Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28515293

RESUMO

Virus-like vesicles (VLVs) are membrane-enclosed vesicles that resemble native enveloped viruses in organization but lack the viral capsid and genome. During the productive infection of tumor-associated gammaherpesviruses, both virions and VLVs are produced and are released into the extracellular space. However, studies of gammaherpesvirus-associated VLVs have been largely restricted by the technical difficulty of separating VLVs from mature virions. Here we report a strategy of selectively isolating VLVs by using a Kaposi's sarcoma-associated herpesvirus (KSHV) mutant that is defective in small capsid protein and is unable to produce mature virions. Using mass spectrometry analysis, we found that VLVs contained viral glycoproteins required for cellular entry, as well as tegument proteins involved in regulating lytic replication, but lacked capsid proteins. Functional analysis showed that VLVs induced the expression of the viral lytic activator RTA, initiating KSHV lytic gene expression. Furthermore, employing RNA sequencing, we performed a genomewide analysis of cellular responses triggered by VLVs and found that PRDM1, a master regulator in cell differentiation, was significantly upregulated. In the context of KSHV replication, we demonstrated that VLV-induced upregulation of PRDM1 was necessary and sufficient to reactivate KSHV by activating its RTA promoter. In sum, our study systematically examined the composition of VLVs and demonstrated their biological roles in manipulating host cell responses and facilitating KSHV lytic replication.IMPORTANCE Cells lytically infected with tumor-associated herpesviruses produce a high proportion of virus-like vesicles (VLVs). The composition and function of VLVs have not been well defined, largely due to the inability to efficiently isolate VLVs that are free of virions. Using a cell system capable of establishing latent KSHV infection and robust reactivation, we successfully isolated VLVs from a KSHV mutant defective in the small capsid protein. We quantitatively analyzed proteins and microRNAs in VLVs and characterized the roles of VLVs in manipulating host cells and facilitating viral infection. More importantly, we demonstrated that by upregulating PRDM1 expression, VLVs triggered differentiation signaling in targeted cells and facilitated viral lytic infection via activation of the RTA promoter. Our study not only demonstrates a new strategy for isolating VLVs but also shows the important roles of KSHV-associated VLVs in intercellular communication and the viral life cycle.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Repressoras/biossíntese , Transdução de Sinais , Virossomos/química , Replicação Viral , Diferenciação Celular , Linhagem Celular , Herpesvirus Humano 8/química , Humanos , Proteínas Imediatamente Precoces/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo , Transativadores/metabolismo , Regulação para Cima
2.
J Gen Virol ; 98(2): 242-250, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27926822

RESUMO

The regulation of the late viral gene expression in betaherpesviruses is largely undefined. We have previously shown that the murine cytomegalovirus proteins pM79 and pM92 are required for late gene transcription. Here, we provide insight into the mechanism of pM79 and pM92 activity by determining their interaction partners during infection. Co-immunoprecipitation-coupled MS studies demonstrate that pM79 and pM92 interact with an array of cellular and viral proteins involved in transcription. Specifically, we identify RNA polymerase II as a cellular target for both pM79 and pM92. We use inter-protein coevolution analysis to show how pM79 and pM92 likely assemble into a late transcription complex composed of late transcription regulators pM49, pM87 and pM95. Combining proteomic methods with coevolution computational analysis provides novel insights into the relationship between pM79, pM92 and RNA polymerase II and allows the generation of a model of the multi-component viral complex that regulates late gene transcription.


Assuntos
Regulação Viral da Expressão Gênica , Muromegalovirus/genética , RNA Polimerase II/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas Virais Reguladoras e Acessórias/metabolismo , Animais , Evolução Molecular , Imunoprecipitação , Camundongos , Muromegalovirus/classificação , Filogenia , Análise Serial de Proteínas , Proteômica , RNA Polimerase II/genética , Fatores de Transcrição/classificação , Fatores de Transcrição/genética , Proteínas Virais Reguladoras e Acessórias/classificação , Proteínas Virais Reguladoras e Acessórias/genética
3.
J Virol ; 88(1): 131-42, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24131717

RESUMO

In this study, we report that murine cytomegalovirus (MCMV) protein pM92 regulates viral late gene expression during virus infection. Previously, we have shown that MCMV protein pM79 and its human cytomegalovirus (HCMV) homologue pUL79 are required for late viral gene transcription. Identification of additional factors involved is critical to dissecting the mechanism of this regulation. We show here that pM92 accumulated abundantly at late times of infection in a DNA synthesis-dependent manner and localized to nuclear viral replication compartments. To investigate the role of pM92, we constructed a recombinant virus SMin92, in which pM92 expression was disrupted by an insertional/frameshift mutation. During infection, SMin92 accumulated representative viral immediate-early gene products, early gene products, and viral DNA sufficiently but had severe reduction in the accumulation of late gene products and was thus unable to produce infectious progeny. Coimmunoprecipitation and mass spectrometry analysis revealed an interaction between pM92 and pM79, as well as between their HCMV homologues pUL92 and pUL79. Importantly, we showed that the growth defect of pUL92-deficient HCMV could be rescued in trans by pM92. This study indicates that pM92 is an additional viral regulator of late gene expression, that these regulators (represented by pM92 and pM79) may need to complex with each other for their activity, and that pM92 and pUL92 share a conserved function in CMV infection. pM92 represents a potential new target for therapeutic intervention in CMV disease, and a gateway into studying a largely uncharted viral process that is critical to the viral life cycle.


Assuntos
Citomegalovirus/genética , Regulação Viral da Expressão Gênica/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Cromossomos Artificiais Bacterianos , Citomegalovirus/crescimento & desenvolvimento , Citomegalovirus/fisiologia , Primers do DNA , Imunoprecipitação , Espectrometria de Massas , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos , Proteínas Virais/química , Proteínas Virais/genética , Replicação Viral
4.
J Virol ; 87(16): 9135-47, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23760242

RESUMO

Herpesvirus genes are temporally expressed during permissive infections, but how their expression is regulated at late times is poorly understood. Previous studies indicate that the human cytomegalovirus (CMV) gene, UL79, is required for late gene expression. However, the mechanism remains to be fully elucidated, and UL79 homologues in other CMVs have not been studied. Here, we characterized the role of the conserved murine CMV (MCMV) gene M79. We showed that M79 encoded a protein (pM79) which was expressed with early-late kinetics and localized to nuclear viral replication compartments. M79 transcription was significantly decreased in the absence of viral DNA synthesis but markedly stimulated by pM79. To investigate its role, we created the recombinant virus SMin79, in which pM79 expression was disrupted. While marker-rescued virus grew efficiently in fibroblasts, SMin79 failed to produce infectious progeny but was rescued by pM79 expression in trans. During SMin79 infection, representative viral immediate-early and early gene products as well as viral DNA accumulated sufficiently. Formation of viral replication compartments also appeared normal. Pulsed-field gel electrophoresis analysis indicated that the overall structure of replicating viral DNA was indistinguishable between wild-type and SMin79 infection. Viral tiled array and quantitative PCR analysis revealed that many late transcripts sensitive to a viral DNA synthesis inhibitor (phosphonoacetic acid) were markedly reduced by pM79 mutation. This study indicates that cytomegaloviruses use a conserved mechanism to promote transcription at late stages of infection and that pM79 is a critical regulator for at least a subset of viral DNA synthesis-dependent transcripts.


Assuntos
Regulação Viral da Expressão Gênica , Muromegalovirus/fisiologia , Transcrição Gênica , Proteínas Virais/metabolismo , Replicação Viral , Animais , Núcleo Celular/química , Fibroblastos/virologia , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Camundongos , Muromegalovirus/genética , Proteínas Virais/genética
5.
J Virol ; 86(12): 6712-23, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22496230

RESUMO

The unfolded-protein response (UPR), activated by sensor molecules PERK, ATF6, and IRE1 to resolve endoplasmic reticulum (ER) stress, has emerged as a key target for host cells and viruses to control the infection outcomes. The UPR regulates ER protein folding, controls cell fate upon ER stress, and plays an important role in innate immunity. We and others have shown that human cytomegalovirus (HCMV) modulates the UPR. We show here that murine CMV (MCMV), the widely used CMV model for small animal infection, regulated the UPR in a manner similar to that of HCMV. This modulatory ability was triggered by virion entry and enhanced by viral immediate-early and early gene expression. Thus, while vulnerable at early times, MCMV became resistant to exogenous ER stress at late times of infection. MCMV activated the PERK-ATF4 pathway but only induced a subset of representative ATF4 targets at levels somewhat lower than those by the ER stress inducer tunicamycin. Moreover, MCMV induced ER chaperone Bip but actively blocked IRE1-mediated Xbp1(s) protein accumulation. ATF4 depletion severely attenuated viral growth at a low multiplicity of infection by modestly reducing viral DNA synthesis and more pronouncedly inhibiting late gene transcription. Collectively, we show that the UPR is a conserved target of CMVs and identify ATF4, a key UPR component, as a factor critical for MCMV infection. This work sets the stage for using the MCMV model to explore the role of this stress response in CMV biology, particularly during infection of the host, which is difficult to study in HCMV.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Infecções por Citomegalovirus/metabolismo , Infecções por Herpesviridae/metabolismo , Muromegalovirus/fisiologia , Resposta a Proteínas não Dobradas , Fator 4 Ativador da Transcrição/genética , Animais , Citomegalovirus/genética , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Modelos Animais de Doenças , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/virologia , Humanos , Camundongos , Muromegalovirus/genética
6.
iScience ; 1: 97-111, 2018 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-30227960

RESUMO

Zika virus (ZIKV) infection causes Guillain-Barré syndrome and severe birth defects. ZIKV envelope (E) protein is the major viral protein involved in cell receptor binding and entry and is therefore considered one of the major determinants in ZIKV pathogenesis. Here we report a gene-wide mapping of functional residues of ZIKV E protein using a mutant library, with changes covering every nucleotide position. By comparing the replication fitness of every viral mutant between mosquito and human cells, we identified that mutations affecting glycosylation display the most divergence. By characterizing individual mutants, we show that ablation of glycosylation selectively benefits ZIKV infection of mosquito cells by enhancing cell entry, whereas it either has little impact on ZIKV infection on certain human cells or leads to decreased infection through the entry factor DC-SIGN. In conclusion, we define the roles of individual residues of ZIKV envelope protein, which contribute to ZIKV replication fitness in human and mosquito cells.

7.
J Bacteriol ; 188(23): 8145-52, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17012401

RESUMO

Bacillus anthracis, the spore-forming agent of anthrax, requires iron for growth and is capable of scavenging heme-iron during infection. We show here that the B. anthracis iron-regulated surface determinants (isd) locus encompasses isdC, specifying a heme-iron binding surface protein. Anchoring of IsdC to the cell wall envelopes of vegetative bacilli requires srtB, which encodes sortase B. Purified sortase B cleaves IsdC between the threonine and the glycine of its NPKTG motif sorting signal. B. anthracis variants lacking either isdC or srtB display defects in heme-iron scavenging, suggesting that IsdC binding to heme-iron in the cell wall envelope contributes to bacterial uptake of heme.


Assuntos
Aminoaciltransferases/fisiologia , Bacillus anthracis/metabolismo , Proteínas de Bactérias/fisiologia , Proteínas de Transporte/fisiologia , Cisteína Endopeptidases/fisiologia , Heme/metabolismo , Ferro/metabolismo , Sequência de Aminoácidos , Bacillus anthracis/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Dados de Sequência Molecular , Peptidoglicano/metabolismo , Ligação Proteica , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA