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.
Inflamm Bowel Dis ; 21(12): 2778-86, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26332312

RESUMO

BACKGROUND: Autophagy is implicated in Crohn's disease (CD) pathogenesis. Recent evidence suggests autophagy regulates the microRNA (miRNA)-induced silencing complex (miRISC). Therefore, autophagy may play a novel role in CD by regulating expression of miRISC, thereby altering miRNA silencing. As microbes associated with CD can alter autophagy, we hypothesized that microbial disruption of autophagy affects the critical miRISC component AGO2. METHODS: AGO2 expression was assessed in epithelial and immune cells, and intestinal organoids with disrupted autophagy. Microarray technology was used to determine the expression of downstream miRNAs in cells with defective autophagy. RESULTS: Increased AGO2 was detected in autophagy-deficient ATG5-/- and ATG16-/- mouse embryonic fibroblast cells (MEFs) in comparison with wild-type MEFs. Chemical agents and VacA toxin, which disrupt autophagy, increased AGO2 expression in MEFs, epithelial cells lines, and human monocytes, respectively. Increased AGO2 was also detected in ATG7-/- intestinal organoids, in comparison with wild-type organoids. Five miRNAs were differentially expressed in autophagy-deficient MEFs. Pathway enrichment analysis of the differentially expressed miRNAs implicated signaling pathways previously associated with CD. CONCLUSIONS: Taken together, our results suggest that autophagy is involved in the regulation of the critical miRISC component AGO2 in epithelial and immune cells and primary intestinal epithelial cells. We propose a mechanism by which autophagy alters miRNA expression, which likely impacts the regulation of CD-associated pathways. Furthermore, as enteric microbial products can manipulate autophagy and AGO2, our findings suggest a novel mechanism by which enteric microbes could influence miRNA to promote disease.


Assuntos
Proteínas Argonautas/metabolismo , Autofagia/genética , Toxinas Bacterianas/metabolismo , MicroRNAs/metabolismo , Transdução de Sinais/genética , Animais , Proteínas Argonautas/genética , Proteína 5 Relacionada à Autofagia , Proteína 7 Relacionada à Autofagia , Proteínas de Transporte , Linhagem Celular , Doença de Crohn/genética , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , MicroRNAs/genética , Proteínas Associadas aos Microtúbulos/deficiência , Monócitos/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo
2.
Gastroenterology ; 142(5): 1160-71, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22333951

RESUMO

BACKGROUND & AIMS: The Helicobacter pylori toxin vacuolating cytotoxin (VacA) promotes gastric colonization, and its presence (VacA(+)) is associated with more-severe disease. The exact mechanisms by which VacA contributes to infection are unclear. We previously found that limited exposure to VacA induces autophagy of gastric cells, which eliminates the toxin; we investigated whether autophagy serves as a defense mechanism against H pylori infection. METHODS: We investigated the effect of VacA on autophagy in human gastric epithelial cells and primary gastric cells from mice. Expression of p62, a marker of autophagy, was also assessed in gastric tissues from patients infected with toxigenic (VacA(+)) or nontoxigenic strains. We analyzed the effect of VacA on autophagy in peripheral blood monocytes obtained from subjects with different genotypes of ATG16L1, which regulates autophagy. We performed genotyping for ATG16L1 in 2 cohorts of infected and uninfected subjects. RESULTS: Prolonged exposure of human gastric epithelial cells and mouse gastric cells to VacA disrupted induction of autophagy in response to the toxin, because the cells lacked cathepsin D in autophagosomes. Loss of autophagy resulted in the accumulation of p62 and reactive oxygen species. Gastric biopsy samples from patients infected with VacA(+), but not nontoxigenic strains of H pylori, had increased levels of p62. Peripheral blood monocytes isolated from individuals with polymorphisms in ATG16L1 that increase susceptibility to Crohn's disease had reduced induction of autophagy in response to VacA(+) compared to cells from individuals that did not have these polymorphisms. The presence of the ATG16L1 Crohn's disease risk variant increased susceptibility to H pylori infection in 2 separate cohorts. CONCLUSIONS: Autophagy protects against infection with H pylori; the toxin VacA disrupts autophagy to promote infection, which could contribute to inflammation and eventual carcinogenesis.


Assuntos
Autofagia/fisiologia , Proteínas de Bactérias/fisiologia , Infecções por Helicobacter/etiologia , Helicobacter pylori , Alelos , Animais , Proteínas de Bactérias/genética , Catepsina D/fisiologia , Doença de Crohn/etiologia , Doença de Crohn/genética , Genótipo , Humanos , Imunidade Inata , Camundongos , Fagossomos/fisiologia
3.
J Innate Immun ; 3(1): 83-98, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20962508

RESUMO

Citrobacter rodentium utilizes a type 3 secretion system (T3SS) to inject effector proteins into host intestinal epithelial cells, causing structural and functional changes in these cells during infection. Here, we examined the effects of C. rodentium infection on host cell protein ubiquitination in vivo. We observed the appearance of ubiquitinated protein (Ub(+)) aggregates in intestinal epithelial cells near the site of bacterial attachment. Formation of aggregates was dependent on T3SS activity and the effector translocated intimin receptor (Tir). Aggregates formed at 6 days after infection, when bacterial loads were maximal, but were absent at 12 days. Aggregates were not observed in MyD88(-/-) mice. Aggregate formation correlated with MyD88-dependent induction of NADPH oxidase 1, implicating reactive oxygen species in their formation. Aggregates were also observed in gastric tissues of mice infected with Helicobacter pylori. This is the first report describing the formation of Ub(+) aggregates in vivo during enteric infection, and reveals that this phenotype is dependent on both bacterial and host factors. Our experiments extend previous in vitro studies suggesting that Ub(+) aggregates play an important role in the initiation of immune responses to infection. Ub(+) aggregates are a novel marker of the cellular response to enteric pathogens and will be useful for studies of host-pathogen interactions in vivo.


Assuntos
Citrobacter rodentium/patogenicidade , Colo/metabolismo , Colo/microbiologia , Células Epiteliais/metabolismo , Interações Hospedeiro-Patógeno , Fator 88 de Diferenciação Mieloide/metabolismo , Proteínas Ubiquitinadas/metabolismo , Animais , Carga Bacteriana , Linhagem Celular , Colo/citologia , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/fisiopatologia , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidase 1
4.
Dig Dis Sci ; 50(12): 2300-6, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16416178

RESUMO

The aim of this study was to delineate the role of the Fas pathway in vaccination against Helicobacter pylori. C57BL/6 and Fas ligand-deficient (gld) mice were divided into 3 groups: control, H. pylori infected, and orally vaccinated (H. pylori whole cell sonicate and cholera toxin adjuvant). Oral vaccination prevented H. pylori colonization in 78% of C57BL/6 mice compared to only 18% of gld mice. Vaccination did not alter the degree of apoptosis in either strain of mice. Vaccination led to significant increase in interleukin (IL)-5 and IL-10 in C57BL/6 but not gld mice. H. pylori infection increased interferon (IFN)-gamma levels in C57BL/6 but not in gld mice while vaccination had no effect on IFN-gamma levels in either strain. Oral vaccination is not effective in Fas ligand-deficient mice likely owing to lack of effective cytokine responses. This indicates that the Fas pathway plays a critical role in promoting an appropriate effector response following H. pylori vaccination.


Assuntos
Anticorpos Antibacterianos/imunologia , Vacinas Bacterianas/farmacologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Interleucinas/imunologia , Glicoproteínas de Membrana/deficiência , Fatores de Necrose Tumoral/deficiência , Administração Oral , Animais , Apoptose/imunologia , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Proteína Ligante Fas , Infecções por Helicobacter/imunologia , Interleucinas/análise , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Valores de Referência , Medição de Risco , Sensibilidade e Especificidade , Falha de Tratamento , Fatores de Necrose Tumoral/imunologia , Vacinação/métodos
5.
J Immunol ; 171(4): 2035-41, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12902508

RESUMO

Helicobacter pylori is a bacterial pathogen evolved to chronically colonize the gastric epithelium, evade immune clearance by the host, and cause gastritis, peptic ulcers, and even gastric malignancies in some infected humans. In view of the known ability of this bacterium to manipulate gastric epithelial cell signal transduction cascades, we determined the effects of H. pylori infection on epithelial IL-4-Stat6 signal transduction. HEp-2 and MKN45 epithelial cells were infected with H. pylori strains LC11 or 8823 (type 1; cagA(+)/cagE(+)/VacA(+)), LC20 (type 2; cagA(-), cagE(-), VacA(-)), and cagA, cagE, and vacA isogenic mutants of strain 8823, with some cells receiving subsequent treatment with the Th2 cytokine IL-4, a known Stat6 activator. Immunofluorescence showed a disruption of Stat6-induced nuclear translocation by IL-4 in LC11-infected HEp-2 cells. IL-4-inducible Stat6 DNA binding in HEp-2 and MKN45 cells was abrogated by infection, but MKN45 cell viability was unaffected. A decrease in IL-4-mediated Stat6 tyrosine phosphorylation in nuclear and whole cell lysates was also observed following infection with strains LC11 and LC20, while neither strain altered IL-4 receptor chain alpha or Janus kinase 1 protein expression. Furthermore, parental strain 8823 and its isogenic cagA, cagE, and vacA mutants also suppressed IL-4-induced Stat6 tyrosine phosphorylation to comparable degrees. Thus, H. pylori did not directly activate Stat6, but blocked the IL-4-induced activation of epithelial Stat6. This may represent an evolutionarily conserved strategy to disrupt a Th2 response and evade the host immune system, allowing for successful chronic infection.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Helicobacter pylori/patogenicidade , Interleucina-4/antagonistas & inibidores , Transdução de Sinais/imunologia , Transativadores/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/imunologia , Morte Celular/imunologia , Linhagem Celular Transformada , Relação Dose-Resposta Imunológica , Regulação para Baixo/imunologia , Células Epiteliais/metabolismo , Helicobacter pylori/imunologia , Humanos , Interleucina-4/metabolismo , Interleucina-4/fisiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Janus Quinase 1 , Fosforilação , Subunidades Proteicas/biossíntese , Proteínas Tirosina Quinases/biossíntese , Receptores de Interleucina-4/biossíntese , Fator de Transcrição STAT6 , Transativadores/metabolismo , Transativadores/fisiologia , Células Tumorais Cultivadas , Tirosina/antagonistas & inibidores , Tirosina/metabolismo
6.
Infect Immun ; 70(5): 2591-7, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11953400

RESUMO

Recent evidence suggests that immune-mediated gastric epithelial cell apoptosis through Fas-Fas ligand interactions participates in Helicobacter pylori disease pathogenesis. To define the role of Fas signaling in vivo, H. pylori strain SS1 infection in C57BL/6 mice was compared to that in mice deficient in the Fas ligand (gld). gld mice had a degree of gastritis similar to that of C57BL/6 mice after 6 weeks (gastritis score, 5.2 +/- 0.6 [mean +/- standard error] versus 3.5 +/- 0.8) and 12 weeks (4.0 +/- 0.7 versus 3.4 +/- 0.5) of infection. Bacterial colonization was comparable in each group of mice at 12 weeks of infection (2.1 +/- 0.3 versus 1.6 +/- 0.3 for gld and C57BL/6, respectively; the difference is not significant). Sixty-seven percent of H. pylori-infected gld mice displayed atrophic changes in the gastric mucosa, compared with 37% of infected C57BL/6 mice, at 12 weeks. In addition, atrophic changes were more severe in H. pylori-infected gld mice (P < 0.05). Splenocytes isolated from H. pylori-infected C57BL/6 mice had a twofold increase in production of the Th1 cytokine gamma interferon (IFN-gamma) in response to H. pylori antigens at both 6 and 12 weeks compared to controls (143 +/- 65 versus 69 +/-26 pg/ml and 336 +/- 73 versus 172 +/- 60, respectively). In contrast, there was a lack of detectable IFN-gamma in gld mice infected with the bacterium. H. pylori-infected C57BL/6 mice had increased epithelial cell apoptosis compared with sham-infected C57BL/6 mice (35.0 +/- 8.9 versus 12.3 +/- 6.9; P < 0.05). Epithelial cell apoptosis did not differ between H. pylori-infected and control gld mice (5.2 +/- 1.6 versus 6.5 +/- 2.9 [not significant]). These data demonstrate that mice with mutations in the Fas ligand develop more severe premalignant mucosal changes in response to infection with H. pylori in association with both an impaired gastric epithelial cell apoptotic response and IFN-gamma production. The Fas death pathway modulates disease pathophysiology following murine infection with H. pylori. Deregulation of the Fas pathway could be involved in the transition from gastritis to gastric cancers during H. pylori infection.


Assuntos
Apoptose , Infecções por Helicobacter/imunologia , Helicobacter pylori , Receptor fas/fisiologia , Animais , Atrofia , Proteína Ligante Fas , Feminino , Mucosa Gástrica/patologia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/patologia , Interferon gama/biossíntese , Interleucina-5/biossíntese , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA