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(3): 1461-72, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24257610

RESUMO

The vesicular stomatitis virus (VSV) RNA-dependent RNA polymerase consists of two viral proteins; the large (L) protein is the main catalytic subunit, and the phosphoprotein (P) is an essential cofactor for polymerase function. The P protein interacts with the L protein and the N-RNA template, thus connecting the polymerase to the template. P protein also binds to free N protein to maintain it in a soluble, encapsidation-competent form. Previously, five sites of phosphorylation were identified on the P protein and these sites were reported to be differentially important for mRNA synthesis or genomic replication. The previous studies were carried out by biochemical analysis of portions of the authentic viral P protein or by analysis of bacterium-expressed, exogenously phosphorylated P protein by mutagenesis. However, there has been no systematic biochemical search for phosphorylation sites on authentic, virus-expressed P protein. In this study, we analyzed the P protein isolated from VSV-infected cells for sites of phosphorylation by mass spectrometry. We report the identification of Tyr14 as a previously unidentified phosphorylation site of VSV P and show that it is essential for viral transcription and replication. However, our mass spectral analysis failed to observe the phosphorylation of previously reported C-terminal residues Ser226 and Ser227 and mutagenic analyses did not demonstrate a role for these sites in RNA synthesis.


Assuntos
Fosfoproteínas/química , Fosfoproteínas/metabolismo , RNA Viral/biossíntese , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo , Motivos de Aminoácidos , Humanos , Espectrometria de Massas , Fosfoproteínas/genética , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Viral/genética , Serina/genética , Serina/metabolismo , Tirosina/genética , Tirosina/metabolismo , Vírus da Estomatite Vesicular Indiana/química , Vírus da Estomatite Vesicular Indiana/genética , Proteínas Estruturais Virais/genética , Replicação Viral
2.
J Virol ; 86(10): 5637-46, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438540

RESUMO

The ORF3 protein of hepatitis E virus (HEV) is a multifunctional protein important for virus replication. The ORF3 proteins from human, swine, and avian strains of HEV contain a conserved PXXP amino acid motif, resembling either Src homology 3 (SH3) cell signaling interaction motifs or "late domains" involved in host cell interactions aiding in particle release. Using an avian strain of HEV, we determined the roles of the conserved prolines within the PREPSAPP motif in HEV replication and infectivity in Leghorn male hepatoma (LMH) chicken liver cells and in chickens. Each proline was changed to alanine to produce 8 avian HEV mutants containing single mutations (P64, P67, P70, and P71 to A), double mutations (P64/67A, P64/70A, and P67/70A), and triple mutations (P64/67/70A). The results showed that avian HEV mutants are replication competent in vitro, and none of the prolines in the PXXPXXPP motif are essential for infectivity in vivo; however, the second and third prolines appear to aid in fecal virus shedding, suggesting that the PSAP motif, but not the PREP motif, is involved in virus release. We also showed that the PSAP motif interacts with the host protein tumor suppressor gene 101 (TSG101) and that altering any proline within the PSAP motif disrupts this interaction. However, we showed that the ORF2 protein expressed in LMH cells is efficiently released from the cells in the absence of ORF3 and that coexpression of ORF2 and ORF3 did not act synergistically in this release, suggesting that another factor(s) such as ORF1 or viral genomic RNA may be necessary for proper particle release.


Assuntos
Hepatite E/virologia , Hepevirus/fisiologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Liberação de Vírus , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular Tumoral , Galinhas , Modelos Animais de Doenças , Vírus da Hepatite E/química , Vírus da Hepatite E/genética , Vírus da Hepatite E/fisiologia , Hepevirus/química , Hepevirus/genética , Humanos , Masculino , Dados de Sequência Molecular , Fases de Leitura Aberta , Proteínas Virais/genética , Replicação Viral
3.
J Virol ; 85(19): 10031-40, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21775444

RESUMO

The RNA genome of the hepatitis E virus (HEV) contains a hypervariable region (HVR) in ORF1 that tolerates small deletions with respect to infectivity. To further investigate the role of the HVR in HEV replication, we constructed a panel of mutants with overlapping deletions in the N-terminal, central, and C-terminal regions of the HVR by using a genotype 1 human HEV luciferase replicon and analyzed the effects of deletions on viral RNA replication in Huh7 cells. We found that the replication levels of the HVR deletion mutants were markedly reduced in Huh7 cells, suggesting a role of the HVR in viral replication efficiency. To further verify the results, we constructed HVR deletion mutants by using a genetically divergent, nonmammalian avian HEV, and similar effects on viral replication efficiency were observed when the avian HEV mutants were tested in LMH cells. Furthermore, the impact of complete HVR deletion on virus infectivity was tested in chickens, using an avian HEV mutant with a complete HVR deletion. Although the deletion mutant was still replication competent in LMH cells, the complete HVR deletion resulted in a loss of avian HEV infectivity in chickens. Since the HVR exhibits extensive variations in sequence and length among different HEV genotypes, we further examined the interchangeability of HVRs and demonstrated that HVR sequences are functionally exchangeable between HEV genotypes with regard to viral replication and infectivity in vitro, although genotype-specific HVR differences in replication efficiency were observed. The results showed that although the HVR tolerates small deletions with regard to infectivity, it may interact with viral and host factors to modulate the efficiency of HEV replication.


Assuntos
Vírus da Hepatite E/fisiologia , Fases de Leitura Aberta , RNA Viral/genética , Replicação Viral , Animais , Linhagem Celular , Galinhas , Análise Mutacional de DNA , Vírus da Hepatite E/genética , Hepatócitos/virologia , Humanos , Deleção de Sequência
4.
Vet Microbiol ; 147(3-4): 310-9, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-20708350

RESUMO

A genetically distinct strain of avian hepatitis E virus (avian HEV-VA strain) was isolated from a healthy chicken in Virginia, and thus it is important to characterize and compare its pathogenicity with the prototype strain (avian HEV-prototype) isolated from a diseased chicken. Here we first constructed an infectious clone of the avian HEV-VA strain. Capped RNA transcripts from the avian HEV-VA clone were replication-competent after transfection of LMH chicken liver cells. Chickens inoculated intrahepatically with RNA transcripts of avian HEV-VA clone developed active infection as evidenced by fecal virus shedding, viremia, and seroconversion. To characterize the pathogenicity, RNA transcripts of both avian HEV-VA and avian HEV-prototype clones were intrahepatically inoculated into the livers of chickens. Avian HEV RNA was detected in feces, serum and bile samples from 10/10 avian HEV-VA-inoculated and 9/9 avian HEV-prototype-inoculated chickens although seroconversion occurred only in some chickens during the experimental period. The histopathological lesion scores were lower for avian HEV-VA group than avian HEV-prototype group in the liver at 3 and 5 weeks post-inoculation (wpi) and in the spleen at 3 wpi, although the differences were not statistically significant. The liver/body weight ratio, indicative of liver enlargement, of both avian HEV-VA and avian HEV-prototype groups were significantly higher than that of the control group at 5 wpi. Overall, the avian HEV-VA strain still induces histological liver lesions even though it was isolated from a healthy chicken. The results also showed that intrahepatic inoculation of chickens with RNA transcripts of avian HEV infectious clone may serve as an alternative for live virus in animal pathogenicity studies.


Assuntos
DNA Complementar/metabolismo , Hepatite E/veterinária , Hepatite Viral Animal/virologia , Hepevirus/patogenicidade , Doenças das Aves Domésticas/virologia , Animais , Células Cultivadas , Galinhas , Clonagem Molecular , DNA Complementar/biossíntese , DNA Complementar/genética , Hepatite E/patologia , Hepatite E/virologia , Hepatite Viral Animal/fisiopatologia , Hepevirus/genética , Fígado/patologia , Fígado/virologia , Doenças das Aves Domésticas/patologia , Capuzes de RNA/genética , Organismos Livres de Patógenos Específicos , Virginia , Eliminação de Partículas Virais
5.
Vet Microbiol ; 139(3-4): 253-61, 2009 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19570623

RESUMO

Avian hepatitis E virus (avian HEV) is the primary causative agent of Hepatitis-Splenomegaly (HS) syndrome in chickens. Recently, a genetically unique strain of avian HEV, designated avian HEV-VA, was recovered from healthy chickens in Virginia. The objective of this study was to experimentally compare the pathogenicity of the prototype strain recovered from a chicken with HS syndrome and the avian HEV-VA strain in specific-pathogen-free chickens. An infectious stock of the avian HEV-VA strain was first generated and its infectivity titer determined in chickens. For the comparative pathogenesis study, 54 chickens of 6-week-old were assigned to 3 groups of 18 chickens each. The group 1 chickens were each intravenously inoculated with 5x10(2.5) 50% chicken infectious dose of the prototype strain. The group 2 received the same dose of the avian HEV-VA strain, and the group 3 served as negative controls. Six chickens from each group were necropsied at 2, 3 and 4 weeks post-inoculation (wpi). Most chickens in both inoculated groups seroconverted by 3wpi, and the mean anti-avian HEV antibody titers were higher for the prototype strain group than the avian HEV-VA strain group. There was no significant difference in the patterns of viremia and fecal virus shedding. Blood analyte profiles did not differ between treatment groups except for serum creatine phosphokinase levels which were higher for prototype avian HEV group than avian HEV-VA group. The hepatic lesion score was higher for the prototype strain group than the other two groups. The results indicated that the avian HEV-VA strain is only slightly attenuated compared to the prototype strain, suggesting that the full spectrum of HS syndrome is likely associated with other co-factors.


Assuntos
Galinhas , Hepatite Viral Animal/virologia , Hepevirus/patogenicidade , Doenças das Aves Domésticas/virologia , Animais , Anticorpos Antivirais/sangue , Fezes/virologia , Hepatite Viral Animal/patologia , Hepevirus/genética , Hepevirus/imunologia , Imunoglobulina G/sangue , Dados de Sequência Molecular , Doenças das Aves Domésticas/patologia , Organismos Livres de Patógenos Específicos , Suínos , Viremia/genética , Viremia/imunologia , Viremia/virologia , Virginia
6.
J Virol ; 83(1): 384-95, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18945785

RESUMO

Hepatitis E virus (HEV) is an important human pathogen, although little is known about its biology and replication. Comparative sequence analysis revealed a hypervariable region (HVR) with extensive sequence variations in open reading frame 1 of HEV. To elucidate the role of the HVR in HEV replication, we first constructed two HVR deletion mutants, hHVRd1 and hHVRd2, with in-frame deletion of amino acids (aa) 711 to 777 and 747 to 761 in the HVR of a genotype 1 human HEV replicon. Evidence of HEV replication was detected in Huh7 cells transfected with RNA transcripts from mutant hHVRd2, as evidenced by expression of enhanced green fluorescent protein. To confirm the in vitro results, we constructed three avian HEV mutants with various HVR deletions: mutants aHVRd1, with deletion of aa 557 to 585 (Delta557-585); aHVRd2 (Delta612-641); and aHVRd3 (Delta557-641). Chickens intrahepatically inoculated with capped RNA transcripts from mutants aHVRd1 and aHVRd2 developed active viral infection, as evidenced by seroconversion, viremia, and fecal virus shedding, although mutant aHVRd3, with complete HVR deletion, was apparently attenuated in chickens. To further verify the results, we constructed four additional HVR deletion mutants using the genotype 3 swine HEV as the backbone. Mutants sHVRd2 (Delta722-781), sHVRd3 (Delta735-765), and sHVRd4 (Delta712-765) were shown to tolerate deletions and were infectious in pigs intrahepatically inoculated with capped RNA transcripts from the mutants, whereas mutant sHVRd1 (Delta712-790), with a nearly complete HVR deletion, exhibited an attenuation phenotype in infected pigs. The data from these studies indicate that deletions in HVR do not abolish HEV infectivity in vitro or in vivo, although evidence for attenuation was observed for HEV mutants with a larger or nearly complete HVR deletion.


Assuntos
Deleção de Genes , Vírus da Hepatite E/patogenicidade , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Galinhas , Fezes/virologia , Genes Reporter , Proteínas de Fluorescência Verde/biossíntese , Anticorpos Anti-Hepatite/sangue , Hepatite E , Vírus da Hepatite E/genética , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , Suínos , Viremia , Virulência , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...