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










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 88(15): 8355-60, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24829348

RESUMO

UNLABELLED: Viruses are dependent on their host cells for replication and thus have evolved in intimate association with them. The identification of host factors required for viral infection has led to advances in both viral and cellular biology. Vesicular stomatitis virus (VSV), a negative-sense RNA virus, replicates in all eukaryotic cells in culture, suggesting that the host requirements for its replication are ubiquitous. In this study, we performed a genome-wide small interfering RNA screen of human cells in culture and identified multiple cellular genes that influence the entry and replication of VSV. From a list of >300 genes, we selected the most promising candidates to perform further analysis to assign their functions to either the entry or intracellular replication step of infection. We implicate 3 new factors in VSV entry and 20 new factors in viral gene expression. These proteins have diverse cellular roles, including S-adenosylmethionine synthesis, respiration, and host translation machinery, underscoring the intimate relationship between VSV and the host cell. Together, these results provide a curated list of genes required for VSV replication. IMPORTANCE: Replication of vesicular stomatitis virus (VSV) has long served as a model for understanding host-virus interactions and neuropathogenesis. We performed a genome-wide analysis of host factors and revealed genes critical for viral replication, including some involved in vesicular trafficking, cell cycling, and protein modification. Our results provide an enriched list of host factors that are required for specific stages of VSV entry and gene expression. This study may also potentially expand the repertoire of targets for antiviral therapy against negative-strand RNA viruses.


Assuntos
Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , RNA Interferente Pequeno/biossíntese , Vesiculovirus/fisiologia , Internalização do Vírus , Replicação Viral , Linhagem Celular , Humanos , RNA Interferente Pequeno/genética , Vesiculovirus/crescimento & desenvolvimento
2.
Proc Natl Acad Sci U S A ; 110(1): 324-9, 2013 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-23169626

RESUMO

Initiation is the primary target of translational control for all organisms. Regulation of eukaryotic translation is traditionally thought to occur through initiation factors and RNA structures. Here, we characterize a transcript-specific translation initiation mechanism that is mediated by the ribosome. By studying vesicular stomatitis virus (VSV), we identify the large ribosomal subunit protein rpL40 as requisite for VSV cap-dependent translation but not bulk cellular or internal ribosome entry site-driven translation. This requirement is conserved among members of the order Mononegavirales, including measles virus and rabies virus. Polysome analyses and in vitro reconstitution of initiation demonstrate that rpL40 is required for 80S formation on VSV mRNAs through a cis-regulatory element. Using deep sequencing, we further uncover a subset of cellular transcripts that are selectively sensitive to rpL40 depletion, suggesting VSV may have usurped an endogenous translation pathway. Together, these findings demonstrate that the ribosome acts as a translational regulator outside of its catalytic role during protein synthesis.


Assuntos
Regulação Viral da Expressão Gênica/fisiologia , Iniciação Traducional da Cadeia Peptídica/fisiologia , RNA Mensageiro/metabolismo , Proteínas Ribossômicas/metabolismo , Subunidades Ribossômicas Maiores de Eucariotos/fisiologia , Vesiculovirus/genética , Células HeLa , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Immunoblotting , Microscopia de Fluorescência , Iniciação Traducional da Cadeia Peptídica/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Saccharomyces cerevisiae
3.
J Virol ; 86(2): 655-66, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22072764

RESUMO

Viruses coopt cellular membrane transport to invade cells, establish intracellular sites of replication, and release progeny virions. Recent genome-wide RNA interference (RNAi) screens revealed that genetically divergent viruses require biosynthetic membrane transport by the COPI coatomer complex for efficient replication. Here we found that disrupting COPI function by RNAi inhibited an early stage of vesicular stomatitis virus (VSV) replication. To dissect which replication stage(s) was affected by coatomer inactivation, we used visual and biochemical assays to independently measure the efficiency of viral entry and gene expression in hamster (ldlF) cells depleted of the temperature-sensitive ε-COP subunit. We show that ε-COP depletion for 12 h caused a primary block to virus internalization and a secondary defect in viral gene expression. Using brefeldin A (BFA), a chemical inhibitor of COPI function, we demonstrate that short-term (1-h) BFA treatments inhibit VSV gene expression, while only long-term (12-h) treatments block virus entry. We conclude that prolonged coatomer inactivation perturbs cellular endocytic transport and thereby indirectly impairs VSV entry. Our results offer an explanation of why COPI coatomer is frequently identified in screens for cellular factors that support cell invasion by microbial pathogens.


Assuntos
Complexo I de Proteína do Envoltório/genética , Regulação para Baixo , Regulação Viral da Expressão Gênica , Inativação Gênica , Estomatite Vesicular/genética , Vírus da Estomatite Vesicular Indiana/fisiologia , Internalização do Vírus , Animais , Linhagem Celular , Complexo I de Proteína do Envoltório/metabolismo , Cricetinae , Humanos , Estomatite Vesicular/metabolismo , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/genética
4.
J Virol ; 83(8): 3429-35, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19176616

RESUMO

The alphavirus Sindbis virus (SINV) causes encephalomyelitis in mice by infecting neurons of the brain and spinal cord. The outcome is age dependent. Young animals develop fatal disease, while older animals recover from infection. Recovery requires noncytolytic clearance of SINV from neurons, and gamma interferon (IFN-gamma) is an important contributor to clearance in vivo. IFN-gamma-dependent clearance has been studied using immortalized CSM14.1 rat neuronal cells that can be differentiated in vitro. Previous studies have shown that differentiated, but not undifferentiated, cells develop prolonged SINV replication and respond to IFN-gamma treatment with noncytolytic clearance of virus preceded by suppression of genomic viral RNA synthesis and reactivation of cellular protein synthesis. To determine the signaling mechanisms responsible for clearance, the responses of SINV-infected differentiated neurons to IFN-gamma were examined. IFN-gamma treatment of SINV-infected differentiated CSM14.1 cells, AP-7 olfactory neuronal cells, and primary dorsal root ganglia neurons triggered prolonged Stat-1 Tyr(701) phosphorylation, Stat-1 Ser(727) phosphorylation, and transient Stat-5 phosphorylation. Inhibition of Jak kinase activity with Jak inhibitor I completely reversed the neuroprotective and antiviral activities of IFN-gamma in differentiated cells. We conclude that activation of the Jak/Stat pathway is the primary mechanism for IFN-gamma-mediated clearance of SINV infection from mature neurons.


Assuntos
Interferon gama/imunologia , Janus Quinases/metabolismo , Neurônios/virologia , Fator de Transcrição STAT1/metabolismo , Sindbis virus/imunologia , Animais , Linhagem Celular , Células Cultivadas , RNA Viral/biossíntese , Ratos , Fator de Transcrição STAT5/metabolismo , Sindbis virus/fisiologia , Replicação Viral
5.
J Virol ; 81(11): 5628-36, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17376910

RESUMO

Sindbis virus (SINV) is an alphavirus that causes infection of neurons and encephalomyelitis in adult immunocompetent mice. Recovery can occur without apparent neurological damage. To better define the factors facilitating noncytolytic clearance of SINV in different regions of the central nervous system (CNS) and the roles of innate and adaptive immune responses at different times during infection, we have characterized SINV infection and clearance in the brain, brain stem, and spinal cords of severe combined immunodeficiency (SCID) and C57BL/6 (wild-type [WT]) mice and mice deficient in beta interferon (IFN-beta) (BKO), antibody (muMT), IFN-gamma (GKO), IFN-gamma receptor (GRKO), and both antibody and IFN-gamma (muMT/GKO). WT mice cleared infectious virus by day 8, while SCID mice had persistent virus replication at all sites. For 3 days after infection, BKO mice had higher titers at all sites than WT mice, despite similar IFN-alpha production, but cleared virus similarly. GKO and GRKO mice cleared infectious virus from all sites by days 8 to 10 and, like WT mice, displayed transient reactivation at 12 to 22 days. muMT mice did not clear virus from the brain, and clearance from the brain stem and lumbar spinal cord was delayed, followed by reactivation. Eighty-one days after infection, muMT/GKO mice had not cleared virus from any site, but titers were lower than for SCID mice. These studies show that IFN-beta is independently important for early control of CNS virus replication, that antiviral antibody is critical for clearance from the brain, and that both antibody and IFN-gamma contribute to prevention of reactivation after initial clearance.


Assuntos
Infecções por Alphavirus/imunologia , Anticorpos Antivirais/fisiologia , Encéfalo/virologia , Interferon beta/fisiologia , Interferon gama/fisiologia , Sindbis virus/imunologia , Medula Espinal/virologia , Infecções por Alphavirus/virologia , Animais , Anticorpos Antivirais/genética , Encéfalo/imunologia , Citotoxicidade Imunológica , Combinação de Medicamentos , Sinergismo Farmacológico , Interferon beta/deficiência , Interferon beta/genética , Interferon gama/deficiência , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Medula Espinal/imunologia , Ativação Viral/imunologia
6.
J Virol ; 79(9): 5374-85, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15827152

RESUMO

Due to the nonrenewable nature of neurons, recovery from viral infection of the central nervous system requires noncytopathic mechanisms for control of virus replication. Recovery from alphavirus encephalitis can occur without apparent neurological damage through the effects of antibody and gamma interferon (IFN-gamma). To establish an in vitro cell culture system that will allow the study of mechanisms of IFN-gamma-mediated control of Sindbis virus (SINV) replication in neurons, we have characterized the susceptibility to SINV infection and IFN-gamma responsiveness of two neuronal cell lines that can be differentiated in vitro: CSM14.1, a rat nigral cell line, and NSC34, a mouse motor neuron cell line. Undifferentiated CSM14.1 and NSC34 cells were permissive for SINV and susceptible to virus-induced cell death. With differentiation, CSM14.1 cells reduced virus replication and became progressively resistant to virus-induced cell death, resulting in prolonged virus replication. NSC34 cells did not differentiate completely and became only partially resistant to SINV infection. Both CSM14.1 and NSC34 cells responded to pretreatment with IFN-gamma by decreasing SINV replication. Differentiated CSM14.1 cells treated 24 h after infection with IFN-gamma responded with increased cell viability and clearance of infectious virus. IFN-gamma treatment sequentially altered the ratio of genomic to subgenomic viral RNA synthesis, promoted recovery of cellular protein synthesis, reduced viral protein synthesis, and inhibited viral RNA transcription within 24 h after treatment. We conclude that CSM14.1 cells provide an excellent model for the study of IFN-gamma-mediated noncytolytic clearance of SINV from mature neurons.


Assuntos
Interferon gama/farmacologia , Neurônios/virologia , Inibidores da Síntese de Proteínas/farmacologia , Sindbis virus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Relação Dose-Resposta a Droga , Interferon gama/biossíntese , Camundongos , Neurônios/imunologia , Neurônios/metabolismo , RNA Viral/biossíntese , Ratos , Proteínas Recombinantes , Sindbis virus/fisiologia , Fatores de Tempo , Proteínas Virais/biossíntese , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...