Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
J Immunol ; 188(8): 3594-602, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22403439

RESUMEN

The proinflammatory cysteine protease caspase-1 is autocatalytically activated upon cytosolic sensing of a variety of pathogen-associated molecular patterns by Nod-like receptors. Active caspase-1 processes pro-IL-1ß and pro-IL-18 to generate the bioactive cytokines and to initiate pathogen-specific immune responses. Little information is available on caspase-1 and inflammasome activation during infection with the gastric bacterial pathogen Helicobacter pylori. In this study, we addressed a possible role for caspase-1 and its cytokine substrates in the spontaneous and vaccine-induced control of Helicobacter infection, as well as the development of gastritis and gastric cancer precursor lesions, using a variety of experimental infection, vaccine-induced protection, and gastric disease models. We show that caspase-1 is activated and IL-1ß and IL-18 are processed in vitro and in vivo as a consequence of Helicobacter infection. Caspase-1 activation and IL-1 signaling are absolutely required for the efficient control of Helicobacter infection in vaccinated mice. IL-1R(-/-) mice fail to develop protective immunity but are protected against Helicobacter-associated gastritis and gastric preneoplasia as a result of their inability to generate Helicobacter-specific Th1 and Th17 responses. In contrast, IL-18 is dispensable for vaccine-induced protective immunity but essential for preventing excessive T cell-driven immunopathology. IL-18(-/-) animals develop strongly accelerated pathology that is accompanied by unrestricted Th17 responses. In conclusion, we show in this study that the processing and release of a regulatory caspase-1 substrate, IL-18, counteracts the proinflammatory activities of another caspase-1 substrate, IL-1ß, thereby balancing control of the infection with the prevention of excessive gastric immunopathology.


Asunto(s)
Caspasa 1/inmunología , Gastritis/inmunología , Infecciones por Helicobacter/inmunología , Neoplasias Gástricas/prevención & control , Inmunidad Adaptativa/efectos de los fármacos , Animales , Vacunas Bacterianas , Caspasa 1/genética , Modelos Animales de Enfermedad , Gastritis/etiología , Gastritis/prevención & control , Regulación de la Expresión Génica , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Transducción de Señal , Estómago/inmunología , Estómago/microbiología , Estómago/patología , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/microbiología , Células TH1/inmunología , Células Th17/inmunología , Vacunación
2.
J Immunol ; 186(2): 878-90, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149607

RESUMEN

B cells regulate autoimmune pathologies and chronic inflammatory conditions such as autoimmune encephalomyelitis and inflammatory bowel disease. The potential counterregulatory role of B cells in balancing pathogen-specific immune responses and the associated immunopathology is less well understood owing to the lack of appropriate persistent infection models. In this paper, we show that B cells have the ability to negatively regulate adaptive immune responses to bacterial pathogens. Using mouse models of infection with Helicobacter felis, a close relative of the human gastrointestinal pathogen H. pylori, we found that B cells activated by Helicobacter TLR-2 ligands induce IL-10-producing CD4(+)CD25(+) T regulatory-1 (Tr-1)-like cells in vitro and in vivo. Tr-1 conversion depends on TCR signaling and a direct T-/B-interaction through CD40/CD40L and CD80/CD28. B cell-induced Tr-1 cells acquire suppressive activity in vitro and suppress excessive gastric Helicobacter-associated immunopathology in vivo. Adoptive cotransfer of MyD88-proficient B cells and Tr-1 cells restores a normal gastric mucosal architecture in MyD88(-/-) and IL-10(-/-) mice in a manner that depends on T cellular, but not B cellular, IL-10 production. Our findings describe a novel mechanism of B cell-dependent Tr-1 cell generation and function in a clinically relevant disease model. In conclusion, we demonstrate that the B cell/Tr-1 cell axis is essential for balancing the control of Helicobacter infection with the prevention of excessive Th1-driven gastric immunopathology, promoting gastric mucosal homeostasis on the one hand and facilitating Helicobacter persistence on the other.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Diferenciación Celular/inmunología , Infecciones por Helicobacter/inmunología , Activación de Linfocitos/inmunología , Lesiones Precancerosas/inmunología , Neoplasias Gástricas/inmunología , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 2/fisiología , Animales , Subgrupos de Linfocitos B/microbiología , Subgrupos de Linfocitos B/patología , Células Cultivadas , Infecciones por Helicobacter/patología , Infecciones por Helicobacter/prevención & control , Helicobacter felis/inmunología , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/fisiología , Lesiones Precancerosas/patología , Lesiones Precancerosas/prevención & control , Neoplasias Gástricas/patología , Neoplasias Gástricas/prevención & control , Linfocitos T Reguladores/microbiología , Linfocitos T Reguladores/patología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/metabolismo
3.
Gastroenterology ; 138(4): 1455-67, 1467.e1-4, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20026064

RESUMEN

BACKGROUND & AIMS: Persistent infection with the human pathogen Helicobacter pylori increases the risk of gastric cancer. In this study, we investigated the role of cyclooxygenase-2 (COX-2) and its main product, prostaglandin E(2) (PGE(2)), in the development of Helicobacter-induced gastritis and gastric cancer precursor lesions. METHODS: We utilized mouse models of Helicobacter-induced gastric preneoplasia and vaccine-induced protection to study the effects of COX-2 inhibition and PGE(2) treatment on the induction of Helicobacter-specific immune responses and gastric premalignant immunopathology. RESULTS: COX-2 and PGE(2) are up-regulated upon Helicobacter infection in cultured epithelial cells and in the gastric mucosa of infected mice. Inhibition of COX-2 activity with celecoxib significantly accelerated early preneoplasia; conversely, systemic administration of synthetic PGE(2) prevented development of premalignant pathology and completely reversed preexisting lesions by suppressing interferon-gamma production in the infected stomachs. The protective effect of PGE(2) was accompanied by increased Helicobacter colonization in all models. All in vivo effects were attributed to immunosuppressive effects of PGE(2) on CD4(+) T-helper 1 cells, which fail to migrate, proliferate, and secrete cytokines when exposed to PGE(2) in vitro and in vivo. T-cell inhibition was found to be due to silencing of interleukin-2 gene transcription, and could be overcome by supplementation with recombinant interleukin-2 in vitro and in vivo. CONCLUSIONS: COX-2-dependent production of PGE(2) has an important immunomodulatory role during Helicobacter infection, preventing excessive local immune responses and the associated immunopathology by inhibiting the effector functions of pathogenic T-helper 1 cells.


Asunto(s)
Dinoprostona/uso terapéutico , Infecciones por Helicobacter/complicaciones , Lesiones Precancerosas/prevención & control , Neoplasias Gástricas/prevención & control , Animales , Ciclooxigenasa 2/fisiología , Inhibidores de la Ciclooxigenasa 2/farmacología , Modelos Animales de Enfermedad , Mucosa Gástrica/patología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/patología , Interleucina-10/fisiología , Interleucina-2/farmacología , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
4.
J Immunol ; 182(11): 7085-101, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19454706

RESUMEN

Chronic infection with the bacterial pathogen Helicobacter pylori is a risk factor for the development of gastric cancer, yet remains asymptomatic in the majority of individuals. We report here that the C57BL/6 mouse model of experimental infection with the closely related Helicobacter felis recapitulates this wide range in host susceptibility. Although the majority of infected animals develop premalignant lesions such as gastric atrophy, compensatory epithelial hyperplasia, and intestinal metaplasia, a subset of mice is completely protected from preneoplasia. Protection is associated with a failure to mount an IFN-gamma response to the infection and with a concomitant high Helicobacter burden. Using a vaccine model as well as primary infection and adoptive transfer models, we demonstrate that IFN-gamma, secreted predominantly by CD4(+)CD25(-) effector T(H) cells, is essential for Helicobacter clearance, but at the same time mediates the formation of preneoplastic lesions. We further provide evidence that IFN-gamma triggers a common transcriptional program in murine gastric epithelial cells in vitro and in vivo and induces their preferential transformation to the hyperplastic phenotype. In summary, our data suggest a dual role for IFN-gamma in Helicobacter pathogenesis that could be the basis for the differential susceptibility to H. pylori-induced gastric pathology in the human population.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Helicobacter/inmunología , Interferón gamma/inmunología , Neoplasias Gástricas/etiología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales , Infecciones por Helicobacter/complicaciones , Interferón gamma/metabolismo , Ratones , Riesgo , Neoplasias Gástricas/microbiología , Transcripción Genética , Vacunación
5.
Eur J Hum Genet ; 10(1): 77-81, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11896459

RESUMEN

Rett syndrome is a severe neurodevelopmental disorder that arises from mutations in the X-linked MECP2 gene. It is almost exclusively seen in girls due to the predominant occurrence of the mutations on the paternal X-chromosome, and also the early postnatal lethal effect of the disease causing mutations in hemizygous boys. We identified a boy with features of classic Rett syndrome who is mosaic for the truncating MECP2 mutation R270X. Chromosome analysis showed normal karyotype. These results indicate that a MECP2 mutation associated with Rett syndrome in females could lead to a similar phenotype in males as a result of somatic mosaicism.


Asunto(s)
Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN/genética , Mosaicismo/genética , Proteínas Represoras , Síndrome de Rett/genética , Niño , Preescolar , Codón sin Sentido , Humanos , Lactante , Recién Nacido , Masculino , Proteína 2 de Unión a Metil-CpG
6.
Discov Med ; 8(41): 68-73, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19788871

RESUMEN

Chronic infection with the bacterial pathogen Helicobacter pylori is closely linked to the development of gastric cancer. Experimental infection of the laboratory mouse strain C57Bl6 mimics the initiation and progression of the disease in humans. Using this model, we have identified a dual role for CD4+ IFN-gamma-secreting T-cells in the control of Helicobacter infection as well as in the induction of preneoplastic gastric pathology. High gastric expression of IFN-gamma was positively correlated with a low Helicobacter burden, and was essential for vaccine-induced protection; on the other hand, elevated levels of the cytokine also, either directly or indirectly, triggered the transformation of the normal gastric mucosa to atrophic, hyperplastic and metaplastic lesions. Based on similar patterns of gene expression changes induced by IFN-gamma in vivo and in cultured gastric epithelial cells, we hypothesize that IFN-gamma may act directly on epithelial cells to stimulate their hyperproliferation, and thus to predispose them to elevated mutation rates and an increased risk of malignant transformation.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Animales , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Mucosa Gástrica/citología , Mucosa Gástrica/efectos de los fármacos , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/inmunología , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interferón gamma/fisiología , Ratones , Ratones Endogámicos C57BL , Neoplasias Gástricas/etiología , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA