Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-22919664

RESUMO

MiR-155 regulates numerous aspects of innate and adaptive immune function. This miR is induced in response to Toll-like receptor ligands, cytokines, and microbial infection. We have previously shown that miR-155 is induced in monocytes/macrophages infected with Francisella tularensis and suppresses expression of the inositol phosphatase SHIP to enhance activation of the PI3K/Akt pathway, which in turn promotes favorable responses for the host. Here we examined how miR-155 expression is regulated during infection. First, our data demonstrate that miR-155 can be induced through soluble factors of bacterial origin and not the host. Second, miR-155 induction is not a direct effect of infection and it requires NF-κB signaling to up-regulate fos/jun transcription factors. Finally, we demonstrate that the requirement for NF-κB-dependent de novo protein synthesis is globally shared by microbial ligands and live bacteria. This study provides new insight into the complex regulation of miR-155 during microbial infection.


Assuntos
Francisella tularensis/imunologia , MicroRNAs/biossíntese , Monócitos/imunologia , NF-kappa B/metabolismo , Células Cultivadas , Humanos , Biossíntese de Proteínas
2.
J Immunol ; 187(2): 635-43, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21697459

RESUMO

The environmental bacterium Burkholderia cenocepacia causes opportunistic lung infections in immunocompromised individuals, particularly in patients with cystic fibrosis. Infections in these patients are associated with exacerbated inflammation leading to rapid decay of lung function, and in some cases resulting in cepacia syndrome, which is characterized by a fatal acute necrotizing pneumonia and sepsis. B. cenocepacia can survive intracellularly in macrophages by altering the maturation of the phagosome, but very little is known on macrophage responses to the intracellular infection. In this study, we have examined the role of the PI3K/Akt signaling pathway in B. cenocepacia-infected monocytes and macrophages. We show that PI3K/Akt activity was required for NF-κB activity and the secretion of proinflammatory cytokines during infection with B. cenocepacia. In contrast to previous observations in epithelial cells infected with other Gram-negative bacteria, Akt did not enhance IκB kinase or NF-κB p65 phosphorylation, but rather inhibited GSK3ß, a negative regulator of NF-κB transcriptional activity. This novel mechanism of modulation of NF-κB activity may provide a unique therapeutic target for controlling excessive inflammation upon B. cenocepacia infection.


Assuntos
Infecções por Burkholderia/imunologia , Burkholderia cenocepacia/imunologia , Quinase 3 da Glicogênio Sintase/fisiologia , Quinase I-kappa B/fisiologia , Mediadores da Inflamação/fisiologia , Macrófagos/imunologia , Monócitos/imunologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Infecções por Burkholderia/patologia , Regulação para Baixo/imunologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Monócitos/microbiologia , Monócitos/patologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação/imunologia , Transdução de Sinais/imunologia , Fator de Transcrição RelA/antagonistas & inibidores
3.
PLoS One ; 4(12): e8508, 2009 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-20041145

RESUMO

The intracellular gram-negative bacterium Francisella tularensis causes the disease tularemia and is known for its ability to subvert host immune responses. Previous work from our laboratory identified the PI3K/Akt pathway and SHIP as critical modulators of host resistance to Francisella. Here, we show that SHIP expression is strongly down-regulated in monocytes and macrophages following infection with F. tularensis novicida (F.n.). To account for this negative regulation we explored the possibility that microRNAs (miRs) that target SHIP may be induced during infection. There is one miR that is predicted to target SHIP, miR-155. We tested for induction and found that F.n. induced miR-155 both in primary monocytes/macrophages and in vivo. Using luciferase reporter assays we confirmed that miR-155 led to down-regulation of SHIP, showing that it specifically targets the SHIP 3'UTR. Further experiments showed that miR-155 and BIC, the gene that encodes miR-155, were induced as early as four hours post-infection in primary human monocytes. This expression was dependent on TLR2/MyD88 and did not require inflammasome activation. Importantly, miR-155 positively regulated pro-inflammatory cytokine release in human monocytes infected with Francisella. In sharp contrast, we found that the highly virulent type A SCHU S4 strain of Francisella tularensis (F.t.) led to a significantly lower miR-155 response than the less virulent F.n. Hence, F.n. induces miR-155 expression and leads to down-regulation of SHIP, resulting in enhanced pro-inflammatory responses. However, impaired miR-155 induction by SCHU S4 may help explain the lack of both SHIP down-regulation and pro-inflammatory response and may account for the virulence of Type A Francisella.


Assuntos
Citocinas/imunologia , Francisella tularensis/patogenicidade , Francisella/fisiologia , Mediadores da Inflamação/imunologia , MicroRNAs/genética , Monoéster Fosfórico Hidrolases/genética , Animais , Sequência de Bases , Caspase 1/metabolismo , Linhagem Celular , Citocinas/biossíntese , Regulação para Baixo/genética , Endocitose , Ativação Enzimática , Francisella/citologia , Infecções por Bactérias Gram-Negativas/enzimologia , Infecções por Bactérias Gram-Negativas/genética , Humanos , Mediadores da Inflamação/metabolismo , Inositol Polifosfato 5-Fosfatases , Camundongos , MicroRNAs/metabolismo , Viabilidade Microbiana , Modelos Biológicos , Dados de Sequência Molecular , Monoéster Fosfórico Hidrolases/metabolismo , Transdução de Sinais , Receptores Toll-Like , Virulência/genética
4.
PLoS One ; 4(11): e7919, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19936232

RESUMO

Francisella tularensis infects macrophages and escapes phago-lysosomal fusion to replicate within the host cytosol, resulting in host cell apoptosis. Here we show that the Fas-mediated death pathway is activated in infected cells and correlates with escape of the bacterium from the phagosome and the bacterial burden. Our studies also demonstrate that constitutive activation of Akt, or deletion of SHIP, promotes phago-lysosomal fusion and limits bacterial burden in the host cytosol, and the subsequent induction of Fas expression and cell death. Finally, we show that phagosomal escape/intracellular bacterial burden regulate activation of the transcription factors sp1/sp3, leading to Fas expression and cell death. These data identify for the first time host cell signaling pathways that regulate the phagosomal escape of Francisella, leading to the induction of Fas and subsequent host cell death.


Assuntos
Francisella/metabolismo , Fagossomos/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor fas/metabolismo , Animais , Células da Medula Óssea/citologia , Morte Celular , Linhagem Celular , Citosol/metabolismo , Inositol Polifosfato 5-Fosfatases , Lisossomos/metabolismo , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose
5.
Autophagy ; 5(1): 125-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19029814

RESUMO

The Gram-negative intracellular pathogen Francisella tularensis is known for its ability to dampen host immune responses. We recently performed a microarray analysis comparing human monocyte responses to the highly virulent F. tularensis tularensis Schu S4 strain (F.t.) versus the less virulent F. tularensis novicida (F.n.).(1) Many groups of genes were affected, including those involved with autophagy and with the regulation of autophagy. Here, we discuss the implications in the context of Francisella virulence and host cell response, then conclude with potential future experiments.


Assuntos
Autofagia , Francisella tularensis/fisiologia , Interações Hospedeiro-Patógeno , Monócitos/citologia , Monócitos/microbiologia , Transdução de Sinais , Autofagia/genética , Regulação da Expressão Gênica , Humanos , Modelos Biológicos , Monócitos/metabolismo
6.
Future Microbiol ; 3(5): 503-6, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18811234

RESUMO

Evaluation of: Ketavarapu JM, Rodriguez AR, Yu J et al.: Mast cells inhibit intramacrophage Francisella tularensis replication via contact and secreted products including IL-4. Proc. Natl Acad. Sci. USA 105(27), 9313-9318 (2008). The intracellular pathogen Francisella tularensis is a highly infectious organism that infects cells of the immune system. Mast cells have been known for their role in anaphylaxis, although they are also important for their ability to aid in the defense against pathogens. The report by Ketavarapu et al. has demonstrated that mast cells function to limit the replication of F. tularensis live vaccine strain within macrophages in vitro as well as in vivo. It was determined that IL-4 is one secreted mediator of this effect thus highlights a previously unknown mechanism of host defense against F. tularensis.

7.
PLoS One ; 3(8): e2924, 2008 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-18698339

RESUMO

Francisella tularensis is a gram-negative facultative bacterium that causes the disease tularemia, even upon exposure to low numbers of bacteria. One critical characteristic of Francisella is its ability to dampen or subvert the host immune response. In order to help understand the mechanisms by which this occurs, we performed Affymetrix microarray analysis on transcripts from blood monocytes infected with the virulent Type A Schu S4 strain. Results showed that expression of several host response genes were reduced such as those associated with interferon signaling, Toll-like receptor signaling, autophagy and phagocytosis. When compared to microarrays from monocytes infected with the less virulent F. tularensis subsp. novicida, we found qualitative differences and also a general pattern of quantitatively reduced pro-inflammatory signaling pathway genes in the Schu S4 strain. Notably, the PI3K/Akt1 pathway appeared specifically down-regulated following Schu S4 infection and a concomitantly lower cytokine response was observed. This study identifies several new factors potentially important in host cell subversion by the virulent Type A F. tularensis that may serve as novel targets for drug discovery.


Assuntos
Francisella tularensis/patogenicidade , Monócitos/microbiologia , Análise de Sequência com Séries de Oligonucleotídeos , Células Cultivadas , Regulação para Baixo , Citometria de Fluxo , Regulação da Expressão Gênica , Interações Hospedeiro-Parasita , Humanos , Interferon gama/genética , Interferons/genética , Monócitos/citologia , Monócitos/fisiologia , Reação em Cadeia da Polimerase , Receptores Toll-Like/genética , Transcrição Gênica
8.
Mol Immunol ; 45(12): 3428-37, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18514317

RESUMO

Tularemia is a zoonotic disease caused by the Gram-negative intracellular pathogen Francisella tularensis. These bacteria evade phagolysosomal fusion, escape from the phagosome and replicate in the host cell cytoplasm. IFNgamma has been shown to suppress the intra-macrophage growth of Francisella through both nitric oxide-dependent and -independent pathways. Since Francisella is known to subvert host immune responses, we hypothesized that this pathogen could interfere with IFNgamma signaling. Here, we report that infection with Francisella suppresses IFNgamma-induced STAT1 expression and phosphorylation in both human and murine mononuclear phagocytes. This suppressive effect of Francisella is independent of phagosomal escape or replication and is mediated by a heat-stable and constitutively expressed bacterial factor. An analysis of the molecular mechanism of STAT1 inhibition indicated that expression of SOCS3, an established negative regulator of IFNgamma signaling, is highly up-regulated during infection and suppresses STAT1 phosphorylation. Functional analyses revealed that this interference with IFNgamma signaling is accompanied by the suppression of IP-10 production and iNOS induction resulting in increased intracellular bacterial survival. Importantly, the suppressive effect on IFNgamma-mediated host cell protection is most effective when IFNgamma is added post infection, suggesting that the bacteria establish a permissive environment within the host cell.


Assuntos
Francisella tularensis/citologia , Interferon gama/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/microbiologia , Viabilidade Microbiana/imunologia , Fagócitos/imunologia , Fagócitos/microbiologia , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Francisella tularensis/efeitos dos fármacos , Humanos , Interferon gama/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/microbiologia , Camundongos , Viabilidade Microbiana/efeitos dos fármacos , Modelos Imunológicos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/microbiologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Fagócitos/efeitos dos fármacos , Fagossomos/efeitos dos fármacos , Fagossomos/microbiologia , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT1/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Tularemia/imunologia , Tularemia/microbiologia
9.
Mol Immunol ; 45(10): 3012-21, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18295889

RESUMO

Francisella tularensis is a highly infectious, Gram-negative intra-cellular pathogen that can cause the zoonotic disease tularemia. Although the receptors critical for internalization of Francisella by macrophages are beginning to be defined, the identity of the downstream signaling pathways essential for the engulfment are not yet identified. In this study we have tested the role of Syk in the phagocytosis of Francisella. We report that Syk is activated during Francisella infection and is critical for the uptake of the organisms. Pharmacologic inhibition of Syk almost completely abrogated uptake, whereas the overexpression of Syk significantly enhanced uptake. However, Syk appears to be dispensable during initial host-pathogen contact. Further analyses of the molecular mechanism of Syk influence on Francisella uptake revealed that the MAPK Erk but not the phosphatidylinositol 3 kinase (PI3K)/Akt pathway is the downstream effector of Syk. Thus, the inhibition of Erk in Syk-overexpressing cells or the inhibition of Syk in Erk-overexpressing cells led to a significant attenuation of uptake. Collectively, these data identify Syk and Erk as key players in the phagocytosis of Francisella.


Assuntos
Francisella/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fagocitose , Proteínas Tirosina Quinases/metabolismo , Animais , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Francisella/efeitos dos fármacos , Infecções por Bactérias Gram-Negativas/enzimologia , Infecções por Bactérias Gram-Negativas/microbiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/microbiologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Fagocitose/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinase Syk
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...