitro and in a novel ex vivo gastric gland culture system. Nuclear translocation of p120 in response to H. pylori relieves Kaiso-mediated transcriptional repression of mmp-7, which is implicated in tumorigenesis. These results indicate that selective and coordinated induction of mmp-7 expression by H. pylori cag(+) isolates may explain in part the augmentation in gastric cancer risk associated with these strains.


Assuntos
Moléculas de Adesão Celular/metabolismo , Regulação Bacteriana da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Fosfoproteínas/metabolismo , Neoplasias Gástricas/microbiologia , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Cateninas , Humanos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Neoplasias Gástricas/patologia , Frações Subcelulares , Transcrição Gênica , delta Catenina
6.
J Biol Chem ; 283(35): 23922-30, 2008 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-18579524

RESUMO

Chronic gastritis induced by Helicobacter pylori is the strongest known risk factor for peptic ulceration and distal gastric cancer, and adherence of H. pylori to gastric epithelial cells is critical for induction of inflammation. One H. pylori constituent that increases disease risk is the cag pathogenicity island, which encodes a secretion system that translocates bacterial effector molecules into host cells. Decay-accelerating factor (DAF) is a cellular receptor for H. pylori and a mediator of the inflammatory response to this pathogen. H. pylori induces DAF expression in human gastric epithelial cells; therefore, we sought to define the mechanism by which H. pylori up-regulates DAF and to extend these findings into a murine model of H. pylori-induced injury. Co-culture of MKN28 gastric epithelial cells with the wild-type H. pylori cag(+) strain J166 induced transcriptional expression of DAF, which was attenuated by disruption of a structural component of the cag secretion system (cagE). H. pylori-induced expression of DAF was dependent upon activation of the p38 mitogen-activated protein kinase pathway but not NF-kappaB. Hypergastrinemic INS-GAS mice infected with wild-type H. pylori demonstrated significantly increased DAF expression in gastric epithelium versus uninfected controls or mice infected with an H. pylori cagE(-) isogenic mutant strain. These results indicate that H. pylori cag(+) strains induce up-regulation of a cognate cellular receptor in vitro and in vivo in a cag-dependent manner, representing the first evidence of regulation of an H. pylori host receptor by the cag pathogenicity island.


Assuntos
Antígenos CD55/biossíntese , Mucosa Gástrica/metabolismo , Ilhas Genômicas , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Animais , Proteínas de Bactérias , Linhagem Celular , Técnicas de Cocultura , Mucosa Gástrica/microbiologia , Ilhas Genômicas/genética , Infecções por Helicobacter/genética , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Interações Hospedeiro-Patógeno/genética , Humanos , Camundongos , Mutação , Transcrição Gênica/genética , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...