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










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 13(9): e1006589, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28953979

RESUMEN

The ability of intracellular pathogens to manipulate host-cell viability is critical to successful infection. Some pathogens promote host-cell survival to protect their replicative niche, whereas others trigger host-cell death to facilitate release and dissemination of the pathogen after intracellular replication has occurred. We previously showed that the intracellular fungal pathogen Histoplasma capsulatum (Hc) uses the secreted protein Cbp1 to actively induce apoptosis in macrophages; interestingly, cbp1 mutant strains are unable to kill macrophages and display severely reduced virulence in the mouse model of Hc infection. To elucidate the mechanism of Cbp1-induced host-cell death, we performed a comprehensive alanine scanning mutagenesis and identified all amino acid residues that are required for Cbp1 to trigger macrophage lysis. Here we demonstrate that Hc strains expressing lytic CBP1 alleles activate the integrated stress response (ISR) in infected macrophages, as indicated by an increase in eIF2α phosphorylation as well as induction of the transcription factor CHOP and the pseudokinase Tribbles 3 (TRIB3). In contrast, strains bearing a non-lytic allele of CBP1 fail to activate the ISR, whereas a partially lytic CBP1 allele triggers intermediate levels of activation. We further show that macrophages deficient for CHOP or TRIB3 are partially resistant to lysis during Hc infection, indicating that the ISR is critical for susceptibility to Hc-mediated cell death. Moreover, we show that CHOP-dependent macrophage lysis is critical for efficient spread of Hc infection to other macrophages. Notably, CHOP knockout mice display reduced macrophage apoptosis and diminished fungal burden and are markedly resistant to Hc infection. Together, these data indicate that Cbp1 is required for Hc to induce the ISR and mediate a CHOP-dependent virulence pathway in the host.


Asunto(s)
Apoptosis/inmunología , Genes Fúngicos/genética , Histoplasma/metabolismo , Histoplasmosis/microbiología , Macrófagos/metabolismo , Factor de Transcripción CHOP/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Femenino , Interacciones Huésped-Patógeno/inmunología , Macrófagos/microbiología , Ratones , Virulencia/genética
2.
PLoS Pathog ; 12(7): e1005749, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27459510

RESUMEN

Innate immune cells shape the host response to microbial pathogens. Here we elucidate critical differences in the molecular response of macrophages vs. dendritic cells (DCs) to Histoplasma capsulatum, an intracellular fungal pathogen of humans. It has long been known that macrophages are permissive for Histoplasma growth and succumb to infection, whereas DCs restrict fungal growth and survive infection. We used murine macrophages and DCs to identify host pathways that influence fungal proliferation and host-cell viability. Transcriptional profiling experiments revealed that DCs produced a strong Type I interferon (IFN-I) response to infection with Histoplasma yeasts. Toll-like receptors 7 and 9 (TLR7/9), which recognize nucleic acids, were required for IFN-I production and restriction of fungal growth in DCs, but mutation of TLR7/9 had no effect on the outcome of macrophage infection. Moreover, TLR7/9 were essential for the ability of infected DCs to elicit production of the critical cytokine IFNγ from primed CD4+ T cells in vitro, indicating the role of this pathway in T cell activation. In a mouse model of infection, TLR7/9 were required for optimal production of IFN-I and IFNγ, host survival, and restriction of cerebral fungal burden. These data demonstrate the critical role of this pathway in eliciting an appropriate adaptive immune response in the host. Finally, although other fungal pathogens have been shown to elicit IFN-I in mouse models, the specific host cell responsible for producing IFN-I has not been elucidated. We found that CD103+ conventional DCs were the major producer of IFN-I in the lungs of wild-type mice infected with Histoplasma. Mice deficient in this DC subtype displayed reduced IFN-I production in vivo. These data reveal a previously unknown role for CD103+ conventional DCs and uncover the pivotal function of these cells in modulating the host immune response to endemic fungi.


Asunto(s)
Células Dendríticas/inmunología , Histoplasmosis/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/inmunología , Inmunidad Adaptativa/inmunología , Animales , Antígenos CD/inmunología , Técnicas de Cocultivo , Citocinas/biosíntesis , Citocinas/inmunología , Células Dendríticas/parasitología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Histoplasma/inmunología , Humanos , Cadenas alfa de Integrinas/inmunología , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/parasitología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos
3.
Infect Immun ; 81(2): 411-20, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23184522

RESUMEN

Histoplasma capsulatum is a fungal respiratory pathogen that survives and replicates within the phagolysosome of macrophages. The molecular factors it utilizes to subvert macrophage antimicrobial defenses are largely unknown. Although the ability of H. capsulatum to prevent acidification of the macrophage phagolysosome is thought to be critical for intracellular survival, this hypothesis has not been tested since H. capsulatum mutants that experience decreased phagosomal pH have not been identified. In a screen to identify H. capsulatum genes required for lysis of bone marrow-derived macrophages (BMDMs), we identified an insertion mutation disrupting the H. capsulatum homolog of 3-hydroxy-methylglutaryl coenzyme A (HMG CoA) lyase (HCL1). In addition to its inability to lyse macrophages, the hcl1 mutant had a severe growth defect in BMDMs, indicating that HMG CoA lyase gene function is critical for macrophage colonization. In other organisms, HMG CoA lyase catalyzes the last step in the leucine catabolism pathway. In addition, both fungi and humans deficient in HMG CoA lyase accumulate acidic intermediates as a consequence of their inability to catabolize leucine. Consistent with observations in other organisms, the H. capsulatum hcl1 mutant was unable to grow on leucine as the major carbon source, caused acidification of its growth medium in vitro, and resided in an acidified vacuole within macrophages. Mice infected with the hcl1 mutant took significantly longer to succumb to infection than mice infected with the wild-type strain. Taken together, these data indicate the importance of Hcl1 function in H. capsulatum replication in the harsh growth environment of the macrophage phagosome.


Asunto(s)
Histoplasma/metabolismo , Histoplasmosis/metabolismo , Macrófagos/metabolismo , Oxo-Ácido-Liasas/metabolismo , Acetil-CoA C-Acetiltransferasa/deficiencia , Acetil-CoA C-Acetiltransferasa/genética , Acetil-CoA C-Acetiltransferasa/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Secuencia de Aminoácidos , Animales , Femenino , Histoplasma/genética , Histoplasma/patogenicidad , Histoplasmosis/genética , Histoplasmosis/microbiología , Humanos , Concentración de Iones de Hidrógeno , Leucina/genética , Leucina/metabolismo , Macrófagos/enzimología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutagénesis Insercional , Oxo-Ácido-Liasas/deficiencia , Oxo-Ácido-Liasas/genética , Fagosomas/genética , Fagosomas/metabolismo , Fagosomas/microbiología
4.
Proc Natl Acad Sci U S A ; 107(48): 20738-43, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21076035

RESUMEN

The human T-cell leukemia virus type 1 (HTLV-1) is the cause of adult T-cell leukemia/lymphoma as well as tropical spastic paraparesis/HTLV-1-associated myelopathy. HTLV-1 is transmitted to T cells through the virological synapse and by extracellular viral assemblies. Here, we uncovered an additional mechanism of virus transmission that is regulated by the HTLV-1-encoded p8 protein. We found that the p8 protein, known to anergize T cells, is also able to increase T-cell contact through lymphocyte function-associated antigen-1 clustering. In addition, p8 augments the number and length of cellular conduits among T cells and is transferred to neighboring T cells through these conduits. p8, by establishing a T-cell network, enhances the envelope-dependent transmission of HTLV-1. Thus, the ability of p8 to simultaneously anergize and cluster T cells, together with its induction of cellular conduits, secures virus propagation while avoiding the host's immune surveillance. This work identifies p8 as a viral target for the development of therapeutic strategies that may limit the expansion of infected cells in HTLV-1 carriers and decrease HTLV-1-associated morbidity.


Asunto(s)
Infecciones por HTLV-I/transmisión , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Linfocitos T/citología , Linfocitos T/virología , Proteínas Virales/metabolismo , Comunicación Celular , Virus Linfotrópico T Tipo 1 Humano/ultraestructura , Humanos , Células Jurkat , Cinética , Linfocitos T/ultraestructura
5.
Mol Aspects Med ; 31(5): 333-43, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20673780

RESUMEN

The non-structural proteins encoded by the orf-I, II, III, and IV genes of the human T-cell leukemia/lymphoma virus type 1 (HTLV-1) genome, are critical for the modulation of cellular gene expression and T-cell proliferation, the escape from cytotoxic T-cells and natural killer cells, and virus expression. In here, we review the main functions of the HTLV-1 orf-I products. The 12kDa product from orf-I (p12) is proteolytically cleaved within the endoplasmic reticulum (ER) to generate the 8kDa protein (p8). At the steady state, both proteins are expressed at similar levels in transfected T-cells. The p12 protein remains in the ER and cis-Golgi, whereas the p8 protein traffics to the cell surface and is recruited to the immunological synapse. The p12 and the p8 proteins have seemingly opposite effects on T-cells; the ER resident p12, modulates T-cell activation and proliferation, whereas p8 induces T-cell anergy. The p8 protein also increases the formation of cellular conduits, is transferred to neighboring T-cells, and increases virus transmission. The requirement for HTLV-1 infectivity of orf-I is demonstrated by the loss of virus infectivity in macaques exposed to an engineered virus, whereby expression of orf-I was ablated. Altogether the current knowledge demonstrates that the concerted activity of p8 and p12 is essential for the persistence of virus infected cells in the host.


Asunto(s)
Comunicación Celular , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Linfocitos T/citología , Linfocitos T/virología , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Sistemas de Lectura Abierta/genética , Linfocitos T/metabolismo
6.
J Immunol ; 182(8): 4776-83, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19342655

RESUMEN

The endoplasmic reticulum (ER) protein Bap31 associates with nascent class I MHC molecules. It appears to mediate the export of class I MHC molecules from the ER and may also be involved in their quality control. In this study, we use Förster resonance energy transfer and quantitative fluorescence imaging to show that in human, HeLa cells, Bap31 clusters with MHC class I (HLA-A2) molecules in the ER, and traffics via export vesicles to the ER/Golgi intermediate compartment. Förster resonance energy transfer between Bap31 and HLA-A2 and forward traffic increases when MHC class I molecules are loaded with a pulse of peptide. The increased forward traffic is blocked by overexpression of Bap29, a partner protein for Bap31, which localizes to the ER. Thus, in HeLa cells, Bap31 is involved in the exit of peptide-loaded MHC class I from the ER, and its function is regulated by its interaction with its homologue, Bap29.


Asunto(s)
Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Biomarcadores , Células HeLa , Humanos , Proteínas de la Membrana/genética , Microscopía Inmunoelectrónica , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...