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











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 90(22): 10414-10422, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27630226

RESUMO

Herpes simplex virus 1 (HSV-1) remodels nuclear membranes during virus egress. Although the UL31 and UL34 proteins control nucleocapsid transit in infected cells, the molecular interactions required for their function are unclear. Here we report that the γ134.5 gene product of HSV-1 facilitates nucleocapsid release to the cytoplasm through bridging the UL31/UL34 complex, cellular p32, and protein kinase C. Unlike wild-type virus, an HSV mutant devoid of γ134.5 or its amino terminus is crippled for viral growth and release. This is attributable to a defect in virus nuclear egress. In infected cells, wild-type virus recruits protein kinase C to the nuclear membrane and triggers its activation, whereas the γ134.5 mutants fail to exert such an effect. Accordingly, the γ134.5 mutants are unable to induce phosphorylation and reorganization of lamin A/C. When expressed in host cells γ134.5 targets p32 and protein kinase C. Meanwhile, it communicates with the UL31/UL34 complex through UL31. Deletion of the amino terminus from γ134.5 disrupts its activity. These results suggest that disintegration of the nuclear lamina mediated by γ134.5 promotes HSV replication. IMPORTANCE: HSV nuclear egress is a key step that determines the outcome of viral infection. While the nuclear egress complex mediates capsid transit across the nuclear membrane, the regulatory components are not clearly defined in virus-infected cells. We report that the γ134.5 gene product, a virulence factor of HSV-1, facilitates nuclear egress cooperatively with cellular p32, protein kinase C, and the nuclear egress complex. This work highlights a viral mechanism that may contribute to the pathogenesis of HSV infection.


Assuntos
Herpesvirus Humano 1/metabolismo , Lamina Tipo A/metabolismo , Fosforilação/fisiologia , Proteínas Virais/metabolismo , Liberação de Vírus/fisiologia , Animais , Capsídeo/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Chlorocebus aethiops , Citoplasma/metabolismo , Citoplasma/virologia , Células HeLa , Humanos , Membrana Nuclear/metabolismo , Membrana Nuclear/virologia , Lâmina Nuclear/metabolismo , Lâmina Nuclear/virologia , Proteínas Nucleares/metabolismo , Nucleocapsídeo/metabolismo , Proteína Quinase C/metabolismo , Células Vero , Montagem de Vírus/fisiologia
2.
J Virol ; 87(9): 5019-27, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23427156

RESUMO

Human cytomegalovirus (HCMV) encodes one conventional protein kinase, UL97. During infection, UL97 phosphorylates the retinoblastoma tumor suppressor protein (pRb) on sites ordinarily phosphorylated by cyclin-dependent kinases (CDK), inactivating the ability of pRb to repress host genes required for cell cycle progression to S phase. UL97 is important for viral DNA synthesis in quiescent cells, but this function can be replaced by human papillomavirus type 16 E7, which targets pRb for degradation. However, viruses in which E7 replaces UL97 are still defective for virus production. UL97 is also required for efficient nuclear egress of viral nucleocapsids, which is associated with disruption of the nuclear lamina during infection, and phosphorylation of lamin A/C on serine 22, which antagonizes lamin polymerization. We investigated whether inactivation of pRb might overcome the requirement of UL97 for these roles, as pRb inactivation induces CDK1, and CDK1 phosphorylates lamin A/C on serine 22. We found that lamin A/C serine 22 phosphorylation during HCMV infection correlated with expression of UL97 and was considerably delayed in UL97-null mutants, even when E7 was expressed. E7 failed to restore gaps in the nuclear lamina seen in wild-type but not UL97-null virus infections. In electron microscopy analyses, a UL97-null virus expressing E7 was as impaired as a UL97-null mutant in cytoplasmic accumulation of viral nucleocapsids. Our results demonstrate that pRb inactivation is insufficient to restore efficient viral nuclear egress of HCMV in the absence of UL97 and instead argue further for a direct role of UL97 in this stage of the infectious cycle.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/enzimologia , Lâmina Nuclear/virologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteína do Retinoblastoma/metabolismo , Liberação de Vírus , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Citomegalovirus/genética , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Humanos , Lamina Tipo A/química , Lamina Tipo A/metabolismo , Lâmina Nuclear/química , Lâmina Nuclear/metabolismo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Polimerização , Proteína do Retinoblastoma/genética
3.
Adv Virus Res ; 80: 69-101, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21762822

RESUMO

Viral replication is a complex process relying on a network of interacting viral and cellular proteins, in which particularly protein kinases play an important regulatory role. The specific phosphorylation of substrate proteins induces activation, inactivation, or other functional modification and thus determines virus-host cell interregulation. During herpesviral infections, both viral and cellular protein kinases are expressed and provide activities crucial for the efficiency of virus replication. The protein kinase pUL97 encoded by human cytomegalovirus (HCMV) is a multifunctional regulatory enzyme which exerts strong regulatory effects on early and late steps of the viral replication cycle. A number of interacting proteins and substrates of pUL97 have been described, including retinoblastoma (Rb) protein, nuclear lamins and viral pUL69. Recently, it was demonstrated that pUL97 has structural and functional resemblance to cyclin-dependent protein kinases (CDKs) and thus represents a CDK ortholog. pUL97 can phosphorylate and inactivate Rb, resulting in a stimulation of cell cycle progression. In addition, the association of pUL97 activity with nucleocytoplasmic export of viral capsids has been demonstrated by several investigators. We could show that pUL97 is able to phosphorylate nuclear lamins and to contribute to the HCMV-induced reorganization of the nuclear lamina. On the basis of very recent findings, it is becoming increasingly clear that pUL97 is a component of a multiprotein nuclear egress complex (NEC). The NEC contains a small number of egress proteins involved in the recruitment of protein kinases, such as pUL97 and cellular protein kinase C (PKC), to specific sites of the nuclear lamina. Current information about the composition, function, and regulatory complexity of the NEC leads to a mechanistic concept which may set the key features of HCMV nuclear egress in a new light.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Citomegalovirus/genética , Regulação Viral da Expressão Gênica , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Linhagem Celular , Quinases Ciclina-Dependentes/genética , Citomegalovirus/enzimologia , Citomegalovirus/fisiologia , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Humanos , Lâmina Nuclear/genética , Lâmina Nuclear/metabolismo , Lâmina Nuclear/virologia , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Retinoblastoma/genética , Retinoblastoma/metabolismo , Replicação Viral
4.
Virology ; 320(1): 85-98, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15003865

RESUMO

Nuclear matrix is a structural framework of important nuclear processes. We studied the effect of two different types of viral infections on nuclear matrix. HeLa cells were infected with human rhinovirus 1B (HRV 1B) or measles virus (MV), and Nuclear Mitotic Apparatus protein (NuMA) and lamins A/C and B were used as markers for internal nuclear matrix and peripheral nuclear lamina, respectively. We show that NuMA, lamins, and poly(ADP-ribose) polymerase-1 are cleaved during viral infection in a virus family-specific manner suggesting that these viruses activate different sets of proteases. Morphologically, NuMA was excluded from the condensed chromatin, lamins showed a folded distribution, and both proteins finally remained around the nuclear fragments. A general caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (z-VAD-FMK) prevented the nuclear disintegration and the cleavage of the proteins studied. Interestingly, z-VAD-FMK rescued MV-infected but not HRV 1B-infected cells from cell death. These results show for the first time that NuMA and lamins are specific target proteins during virus-induced programmed cell death.


Assuntos
Inibidores de Caspase , Laminas/metabolismo , Vírus do Sarampo , Lâmina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Rhinovirus , Fuso Acromático/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Antígenos Nucleares , Proteínas de Ciclo Celular , Morte Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Células HeLa , Humanos , Lâmina Nuclear/virologia , Proteínas Associadas à Matriz Nuclear , Poli(ADP-Ribose) Polimerases/metabolismo , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA