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











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 85(1): 98-111, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20962081

RESUMO

Varicella-zoster virus (VZV) is a neurotropic alphaherpesvirus. VZV infection of human dorsal root ganglion (DRG) xenografts in immunodeficient mice models the infection of sensory ganglia. We examined DRG infection with recombinant VZV (recombinant Oka [rOka]) and the following gE mutants: gEΔ27-90, gEΔCys, gE-AYRV, and gE-SSTT. gEΔ27-90, which lacks the gE domain that interacts with a putative receptor insulin-degrading enzyme (IDE), replicated as extensively as rOka, producing infectious virions and significant cytopathic effects within 14 days of inoculation. Since neural cells express IDE, the gE/IDE interaction was dispensable for VZV neurotropism. In contrast, gEΔCys, which lacks gE/gI heterodimer formation, was significantly impaired at early times postinfection; viral genome copy numbers increased slowly, and infectious virus production was not detected until day 28. Delayed replication was associated with impaired cell-cell spread in ganglia, similar to the phenotype of a gI deletion mutant (rOkaΔgI). However, at later time points, infection of satellite cells and other supportive nonneuronal cells resulted in extensive DRG tissue damage and cell loss such that cytopathic changes observed at day 70 were more severe than those for rOka-infected DRG. The replication of gE-AYRV, which is impaired for trans-Golgi network (TGN) localization, and the replication of gE-SSTT, which contains mutations in an acidic cluster, were equivalent to that of rOka, causing significant cytopathic effects and infectious virus production by day 14; genome copy numbers were equivalent to those of rOka. These experiments suggest that the gE interaction with cellular IDE, gE targeting to TGN sites of virion envelopment, and phosphorylation at SSTT are dispensable for VZV DRG infection, whereas the gE/gI interaction is critical for VZV neurovirulence.


Assuntos
Gânglios Sensitivos/patologia , Herpes Zoster/patologia , Herpesvirus Humano 3/patogenicidade , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Gânglios Sensitivos/metabolismo , Gânglios Sensitivos/virologia , Herpes Zoster/virologia , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/metabolismo , Humanos , Masculino , Camundongos , Camundongos SCID , Pele/metabolismo , Pele/patologia , Pele/virologia , Proteínas do Envelope Viral/genética , Virulência , Internalização do Vírus , Replicação Viral
2.
Curr Top Microbiol Immunol ; 342: 129-46, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20186616

RESUMO

The two VZV glycoproteins, gE and gI, are encoded by genes that are designated open reading frames, ORF67 and ORF68, located in the short unique region of the VZV genome. These proteins have homologs in the other alphaherpesviruses. Like their homologues, VZV gE and gI exhibit prominent co-localization in infected cells and form heterodimers. However, VZV gE is much larger than its homologues because it has a unique N-terminal domain, consisting of 188 amino acids that are not present in these other gene products. VZV gE also differs from the related gE proteins, in that it is essential for viral replication. Targeted mutations of gE that are compatible with VZV replication in cultured cells have varying phenotypes in skin and T-cell xenografts in the SCID mouse model of VZV pathogenesis in vivo. While gI is dispensable for growth in cultured cells in vitro, this glycoprotein is essential for VZV infection of differentiated human skin and T cells in vivo. The promoter regions of gE and gI are regulated by the cellular transactivator, specificity protein factor 1 (Sp1) in combination with the major VZV transactivator in reporter construct experiments and some Sp1 promoter elements are important for VZV virulence in vivo. Further analysis of VZV gE and gI functions and their interactions with other viral and host cell proteins are important areas for studies of VZV replication and pathogenesis.


Assuntos
Herpesvirus Humano 3/fisiologia , Regiões Promotoras Genéticas/fisiologia , Proteínas do Envelope Viral/fisiologia , Replicação Viral/fisiologia , Animais , Modelos Animais de Doenças , Herpesvirus Humano 3/genética , Camundongos , Camundongos SCID , Mutação , Fator de Transcrição Sp1/fisiologia , Transcrição Gênica , Proteínas do Envelope Viral/genética
3.
Proc Natl Acad Sci U S A ; 107(1): 282-7, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-19966293

RESUMO

Varicella-zoster virus (VZV) is an alphaherpesvirus that infects skin, lymphocytes, and sensory ganglia. VZV glycoprotein E (gE) has a unique N-terminal region (aa1-188), which is required for replication and includes domains involved in secondary envelopment, efficient cell-cell spread, and skin infection in vivo. The nonconserved N-terminal region also mediates binding to the insulin-degrading enzyme (IDE), which is proposed to be a VZV receptor. Using viral mutagenesis to make the recombinant rOka-DeltaP27-G90, we showed that amino acids in this region are required for gE/IDE binding in infected cells; this deletion reduced cell-cell spread in vitro and skin infection in vivo. However, a gE point mutation, linker insertions, and partial deletions in the aa27-90 region, and deletion of a large portion of the unique N-terminal region, aa52-187, had similar or more severe effects on VZV replication in vitro and in vivo without disrupting the gE/IDE interaction. VZV replication in T cells in vivo was not impaired by deletion of gE aa27-90, suggesting that these gE residues are not essential for VZV T cell tropism. However, the rOka-DeltaY51-P187 mutant failed to replicate in T cell xenografts as well as skin in vivo. VZV tropism for T cells and skin, which is necessary for its life cycle in the human host, requires this nonconserved region of the N-terminal region of VZV gE.


Assuntos
Varicela/fisiopatologia , Herpesvirus Humano 3/patogenicidade , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular Tumoral , Varicela/metabolismo , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiologia , Humanos , Camundongos , Camundongos SCID , Mutagênese , Estrutura Terciária de Proteína , Pele/citologia , Pele/patologia , Pele/virologia , Dermatopatias/patologia , Dermatopatias/virologia , Transplante de Pele , Linfócitos T/imunologia , Linfócitos T/virologia , Transplante Heterólogo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Replicação Viral/genética
4.
J Virol ; 83(1): 228-40, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18945783

RESUMO

Varicella-zoster virus (VZV) glycoprotein E (gE) is the most abundant glycoprotein in infected cells and, in contrast to those of other alphaherpesviruses, is essential for viral replication. The gE ectodomain contains a unique N-terminal region required for viral replication, cell-cell spread, and secondary envelopment; this region also binds to the insulin-degrading enzyme (IDE), a proposed VZV receptor. To identify new functional domains of the gE ectodomain, the effect of mutagenesis of the first cysteine-rich region of the gE ectodomain (amino acids 208 to 236) was assessed using VZV cosmids. Deletion of this region was compatible with VZV replication in vitro, but cell-cell spread of the rOka-DeltaCys mutant was reduced significantly. Deletion of the cysteine-rich region abolished the binding of the mutant gE to gI but not to IDE. Preventing gE binding to gI altered the pattern of gE expression at the plasma membrane of infected cells and the posttranslational maturation of gI and its incorporation into viral particles. In contrast, deletion of the first cysteine-rich region did not affect viral entry into human tonsil T cells in vitro or into melanoma cells infected with cell-free VZV. These experiments demonstrate that gE/gI heterodimer formation is essential for efficient cell-cell spread and incorporation of gI into viral particles but that it is dispensable for infectious varicella-zoster virion formation and entry into target cells. Blocking gE binding to gI resulted in severe impairment of VZV infection of human skin xenografts in SCIDhu mice in vivo, documenting the importance of cell fusion mediated by this complex for VZV virulence in skin.


Assuntos
Herpesvirus Humano 3/fisiologia , Herpesvirus Humano 3/patogenicidade , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Montagem de Vírus , Internalização do Vírus , Animais , Linhagem Celular , Herpes Zoster , Humanos , Camundongos , Camundongos SCID , Ligação Proteica , Deleção de Sequência , Transplante Heterólogo , Virulência
5.
J Virol ; 81(19): 10258-67, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17634217

RESUMO

Varicella-zoster virus (VZV) glycoprotein E (gE) is essential for viral replication and is involved in cell-to-cell spread, secondary envelopment, and entry. We created a set of mutations in the gE promoter to investigate the role of viral and cellular transcriptional factors in regulation of the gE promoter. Deletion or point mutation of the two Sp1 sites in the gE promoter abolished Sp1 binding and IE62-mediated transactivation of the gE promoter in vitro. Incorporation of the deletion or the point mutations disrupting both of the Sp1 binding sites into the VZV genome was not compatible with viral replication. A point mutation altering the atypical Sp1 binding site was lethal, while altering the second site impaired VZV replication significantly, indicating functional differences between the two Sp1 binding sites. Deletions in the gE promoter that abolished putative binding sites for cellular transcriptional factors other than Sp1, identified by bioinformatics analysis, were inserted in the VZV genome. Replication of the viruses with mutations of the gE promoter did not differ from control recombinants in melanoma cells or primary human tonsil T cells in vitro. These deletions did not affect infection of human skin xenografts in SCIDhu mice. These results indicate that Sp1 is required for IE62-mediated transactivation of the gE promoter and that this transcriptional factor appears to be the only cellular factor essential for regulation of the gE promoter.


Assuntos
Regulação Viral da Expressão Gênica , Glicoproteínas/genética , Herpesvirus Humano 3/genética , Proteínas Imediatamente Precoces/metabolismo , Fator de Transcrição Sp1/metabolismo , Transativadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Replicação Viral/genética , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular Tumoral , Herpesvirus Humano 3/fisiologia , Humanos , Melanoma , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Deleção de Sequência , Pele/virologia , Linfócitos T/virologia , Ativação Transcricional
6.
J Virol ; 80(19): 9481-96, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973553

RESUMO

Varicella-zoster virus (VZV) glycoprotein E (gE) is a multifunctional protein important for cell-cell spread, envelopment, and possibly entry. In contrast to other alphaherpesviruses, gE is essential for VZV replication. Interestingly, the N-terminal region of gE, comprised of amino acids 1 to 188, was shown not to be conserved in the other alphaherpesviruses by bioinformatics analysis. Mutational analysis was performed to investigate the functions associated with this unique gE N-terminal region. Linker insertions, serine-to-alanine mutations, and deletions were introduced in the gE N-terminal region in the VZV genome, and the effects of these mutations on virus replication and cell-cell spread, gE trafficking and localization, virion formation, and replication in vivo in the skin were analyzed. In summary, mutagenesis of the gE N-terminal region identified a new functional region in the VZV gE ectodomain essential for cell-cell spread and the pathogenesis of VZV skin tropism and demonstrated that different subdomains of the unique N-terminal region had specific roles in viral replication, cell-cell spread, and secondary envelopment.


Assuntos
Glicoproteínas/metabolismo , Herpesvirus Humano 3/fisiologia , Herpesvirus Humano 3/patogenicidade , Dermatopatias Infecciosas/metabolismo , Dermatopatias Infecciosas/virologia , Proteínas Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/ultraestrutura , Herpesvirus Humano 3/ultraestrutura , Humanos , Cinética , Camundongos , Camundongos SCID , Microscopia Eletrônica , Dados de Sequência Molecular , Mutação/genética , Alinhamento de Sequência , Dermatopatias Infecciosas/patologia , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/ultraestrutura , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Development ; 133(18): 3723-32, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936075

RESUMO

Canonical Wnt/beta-catenin signaling regulates the activation of the myogenic determination gene Myf5 at the onset of myogenesis, but the underlying molecular mechanism is unknown. Here, we report that the Wnt signal is transduced in muscle progenitor cells by at least two Frizzled (Fz) receptors (Fz1 and/or Fz6), through the canonical beta-catenin pathway, in the epaxial domain of newly formed somites. We show that Myf5 activation is dramatically reduced by blocking the Wnt/beta-catenin pathway in somite progenitor cells, whereas expression of activated beta-catenin is sufficient to activate Myf5 in somites but not in the presomitic mesoderm. In addition, we identified Tcf/Lef sequences immediately 5' to the Myf5 early epaxial enhancer. These sites determine the correct spatiotemporal expression of Myf5 in the epaxial domain of the somite, mediating the synergistic action of the Wnt/beta-catenin and the Shh/Gli pathways. Taken together, these results demonstrate that Myf5 is a direct target of Wnt/beta-catenin, and that its full activation requires a cooperative interaction between the canonical Wnt and the Shh/Gli pathways in muscle progenitor cells.


Assuntos
Fatores de Transcrição Kruppel-Like/genética , Fator Regulador Miogênico 5/genética , Somitos/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Indução Embrionária/genética , Indução Embrionária/fisiologia , Elementos Facilitadores Genéticos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Globinas/genética , Globinas/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Fator Regulador Miogênico 5/metabolismo , Células NIH 3T3 , Ligação Proteica , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Somitos/citologia , Fatores de Transcrição TCF/genética , Fatores de Transcrição TCF/metabolismo , Ativação Transcricional/genética , Proteínas Wnt/metabolismo , Proteína GLI1 em Dedos de Zinco , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA