Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
FEMS Immunol Med Microbiol ; 47(3): 414-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16872378

RESUMEN

Helicobacter pylori infection is characterized by infiltration of cells of the immune system, including dendritic cells, into the gastric mucosa. During chronic inflammation with Helicobacter pylori infection, a variety of cytokines are secreted into the mucosa, including interleukin-1beta (IL-1beta). The role of IL-1 in H. pylori infection was investigated using bone-marrow-derived dendritic cells from wild-type and IL-1 receptor-deficient (IL-1R-/-) mice. Dendritic cells were incubated with H. pylori at a multiplicity of infection of 10 and 100, and cytokine production evaluated. Helicobacter pylori SS1, H. pylori SD4, and an isogenic cagE mutant of SD4 stimulated IL-12, IL-6, IL-1beta, IL-10, and tumor necrosis factor-alpha at comparable levels in dendritic cells from both wild-type and IL-1R-/- mice. IL-10 production required the higher inoculum, while IL-12 was decreased at this bacterial load. Pretreatment of dendritic cells with an antibody to IL-10 resulted in an increased production of IL-12, confirming the down-regulation of IL-12 by IL-10. cagE was required for maximum stimulation of IL-12 by H. pylori. We speculate that the down-regulation of IL-12 by IL-10 at the higher multiplicity of infection represents the modulation of the host inflammatory response in vivo by H. pylori when the bacterial load is high, allowing for persistent colonization of the gastric mucosa.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Interleucina-10/inmunología , Interleucina-12/inmunología , Animales , Células de la Médula Ósea , Regulación hacia Abajo , Mucosa Gástrica/inmunología , Mucosa Gástrica/metabolismo , Interleucina-1/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-1/deficiencia , Factor de Necrosis Tumoral alfa/metabolismo
2.
Infect Immun ; 75(5): 2408-14, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17353291

RESUMEN

Helicobacter pylori is a gram-negative microaerophilic bacterium that colonizes the gastric mucosa, leading to disease conditions ranging from gastritis to cancer. Toll-like receptors (TLRs) play a central role in innate immunity by their recognition of conserved molecular patterns on bacteria, fungi, and viruses. Upon recognition of microbial components, these TLRs associate with several adaptor molecules, including myeloid differentiation factor 88 (MyD88). To investigate the contribution of the innate immune system to H. pylori infection, bone marrow-derived macrophages from mice deficient in TLR2, TLR4, TLR9, and MyD88 were infected with H. pylori SS1 and SD4 for 24 or 48 h. We demonstrate that MyD88 was essential for H. pylori induction of all cytokines investigated except alpha interferon (IFN-alpha). The secretion of IFN-alpha was substantially increased from cells deficient in MyD88. H. pylori induced interleukin-12 (IL-12) and IL-10 through TLR4/MyD88 signaling. In addition, H. pylori induced less IL-6 and IL-1beta in TLR2-deleted macrophages, suggesting that the MyD88 pathway activated by TLR2 stimulation is responsible for H. pylori induction of the host proinflammatory response (IL-6 and IL-1beta). These observations are important in light of a recent report on IL-6 and IL-1beta playing a role in the development of H. pylori-related gastric cancer. In conclusion, our study demonstrates that H. pylori activates TLR2 and TLR4, leading to the secretion of distinct cytokines by macrophages.


Asunto(s)
Citocinas/metabolismo , Helicobacter pylori/patogenicidad , Macrófagos/microbiología , Factor 88 de Diferenciación Mieloide , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Animales , Células de la Médula Ósea , Células Cultivadas , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
3.
Infect Immun ; 71(7): 4163-6, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12819109

RESUMEN

Dendritic cells are potent antigen-presenting cells that are present in the gastrointestinal tract and are required for the induction of a Th1 T-cell acquired immune response. Since infection with the gastric pathogen Helicobacter pylori elicits a Th1 cell response, the interaction of these organisms with dendritic cells should reflect the Th1 bias. We incubated H. pylori with cultured human dendritic cells and measured the cytokine induction profile, comparing the response to that induced by Salmonella enterica serovar Typhimurium. We found that H. pylori induced little interleukin 6 (IL-6) and essentially no IL-10 in contrast to S. enterica. However, H. pylori induced levels of IL-12 that were 30% of those induced by S. enterica, indicating a Th1 response. An isogenic cagE mutant of H. pylori lost about 50% of its IL-12-inducing ability, suggesting a role for the cag type IV secretion system in the stimulation of dendritic cells.


Asunto(s)
Células Dendríticas/metabolismo , Helicobacter pylori/patogenicidad , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Interleucina-6/biosíntesis , Proteínas Bacterianas/fisiología , Células Cultivadas , Humanos , Salmonella enterica/patogenicidad
4.
J Bacteriol ; 186(10): 3124-32, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15126474

RESUMEN

As all bacteria studied to date, the gastric pathogen Helicobacter pylori has an alternate lifestyle as a biofilm. H. pylori forms biofilms on glass surfaces at the air-liquid interface in stationary or shaking batch cultures. By light microscopy, we have observed attachment of individual, spiral H. pylori to glass surfaces, followed by division to form microcolonies, merging of individual microcolonies, and growth in the third dimension. Scanning electron micrographs showed H. pylori arranged in a matrix on the glass with channels for nutrient flow, typical of other bacterial biofilms. To understand the importance of biofilms to the H. pylori life cycle, we tested the effect of mucin on biofilm formation. Our results showed that 10% mucin greatly increased the number of planktonic H. pylori while not affecting biofilm bacteria, resulting in a decline in percent adherence to the glass. This suggests that in the mucus-rich stomach, H. pylori planktonic growth is favored over biofilm formation. We also investigated the effect of specific mutations in several genes, including the quorum-sensing gene, luxS, and the cagE type IV secretion gene. Both of these mutants were found to form biofilms approximately twofold more efficiently than the wild type in both assays. These results indicate the relative importance of these genes to the production of biofilms by H. pylori and the selective enhancement of planktonic growth in the presence of gastric mucin.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Helicobacter pylori/fisiología , Adhesión Bacteriana , Proteínas Bacterianas/fisiología , Liasas de Carbono-Azufre , Mucinas/farmacología
5.
Infect Immun ; 70(6): 3295-9, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12011029

RESUMEN

Gamma interferon (IFN-gamma) has been proposed to play an important role in Helicobacter-related gastritis. Using the IFN-gamma gene knockout (IFN-gamma(-/-)) mouse model and a murine gastric epithelial cell line, GSM06, we demonstrated that Helicobacter pylori maximally induced macrophage inflammatory protein-2 (MIP-2) and inducible nitric oxide synthase (iNOS) mRNA only in wild-type mice. MIP-2 and iNOS mRNA were also induced by H. pylori in GSM06 cells. Induction of cyclooxygenase 2 mRNA through IFN-gamma was demonstrated in GSM06 cells. These data indicate that IFN-gamma mediates the induction of MIP-2 and iNOS mRNA expression by H. pylori in mice.


Asunto(s)
Quimiocinas/genética , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Interferón gamma/inmunología , Isoenzimas/genética , Óxido Nítrico Sintasa/genética , Prostaglandina-Endoperóxido Sintasas/genética , Animales , Línea Celular , Quimiocina CXCL2 , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Modelos Animales de Enfermedad , Gastritis/genética , Gastritis/inmunología , Expresión Génica , Infecciones por Helicobacter/genética , Interferón gamma/genética , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II , ARN Mensajero , Estómago/citología
6.
Helicobacter ; 8(5): 495-502, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14535996

RESUMEN

BACKGROUND: Recent studies in both humans and animal models strongly suggest the contribution of the host immune response to Helicobacter pylori-related disease. Inducible nitric oxide synthase has been shown to be up-regulated in the gastric epithelium during H. pylori gastritis, suggesting a role in inflammation. MATERIALS AND METHODS: C57BL/6 wild-type and inducible nitric oxide synthase gene knockout mice were infected with H. pylori strain SS1. Expression of macrophage inflammatory protein-2 (MIP-2), interleukin-1 beta (IL-1 beta), Th1 (IL-2 and gamma interferon) and Th2 (IL-4 and IL-10) cytokines, and inducible cyclooxygenase mRNA in mice was determined in mouse gastric tissues and quantified using either competitive reverse transcription-polymerase chain reaction or competitive polymerase chain reaction following reverse transcription. RESULTS: The Th1 cytokine gamma interferon was only detected in wild-type and inducible nitric oxide synthase gene knockout infected mice, while a Th2 (IL-4) response was not detected. H. pylori induced MIP-2 and IL-1 beta mRNA in mice. CONCLUSIONS: Because similar levels of inflammatory mediators were noted in both wild-type and nitric oxide synthase gene knockout infected mice, our data suggest that inducible nitric oxide synthase does not influence expression of these inflammatory mediators in the early stages of H. pylori infection in mice.


Asunto(s)
Citocinas/biosíntesis , Infecciones por Helicobacter/enzimología , Infecciones por Helicobacter/inmunología , Helicobacter pylori , Óxido Nítrico Sintasa/metabolismo , Animales , Quimiocina CXCL2 , Quimiocinas/biosíntesis , Citocinas/genética , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Interferón gamma/biosíntesis , Interleucina-1/biosíntesis , Interleucina-10/biosíntesis , Interleucina-2/biosíntesis , Interleucina-4/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Células TH1/metabolismo , Células Th2/metabolismo
7.
Gastroenterology ; 125(6): 1613-25, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14724813

RESUMEN

BACKGROUND & AIMS: LL-37/human cationic antimicrobial peptide 18 (hCAP18) is a human cathelicidin with broad-spectrum antimicrobial, lipopolysaccharide binding, and chemotactic activities. This study examined the role of LL-37/hCAP18 in gastric innate immune defense by characterizing its constitutive and regulated expression by human gastric mucosa and its bactericidal activity against the gastric pathogen Helicobacter pylori. METHODS: LL-37/hCAP18 messenger RNA expression in normal and H. pylori -infected gastric mucosa and gastric epithelial cells was determined by in situ hybridization, real-time polymerase chain reaction, immunostaining, and immunoblot analysis. Bactericidal activity was measured by using a colony-forming unit assay. RESULTS: LL-37/hCAP18 messenger RNA and protein were expressed in a distinct distribution by surface epithelial cells as well as chief and parietal cells in the fundic glands of normal gastric mucosa. LL-37/hCAP18 was significantly increased in the epithelium and gastric secretions of H. pylori -infected patients, but not in individuals with non-H. pylori -induced gastric inflammation. Infection of cultured gastric epithelial cells with a wild-type but not an isogenic Delta cagE mutant strain of H. pylori increased LL-37/hCAP18 expression, indicating that H. pylori -induced regulation of LL-37/hCAP18 production required an intact type IV secretion system. LL-37, the C-terminal peptide of LL-37/hCAP18, alone or in synergy with human beta-defensin 1, was bactericidal for several H. pylori strains. CONCLUSIONS: These data indicate that H. pylori up-regulates production of LL-37/hCAP18 by gastric epithelium and suggest this cathelicidin contributes to determining the balance between host mucosal defense and H. pylori survival mechanisms that govern chronic infection with this gastric pathogen.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/biosíntesis , Mucosa Gástrica/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Adenocarcinoma/inmunología , Adenoma/inmunología , Catelicidinas , Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica , Humanos , Pólipos/inmunología , ARN Mensajero/análisis , Neoplasias Gástricas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA