Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros




Base de datos
Asunto de la revista
Intervalo de año de publicación
1.
J Virol ; 88(14): 7703-14, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24760892

RESUMEN

Successful adaptive immunity to virus infection often depends on the initial innate response. Previously, we demonstrated that Junín virus, the etiological agent responsible for Argentine hemorrhagic fever (AHF), activates an early innate immune response via an interaction between the viral glycoprotein and Toll-like receptor 2 (TLR2). Here we show that TLR2/6 but not TLR1/2 heterodimers sense Junín virus glycoprotein and induce a cytokine response, which in turn upregulates the expression of the RNA helicases RIG-I and MDA5. NF-κB and Erk1/2 were important in the cytokine response, since both proteins were phosphorylated as a result of the interaction of virus with TLR2, and treatment with an Erk1/2-specific inhibitor blocked cytokine production. We show that the Junín virus glycoprotein activates cytokine production in a human macrophage cell line as well. Moreover, we show that TLR2-mediated immune response plays a role in viral clearance because wild-type mice cleared Candid 1 (JUNV C1), the vaccine strain of Junín virus, more rapidly than did TLR2 knockout mice. This clearance correlated with the generation of Junín virus-specific CD8(+) T cells. However, infected wild-type and TLR2 knockout mice developed TLR2-independent blocking antibody responses with similar kinetics. We also show that microglia and astrocytes but not neurons are susceptible to infection with JUNV C1. Although JUNV C1 infection of the brain also triggered a TLR2-dependent cytokine response, virus levels were equivalent in wild-type and TLR2 knockout mice. Importance: Junín virus is transmitted by rodents native to Argentina and is associated with both systemic disease and, in some patients, neurological symptoms. Humans become infected when they inhale aerosolized Junín virus. AHF has a 15 to 30% mortality rate, and patients who clear the infection develop a strong antibody response to Junín virus. Here we investigated what factors determine the immune response to Junín virus. We show that a strong initial innate immune response to JUNV C1 determines how quickly mice can clear systemic infection and that this depended on the cellular immune response. In contrast, induction of an innate immune response in the brain had no effect on virus infection levels. These findings may explain how the initial immune response to Junín virus infection could determine different outcomes in humans.


Asunto(s)
Inmunidad Adaptativa , Sangre/inmunología , Encéfalo/inmunología , Inmunidad Innata , Virus Junin/inmunología , Receptor Toll-Like 2/inmunología , Replicación Viral , Animales , Sangre/virología , Encéfalo/virología , Línea Celular , Citocinas/metabolismo , Humanos , Virus Junin/fisiología , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Sci Transl Med ; 5(204): 204ra131, 2013 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-24068738

RESUMEN

New World hemorrhagic fever arenavirus infection results in 15 to 30% mortality in humans. We performed a high-throughput small interfering RNA screen with Junín virus glycoprotein-pseudotyped viruses to find potential host therapeutic targets. Voltage-gated calcium channel (VGCC) subunits, for which there are Food and Drug Administration (FDA)-approved drugs, were identified in the screen. Knockdown of VGCC subunits or treatment with channel blockers diminished Junín virus-cell fusion and entry into cells and thereby decreased infection. Gabapentin, an FDA-approved drug used to treat neuropathic pain that targets the α2δ2 subunit, inhibited infection of mice by the Candid 1 vaccine strain of the virus. These findings demonstrate that VGCCs play a role in virus infection and have the potential to lead to therapeutic intervention of New World arenavirus infection.


Asunto(s)
Canales de Calcio/genética , Canales de Calcio/metabolismo , Ensayos Analíticos de Alto Rendimiento , Virus Junin/fisiología , Terapia Molecular Dirigida , ARN Interferente Pequeño/genética , Internalización del Virus , Aminas/farmacología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Fusión Celular , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Chlorocebus aethiops , Ácidos Ciclohexanocarboxílicos/farmacología , Gabapentina , Técnicas de Silenciamiento del Gen , Interacciones Huésped-Patógeno/genética , Humanos , Virus Junin/efectos de los fármacos , Ratones , Unión Proteica/efectos de los fármacos , Subunidades de Proteína/metabolismo , Interferencia de ARN , Receptores de Transferrina/metabolismo , Células Vero , Internalización del Virus/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología
3.
J Virol ; 85(21): 11058-68, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21880772

RESUMEN

Junín virus is the causative agent for Argentine hemorrhagic fever, and its natural host is the New World rodent Calomys musculinus. The virus is transmitted to humans by aerosolization, and it is believed that many of the clinical symptoms are caused by cytokines produced by sentinel cells of the immune system. Here we used the Junín virus vaccine strain Candid 1 to determine whether mouse cells could be used to study virus entry and antiviral innate immune responses. We show that Candid 1 can infect and propagate in different mouse-derived cell lines through a low-pH-dependent, transferrin receptor 1-independent mechanism, suggesting that there is a second entry receptor. In addition, Candid 1 induced expression of the antiviral cytokines tumor necrosis factor alpha and beta interferon in macrophages, and this induction was independent of viral replication. Using Candid 1, as well as virus-like particles bearing the viral glycoprotein, to infect different primary cells and established macrophage cell lines with deletions in the Toll-like receptor (TLR) pathway, we show that TLR2 is a cellular sensor of both the Parodi and Candid 1 viral glycoproteins. Because Junín virus is highly lethal in humans, the use of an experimentally tractable model system, such as the mouse, could provide a better understanding of the antiviral innate cellular responses to Junín virus and the role of these responses in pathogenesis.


Asunto(s)
Inmunidad Innata , Virus Junin/crecimiento & desarrollo , Virus Junin/inmunología , Animales , Células Cultivadas , Citocinas/biosíntesis , Expresión Génica , Humanos , Ratones , Receptores Virales/metabolismo , Receptor Toll-Like 2/inmunología , Internalización del Virus
4.
J Virol ; 84(15): 7803-14, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20504932

RESUMEN

The human cytomegalovirus (HCMV) virion protein pUL83 (also termed pp65) inhibits the expression of interferon-inducible cellular genes. In this work we demonstrate that pUL83 is also important for efficient induction of transcription from the viral major immediate-early promoter. Infection with a mutant virus containing a premature translation termination codon in the UL83 open reading frame (ORF) (UL83Stop) resulted in decreased transcription from the major immediate-early promoter in a time- and multiplicity-dependent manner. Expression of pUL83 alone is capable of transactivating the promoter in a reporter assay, and pUL83 associates with the promoter in infected cells. To investigate the mechanism by which the protein regulates the major immediate-early promoter, we utilized a mutant virus expressing an epitope-tagged pUL83 from its own promoter to identify protein binding partners for pUL83 during infection. We identified and confirmed the interaction of pUL83 with cellular IFI16 family members throughout the course of HCMV infection. pUL83 recruits IFI16 to the major immediate-early promoter, and IFI16 binding at the promoter is dependent upon the presence of pUL83. Consistent with the results obtained with the UL83Stop virus, infection of IFI16 knockdown cells with wild-type virus resulted in decreased levels of immediate-early transcripts compared to those of control cells. These data identify a previously unknown role for pUL83 in the initiation of the human cytomegalovirus gene expression cascade.


Asunto(s)
Citomegalovirus/fisiología , Genes Inmediatos-Precoces , Interacciones Huésped-Patógeno , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Proteínas de la Matriz Viral/metabolismo , Células Cultivadas , Codón sin Sentido , ADN Viral/metabolismo , Fibroblastos/virología , Técnicas de Silenciamiento del Gen , Genes Reporteros , Humanos , Mutación Missense , Proteínas Nucleares/antagonistas & inhibidores , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas de la Matriz Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA