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
2.
J Biol Chem ; 288(5): 3571-84, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23233667

RESUMO

Autophagy has been shown to facilitate replication or production of avian reovirus (ARV); nevertheless, how ARV induces autophagy remains largely unknown. Here, we demonstrate that the nonstructural protein p17 of ARV functions as an activator of autophagy. ARV-infected or p17-transfected cells present a fast and strong induction of autophagy, resulting in an increased level of autophagic proteins Beclin 1 and LC3-II. Although autophagy was suppressed by 3-methyladenine or shRNAs targeting autophagic proteins (Beclin 1, ATG7, and LC3) as well as by overexpression of Bcl-2, viral transcription, σC protein synthesis, and virus yield were all significantly reduced, suggesting a key role of autophagosomes in supporting ARV replication. Furthermore, we revealed for the first time that p17 positively regulates phosphatase and tensin deleted on chromosome 10 (PTEN), AMP-activated protein kinase (AMPK), and dsRNA dependent protein kinase RNA (PKR)/eIF2α signaling pathways, accompanied by down-regulation of Akt and mammalian target of rapamycin complex 1, thereby triggering autophagy. By using p53, PTEN, PKR, AMPK, and p17 short hairpin RNA (shRNA), activation of signaling pathways and LC3-II levels was significantly suppressed, suggesting that p17 triggers autophagy through activation of p53/PTEN, AMPK, and PKR signaling pathways. Furthermore, colocalization of LC3 with viral proteins (p17 and σC), p62 with LAMP2 and LC3 with Rab7 was observed under a fluorescence microscope. The expression level of p62 was increased at 18 h postinfection and then slightly decreased 24 h postinfection compared with mock infection and thapsigargin treatment. Furthermore, disruption of autophagosome-lysosome fusion by shRNAs targeting LAMP2 or Rab7a resulted in inhibition of viral protein synthesis and virus yield, suggesting that formation of autolysosome benefits virus replication. Taken together, our results suggest that ARV induces formation of autolysosome but does not induce complete autophagic flux.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia , Orthoreovirus Aviário/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Proteínas Quinases Ativadas por AMP/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Galinhas , Ativação Enzimática , Fator de Iniciação 2 em Eucariotos/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Orthoreovirus Aviário/crescimento & desenvolvimento , Orthoreovirus Aviário/fisiologia , PTEN Fosfo-Hidrolase/genética , Fagossomos/metabolismo , RNA de Cadeia Dupla/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/metabolismo , eIF-2 Quinase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
J Virol ; 86(24): 13653-61, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23055561

RESUMO

The specific cell pathways involved in bovine ephemeral fever virus (BEFV) cell entry have not been determined. In this work, colocalization of the M protein of BEFV with clathrin or dynamin 2 was observed under a fluorescence microscope. To better understand BEFV entry, we carried out internalization studies with a fluorescently labeled BEFV by using a lipophilic dye, 3,30-dilinoleyloxacarbocyanine perchlorate (DiO), further suggesting that BEFV uses a clathrin-mediated endocytosis pathway. Our results suggest that clathrin-mediated and dynamin 2-dependent endocytosis is an important avenue of BEFV entry. Suppression of Rab5 or Rab7a through the use of a Rab5 dominant negative mutant and Rab7a short hairpin RNA (shRNA) demonstrated that BEFV requires both early and late endosomes for endocytosis and subsequent infection in MDBK and Vero cells. Treatment of BEFV-infected cells with nocodazole significantly decreased the M protein synthesis and viral yield, indicating that microtubules play an important role in BEFV productive infection, likely by mediating trafficking of BEFV-containing endosomes. Furthermore, BEFV infection was strongly blocked by different inhibitors of endosomal acidification, suggesting that virus enters host cells by clathrin-mediated and dynamin 2-dependent endocytosis in a pH-dependent manner.


Assuntos
Clatrina/fisiologia , Dinamina II/fisiologia , Endocitose/fisiologia , Vírus da Febre Efêmera Bovina/fisiologia , Microtúbulos/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Proteínas rab5 de Ligação ao GTP/fisiologia , Animais , Sequência de Bases , Bovinos , Linhagem Celular , Primers do DNA , proteínas de unión al GTP Rab7
4.
J Biol Chem ; 286(35): 30780-30794, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21705803

RESUMO

Very little is known about the mechanism of cell entry of avian reovirus (ARV). The aim of this study was to explore the mechanism of ARV entry and subsequent infection. Cholesterol mainly affected the early steps of the ARV life cycle, because the presence of cholesterol before and during viral adsorption greatly blocked ARV infectivity. Although we have demonstrated that ARV facilitating p38 MAPK is beneficial for virus replication, its mechanism remains unknown. Here, we show that ARV-induced phosphorylation of caveolin-1 (Tyr(14)), dynamin-2 expression, and Rac1 activation through activation of p38 MAPK and Src in the early stage of the virus life cycle is beneficial for virus entry and productive infection. The strong inhibition by dynasore, a specific inhibitor of dynamin-2, and depletion of endogenous caveolin-1 or dynamin-2 by siRNAs as well as the caveolin-1 colocalization study implicate caveolin-1-mediated and dynamin-2-dependent endocytosis as a significant avenue of ARV entry. By means of pharmacological inhibitors, dominant negative mutants, and siRNA of various cellular proteins and signaling molecules, phosphorylation of caveolin-1, dynamin-2 expression, and Rac1 activation were suppressed, suggesting that by orchestrating p38 MAPK, Src, and Rac1 signaling cascade in the target cells, ARV creates an appropriate intracellular environment facilitating virus entry and productive infection. Furthermore, disruption of microtubules, Rab5, or endosome acidification all inhibited ARV infection, suggesting that microtubules and small GTPase Rab5, which regulate transport to early endosome, are crucial for survival of ARV and that exposure of the virus to acidic pH is required for productive infection.


Assuntos
Caveolina 1/metabolismo , Dinamina II/metabolismo , Microtúbulos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/química , Orthoreovirus Aviário/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Colesterol/metabolismo , Endocitose , Ativação Enzimática , Regulação da Expressão Gênica , Concentração de Íons de Hidrogênio , Lisossomos/metabolismo , RNA Interferente Pequeno/metabolismo , Células Vero
5.
J Virol ; 84(15): 7683-94, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20484520

RESUMO

The effects of avian reovirus (ARV) p17 protein on cell cycle progression and host cellular protein translation were studied. ARV infection and ARV p17 transfection resulted in the accumulation of infected and/or transfected cells in the G(2)/M phase of the cell cycle. The accumulation of cells in the G(2)/M phase was accompanied by upregulation and phosphorylation of the G(2)/M-phase proteins ATM, p53, p21(cip1/waf1), Cdc2, cyclin B1, Chk1, Chk2, and Cdc25C, suggesting that p17 induces a G(2)/M cell cycle arrest through activation of the ATM/p53/p21(cip1/waf1)/Cdc2/cyclin B1 and ATM/Chk1/Chk2/Cdc25C pathways. The G(2)/M cell cycle arrest resulted in increased virus replication. In the present study, we also provide evidence demonstrating that p17 protein is responsible for ARV-induced host cellular protein translation shutoff. Increased phosphorylation levels of the eukaryotic translation elongation factor 2 (eEF2) and initiation factor eIF2alpha and reduced phosphorylation levels of the eukaryotic translation initiation factors eIF4E, eIF4B, and eIF4G, as well as 4E-BP1 and Mnk-1 in p17-transfected cells, demonstrated that ARV p17 suppresses translation initiation factors and translation elongation factors to induce host cellular protein translation shutoff. Inhibition of mTOR by rapamycin resulted in a decrease in the levels of phosphorylated 4E-BP1, eIF4B, and eIF4G and an increase in the levels eEF2 but did not affect ARV replication, suggesting that ARV replication was not hindered by inhibition of cap-dependent translation. Taken together, our data indicate that ARV p17-induced G(2)/M arrest and host cellular translation shutoff resulted in increased ARV replication.


Assuntos
Ciclo Celular , Interações Hospedeiro-Patógeno , Orthoreovirus Aviário/patogenicidade , Biossíntese de Proteínas , Proteína Supressora de Tumor p53/metabolismo , Proteínas não Estruturais Virais/fisiologia , Fatores de Virulência/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Fatores de Iniciação em Eucariotos/metabolismo , Humanos , Fator 2 de Elongação de Peptídeos/metabolismo , Fosforilação
6.
Biochem Biophys Res Commun ; 384(3): 301-5, 2009 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-19406104

RESUMO

Viral infection usually influences cellular protein synthesis either actively or passively via modification of various translation initiation factors. Here we demonstrated that infection with avian reovirus (ARV) interfered with cellular protein synthesis. This study demonstrated for the first time that ARV influenced the phosphorylation of translation initiation factors including eIF4E and eIF-4G. Interestingly, ARV also induced phosphorylation of eukaryotic translation elongation factor (eEF2) in a time- and dose-dependent manner. Inhibition of mTOR by rapamycin notably increased the level of phosphorylated eEF2 in infected cells. However, rapamycin did not show any negative effects on ARV replication, suggesting that phosphorylation of eEF2 in infected cells did not reduce ARV propagation. These results demonstrated for the first time that ARV promotes phosphorylation of eEF2 which in turn influenced host protein production not simply by modulating the function of translation initiation factors but also by regulating elongation factor eEF2.


Assuntos
Orthoreovirus Aviário/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo , Biossíntese de Proteínas , Infecções por Reoviridae/metabolismo , Animais , Chlorocebus aethiops , Fosforilação , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA