Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 92(11)2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29563288

RESUMO

Varicella-zoster virus (VZV) is the skin-tropic human alphaherpesvirus responsible for both varicella-zoster and herpes zoster. Varicella-zoster and herpes zoster skin lesions have similar morphologies, but herpes zoster occurs disproportionally in older individuals and is often associated with a more extensive local rash and severe zoster-related neuralgia. We hypothesized that skin aging could also influence the outcome of the anterograde axonal transport of VZV to skin. We utilized human skin xenografts maintained in immunodeficient (SCID) mice to study VZV-induced skin pathology in vivo in fetal and adult skin xenografts. Here we found that VZV replication is enhanced in skin from older compared to younger adults, correlating with clinical observations. In addition to measures of VZV infection, we examined the expression of type I interferon (IFN) pathway components in adult skin and investigated elements of the cutaneous proliferative and inflammatory response to VZV infection in vivo Our results demonstrated that VZV infection of adult skin triggers intrinsic IFN-mediated responses such as we have described in VZV-infected fetal skin xenografts, including MxA as well as promyelocytic leukemia protein (PML), in skin cells surrounding lesions. Further, we observed that VZV elicited altered cell signaling and proliferative and inflammatory responses that are involved in wound healing, driven by follicular stem cells. These cellular changes are consistent with VZV-induced activation of STAT3 and suggest that VZV exploits the wound healing process to ensure efficient delivery of the virus to keratinocytes. Adult skin xenografts offer an approach to further investigate VZV-induced skin pathologies in vivoIMPORTANCE Varicella-zoster virus (VZV) is the agent responsible for both varicella-zoster and herpes zoster. Herpes zoster occurs disproportionally in older individuals and is often associated with a more extensive local rash and severe zoster-related neuralgia. To examine the effect of skin aging on VZV skin lesions, we utilized fetal and adult human skin xenografts maintained in immunodeficient (SCID) mice. We measured VZV-induced skin pathology, examined the expression of type I interferon (IFN) pathway components in adult skin, and investigated elements of the cutaneous proliferative and inflammatory response to VZV infection in vivo Our results demonstrate that characteristics of aging skin are preserved in xenografts; that VZV replication is enhanced in skin from older compared to younger adults, correlating with clinical observations; and that VZV infection elicits altered cell signaling and inflammatory responses. Adult skin xenografts offer an approach to further investigate VZV-induced skin pathologies in vivo.


Assuntos
Envelhecimento/patologia , Herpesvirus Humano 3/crescimento & desenvolvimento , Fator de Transcrição STAT3/metabolismo , Infecção pelo Vírus da Varicela-Zoster/patologia , Replicação Viral/fisiologia , Adulto , Fatores Etários , Idoso , Animais , Modelos Animais de Doenças , Ativação Enzimática , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Proteína da Leucemia Promielocítica/metabolismo , Pele/virologia , Dermatopatias/patologia , Dermatopatias/virologia , Transplante de Pele , Transplante Heterólogo , Infecção pelo Vírus da Varicela-Zoster/virologia , Cicatrização/fisiologia
2.
J Virol ; 87(7): 4075-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23345513

RESUMO

The varicella-zoster virus (VZV) ORF61 protein is necessary for normal replication in vitro and virulence in human skin xenografts in the severe combined immunodeficiency mouse model in vivo. These experiments identify a hydrophobic domain that mediates ORF61 self-interaction. While not needed to inhibit host cell defenses, disruption of this domain (residues 250 to 320) severely impairs VZV growth, transactivation of the immediate early 63 and glycoprotein E genes, and the pathogenesis of VZV skin infection in vivo.


Assuntos
Varicela/fisiopatologia , Herpesvirus Humano 3/metabolismo , Pele/virologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/patogenicidade , Humanos , Interações Hidrofóbicas e Hidrofílicas , Proteínas Imediatamente Precoces/metabolismo , Immunoblotting , Imunoprecipitação , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Análise de Sequência de DNA , Pele/patologia , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Replicação Viral/genética
3.
Proc Natl Acad Sci U S A ; 109(2): 600-5, 2012 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-22190485

RESUMO

Varicella-zoster virus (VZV) is a human α-herpesvirus that causes varicella (chickenpox) during primary infection and zoster (shingles) upon reactivation. Like other viruses, VZV must subvert the intrinsic antiviral defenses of differentiated human cells to produce progeny virions. Accordingly, VZV inhibits the activation of the cellular transcription factors IFN regulatory factor 3 (IRF3) and signal transducers and activators of transcription 1 (STAT1), thereby downregulating antiviral factors, including IFNs. Conversely, in this study, we found that VZV triggers STAT3 phosphorylation in cells infected in vitro and in human skin xenografts in SCID mice in vivo and that STAT3 activation induces the anti-apoptotic protein survivin. Small-molecule inhibitors of STAT3 phosphorylation and survivin restrict VZV replication in vitro, and VZV infection of skin xenografts in vivo is markedly impaired by the administration of the phospho-STAT3 inhibitor S3I-201. STAT3 and survivin are required for malignant transformation caused by γ-herpesviruses, such as Kaposi's sarcoma virus. We show that STAT3 activation is also critical for VZV, a nononcogenic herpesvirus, via a survivin-dependent mechanism. Furthermore, STAT3 activation is critical for the life cycle of the virus because VZV skin infection is necessary for viral transmission and persistence in the human population. Therefore, we conclude that takeover of this major cell-signaling pathway is necessary, independent of cell transformation, for herpesvirus pathogenesis and that STAT3 activation and up-regulation of survivin is a common mechanism important for the pathogenesis of lytic as well as tumorigenic herpesviruses.


Assuntos
Herpesvirus Humano 3/fisiologia , Proteínas Inibidoras de Apoptose/genética , Fator de Transcrição STAT3/genética , Ativação Transcricional/fisiologia , Replicação Viral/fisiologia , Ácidos Aminossalicílicos/farmacologia , Animais , Benzenossulfonatos/farmacologia , Citometria de Fluxo , Humanos , Medições Luminescentes , Camundongos , Camundongos SCID , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Pele/metabolismo , Pele/virologia , Survivina , Ativação Transcricional/genética , Replicação Viral/genética
4.
J Neurovirol ; 17(6): 570-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22161683

RESUMO

Varicella-zoster virus (VZV) is a medically important human alphaherpesvirus. Investigating pathogenic mechanisms that contribute to VZV neurovirulence are made difficult by a marked host restriction. Our approach to investigating VZV neurotropism and neurovirulence has been to develop a mouse-human xenograft model in which human dorsal root ganglia (DRG) are maintained in severe compromised immunodeficient (SCID) mice. In this review, we will describe our key findings using this model in which we have demonstrated that VZV infection of SCID DRG xenograft results in rapid and efficient spread, enabled by satellite cell infection and polykaryon formation, which facilitates robust viral replication and release of infectious virus. In neurons that persist following this acute replicative phase, VZV genomes are present at low frequency with limited gene transcription and no protein synthesis, a state that resembles VZV latency in the natural human host. VZV glycoprotein I and interaction between glycoprotein I and glycoprotein E are critical for neurovirulence. Our work demonstrates that the DRG model can reveal characteristics about VZV replication and long-term persistence of latent VZV genomes in human neuronal tissues, in vivo, in an experimental system that may contribute to our knowledge of VZV neuropathogenesis.


Assuntos
Varicela/virologia , Regulação Viral da Expressão Gênica , Herpes Zoster/virologia , Herpesvirus Humano 3/genética , Células Receptoras Sensoriais/virologia , Proteínas do Envelope Viral/genética , Replicação Viral/genética , Animais , Gânglios Espinais/metabolismo , Gânglios Espinais/transplante , Gânglios Espinais/virologia , Herpesvirus Humano 3/patogenicidade , Humanos , Camundongos , Camundongos SCID , Células Satélites Perineuronais/virologia , Células Receptoras Sensoriais/metabolismo , Pele/virologia , Transcrição Gênica , Transplante Heterólogo , Proteínas do Envelope Viral/metabolismo , Virulência/genética , Latência Viral/genética
5.
PLoS Pathog ; 7(2): e1001266, 2011 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-21304940

RESUMO

The herpesviruses, like most other DNA viruses, replicate in the host cell nucleus. Subnuclear domains known as promyelocytic leukemia protein nuclear bodies (PML-NBs), or ND10 bodies, have been implicated in restricting early herpesviral gene expression. These viruses have evolved countermeasures to disperse PML-NBs, as shown in cells infected in vitro, but information about the fate of PML-NBs and their functions in herpesvirus infected cells in vivo is limited. Varicella-zoster virus (VZV) is an alphaherpesvirus with tropism for skin, lymphocytes and sensory ganglia, where it establishes latency. Here, we identify large PML-NBs that sequester newly assembled nucleocapsids (NC) in neurons and satellite cells of human dorsal root ganglia (DRG) and skin cells infected with VZV in vivo. Quantitative immuno-electron microscopy revealed that these distinctive nuclear bodies consisted of PML fibers forming spherical cages that enclosed mature and immature VZV NCs. Of six PML isoforms, only PML IV promoted the sequestration of NCs. PML IV significantly inhibited viral infection and interacted with the ORF23 capsid surface protein, which was identified as a target for PML-mediated NC sequestration. The unique PML IV C-terminal domain was required for both capsid entrapment and antiviral activity. Similar large PML-NBs, termed clastosomes, sequester aberrant polyglutamine (polyQ) proteins, such as Huntingtin (Htt), in several neurodegenerative disorders. We found that PML IV cages co-sequester HttQ72 and ORF23 protein in VZV infected cells. Our data show that PML cages contribute to the intrinsic antiviral defense by sensing and entrapping VZV nucleocapsids, thereby preventing their nuclear egress and inhibiting formation of infectious virus particles. The efficient sequestration of virion capsids in PML cages appears to be the outcome of a basic cytoprotective function of this distinctive category of PML-NBs in sensing and safely containing nuclear aggregates of aberrant proteins.


Assuntos
Capsídeo/metabolismo , Herpesvirus Humano 3/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Corpos de Inclusão Viral/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Células Cultivadas , Citoproteção/fisiologia , Embrião de Mamíferos , Herpesvirus Humano 3/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Corpos de Inclusão Viral/virologia , Corpos de Inclusão Intranuclear/virologia , Camundongos , Camundongos SCID , Proteínas Nucleares/fisiologia , Proteína da Leucemia Promielocítica , Ligação Proteica , Multimerização Proteica/fisiologia , Fatores de Transcrição/fisiologia , Transplante Heterólogo , Proteínas Supressoras de Tumor/fisiologia
6.
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
7.
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
8.
J Virol ; 84(7): 3421-30, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20106930

RESUMO

Varicella-zoster virus (VZV) causes varicella and establishes latency in sensory nerve ganglia, but the characteristics of VZV latency are not well defined. Immunohistochemical detection of the VZV immediate-early 63 (IE63) protein in ganglion neurons has been described, but there are significant discrepancies in estimates of the frequency of IE63-positive neurons, varying from a rare event to abundant expression. We examined IE63 expression in cadaver ganglia using a high-potency rabbit anti-IE63 antibody and corresponding preimmune serum. Using standard immunohistochemical techniques, we evaluated 10 ganglia that contained VZV DNA from seven individuals. These experiments showed that neuronal pigments were a confounding variable; however, by examining sections coded to prevent investigator bias and applying statistical analysis, we determined that IE63 protein, if present, is in a very small proportion of neurons (<2.8%). To refine estimates of IE63 protein abundance, we modified our protocol by incorporating a biological stain to exclude the pigment signal and evaluated 27 ganglia from 18 individuals. We identified IE63 protein in neurons within only one ganglion, in which VZV glycoprotein E and an immune cell infiltrate were also demonstrated. Antigen preservation was shown by detection of neuronal synaptophysin. These data provide evidence that the expression of IE63 protein, which has been referred to as a latency-associated protein, is rare. Refining estimates of VZV protein expression in neurons is important for developing a hypothesis about the mechanisms by which VZV latency may be maintained.


Assuntos
Gânglios Sensitivos/virologia , Proteínas Imediatamente Precoces/análise , Neurônios/virologia , Proteínas do Envelope Viral/análise , Latência Viral , Adulto , Idoso , Idoso de 80 Anos ou mais , DNA Viral/análise , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Melaninas/análise , Pessoa de Meia-Idade , Sinaptofisina/análise
9.
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
10.
J Virol ; 83(15): 7495-506, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19474103

RESUMO

Glycoprotein B (gB), the most conserved protein in the family Herpesviridae, is essential for the fusion of viral and cellular membranes. Information about varicella-zoster virus (VZV) gB is limited, but homology modeling showed that the structure of VZV gB was similar to that of herpes simplex virus (HSV) gB, including the putative fusion loops. In contrast to HSV gB, VZV gB had a furin recognition motif ([R]-X-[KR]-R-|-X, where | indicates the position at which the polypeptide is cleaved) at residues 491 to 494, thought to be required for gB cleavage into two polypeptides. To investigate their contribution, the putative primary fusion loop or the furin recognition motif was mutated in expression constructs and in the context of the VZV genome. Substitutions in the primary loop, W180G and Y185G, plus the deletion mutation Delta491RSRR494 and point mutation 491GSGG494 in the furin recognition motif did not affect gB expression or cellular localization in transfected cells. Infectious VZV was recovered from parental Oka (pOka)-bacterial artificial chromosomes that had either the Delta491RSRR494 or 491GSGG494 mutation but not the point mutations W180G and Y185G, demonstrating that residues in the primary loop of gB were essential but gB cleavage was not required for VZV replication in vitro. Virion morphology, protein localization, plaque size, and replication were unaffected for the pOka-gBDelta491RSRR494 or pOka-gB491GSGG494 virus compared to pOka in vitro. However, deletion of the furin recognition motif caused attenuation of VZV replication in human skin xenografts in vivo. This is the first evidence that cleavage of a herpesvirus fusion protein contributes to viral pathogenesis in vivo, as seen for fusion proteins in other virus families.


Assuntos
Varicela/virologia , Furina/metabolismo , Herpesvirus Humano 3/patogenicidade , Mutagênese , Pele/virologia , Proteínas do Envelope Viral/genética , Replicação Viral , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Varicela/metabolismo , Varicela/patologia , Herpesvirus Humano 3/química , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Mutação , Ligação Proteica , Alinhamento de Sequência , Pele/metabolismo , Pele/patologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
11.
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
12.
J Virol ; 82(20): 10231-46, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18684828

RESUMO

The assembly of herpesvirus capsids is a complex process involving interactions of multiple proteins in the cytoplasm and in the nucleus. Based on comparative genome analyses, varicella-zoster virus (VZV) open reading frame 23 (ORF23) encodes a conserved capsid protein, referred to as VP26 (UL35) in other alphaherpesviruses. Mutagenesis using a VZV bacterial artificial chromosome system showed that ORF23 was dispensable for replication in vitro. However, the absence of ORF23 disrupted capsid assembly in a melanoma cell line. Expression of ORF23 as a red fluorescent protein (RFP) fusion protein appeared to have a dominant negative effect on replication that was rescued by ORF23 expression from a nonnative site in the VZV genome. In contrast to its VP26 homolog, ORF23 has an intrinsic nuclear localization capacity that was mapped to an SRSRVV motif at residues 229 to 234 in the extreme C terminus of ORF23. In addition, coexpression with ORF23 resulted in nuclear import of the major capsid protein, ORF40. VZV ORF33.5 also translocated ORF40, which may provide a redundant mechanism in vitro but appears insufficient to overcome the dominant negative effect of the monomeric RFP-ORF23 (mRFP23) fusion protein. ORF23 was required for VZV infection of human skin xenografts, indicating that ORF33.5 does not compensate for lack of ORF23 in vivo. These observations suggest a model of VZV capsid assembly in which nuclear transport of the major capsid protein and associated proteins requires ORF23 during VZV replication in the human host. If so, ORF23 expression could be a target for a novel antiviral drug against VZV.


Assuntos
Proteínas do Capsídeo/metabolismo , Infecções por Herpesviridae/fisiopatologia , Herpesvirus Humano 3/fisiologia , Fases de Leitura Aberta , Dermatopatias Infecciosas/fisiopatologia , Replicação Viral/fisiologia , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Linhagem Celular , Herpesvirus Humano 3/genética , Humanos , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Dermatopatias Infecciosas/virologia , Transplante de Pele , Transplante Heterólogo , Montagem de Vírus
13.
J Virol ; 82(12): 5825-34, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18400847

RESUMO

The gene cluster composed of varicella-zoster virus (VZV) open reading frame 9 (ORF9) to ORF12 encodes four putative tegument proteins and is highly conserved in most alphaherpesviruses. In these experiments, the genes within this cluster were deleted from the VZV parent Oka (POKA) individually or in combination, and the consequences for VZV replication were evaluated with cultured cells in vitro and with human skin xenografts in SCID mice in vivo. As has been reported for ORF10, ORF11 and ORF12 were dispensable for VZV replication in melanoma and human embryonic fibroblast cells. In contrast, deletion of ORF9 was incompatible with the recovery of infectious virus. ORF9 localized to the virion tegument and formed complexes with glycoprotein E, which is an essential protein, in VZV-infected cells. Recombinants lacking ORF10 and ORF11 (POKADelta10/11), ORF11 and ORF12 (POKADelta11/12), or ORF10, ORF11 and ORF12 (POKADelta10/11/12) were viable in cultured cells. Their growth kinetics did not differ from those of POKA, and nucleocapsid formation and virion assembly were not disrupted. In addition, these deletion mutants showed no differences compared to POKA in infectivity levels for primary human tonsil T cells. Deletion of ORF12 had no effect on skin infection, whereas replication of POKADelta11, POKADelta10/11, and POKADelta11/12 was severely reduced, and no virus was recovered from skin xenografts inoculated with POKADelta10/11/12. These results indicate that with the exception of ORF9, the individual genes within the ORF9-to-ORF12 gene cluster are dispensable and can be deleted simultaneously without any apparent effect on VZV replication in vitro but that the ORF10-to-ORF12 cluster is essential for VZV virulence in skin in vivo.


Assuntos
Herpesvirus Humano 3/patogenicidade , Família Multigênica , Fases de Leitura Aberta , Dermatopatias Infecciosas/etiologia , Replicação Viral , Animais , Linhagem Celular Tumoral , Células Cultivadas , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiologia , Humanos , Imuno-Histoquímica , Masculino , Melanoma/patologia , Melanoma/ultraestrutura , Melanoma/virologia , Camundongos , Camundongos SCID , Transplante de Pele , Transplante Heterólogo
14.
Proc Natl Acad Sci U S A ; 104(35): 14086-91, 2007 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-17709745

RESUMO

Varicella-zoster virus (VZV) causes varicella, establishes latency in sensory ganglia, and reactivates as herpes zoster. Human dorsal root ganglia (DRGs) xenografts in immunodeficient mice provide a model for evaluating VZV neuropathogenesis. Our investigation of the role of glycoprotein I (gI), which is dispensable in vitro, examines the functions of a VZV gene product during infection of human neural cells in vivo. Whereas intact recombinant Oka (rOka) initiated a short replicative phase followed by persistence in DRGs, the gI deletion mutant, rOkaDeltagI, showed prolonged replication with no transition to persistence up to 70 days after infection. Only a few varicella-zoster nucleocapsids and cytoplasmic virions were observed in neurons, and the major VZV glycoprotein, gE, was retained in the rough endoplasmic reticulum in the absence of gI. VZV neurotropism was not disrupted when DRG xenografts were infected with rOka mutants lacking gI promoter elements that bind cellular transactivators, specificity factor 1 (Sp1) and upstream stimulatory factor (USF). Because gI is essential and Sp1 and USF contribute to VZV pathogenesis in skin and T cells in vivo, these DRG experiments indicate that the genetic requirements for VZV infection are less stringent in neural cells in vivo. The observations demonstrate that gI is important for VZV neurotropism and suggest that a strategy to reduce neurovirulence by deleting gI could prolong active infection in human DRGs.


Assuntos
Varicela/imunologia , Gânglios Espinais/virologia , Herpesvirus Humano 3/fisiologia , Proteínas do Envelope Viral/genética , Animais , Varicela/patologia , DNA Viral/genética , Gânglios Espinais/patologia , Gânglios Espinais/transplante , Deleção de Genes , Genoma Viral , Herpesvirus Humano 3/enzimologia , Herpesvirus Humano 3/genética , Humanos , Camundongos , Fases de Leitura Aberta , Regiões Promotoras Genéticas , Recombinação Genética , Transcrição Gênica , Transplante Heterólogo , Proteínas Virais/genética
15.
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
16.
J Virol ; 80(7): 3238-48, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16537591

RESUMO

The open reading frame 10 (ORF10) of varicella-zoster virus (VZV) encodes a tegument protein that enhances transactivation of VZV genes and has homology to herpes simplex virus type 1 (HSV-1) VP16. While VP16 is essential for HSV replication, ORF10 is dispensable for vaccine OKA (VOKA) growth in vitro. We used parent OKA (POKA) cosmids to delete ORF10, producing POKA delta10; point mutations that disrupted the acidic activation domain and the putative motif for binding human cellular factor 1 (HCF-1) in ORF10 protein yielded POKA10-Phe28Ala, POKA10-Phe28Ser, and POKA10-mHCF viruses. Deleting ORF10 or mutating these two functional domains had no effect on VZV replication, immediate-early gene transcription, or virion assembly in vitro. However, deleting ORF10 reduced viral titers and the extent of cutaneous lesions significantly in SCIDhu skin xenografts in vivo compared to POKA. Epidermal cells infected with POKA delta10 had significantly fewer DNA-containing nucleocapsids and complete virions compared to POKA; extensive aggregates of intracytoplasmic viral particles were also observed. Altering the activation or the putative HCF-1 domains of ORF10 protein had no consequences for VZV replication in vivo. Thus, the decreased pathogenic potential of POKA delta10 in skin could not be attributed to absence of these ORF10 protein functions. In contrast to skin cells, deleting ORF10 did not impair VZV T-cell tropism in vivo, as assessed by infectious virus yields. We conclude that ORF10 protein is required for efficient VZV virion assembly and is a specific determinant of VZV virulence in epidermal and dermal cells in vivo.


Assuntos
Herpesvirus Humano 3/genética , Herpesvirus Humano 3/patogenicidade , Fases de Leitura Aberta , Pele/virologia , Linfócitos T/virologia , Animais , Cosmídeos , Humanos , Imuno-Histoquímica , Masculino , Melanoma/patologia , Melanoma/ultraestrutura , Melanoma/virologia , Camundongos , Camundongos SCID , Mutação Puntual , Recombinação Genética , Pele/patologia , Pele/ultraestrutura , Fatores de Tempo , Transplante Heterólogo , Vacinas Atenuadas/imunologia , Vírion/ultraestrutura , Virulência , Replicação Viral
17.
J Virol ; 79(20): 12921-33, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16188994

RESUMO

The pathogenesis of varicella-zoster virus (VZV) involves a cell-associated viremia during which infectious virus is carried from sites of respiratory mucosal inoculation to the skin. We now demonstrate that VZV infection of T cells is associated with robust virion production and modulation of the apoptosis and interferon pathways within these cells. The VZV serine/threonine protein kinase encoded by ORF66 is essential for the efficient replication of VZV in T cells. Preventing ORF66 protein expression by stop codon insertion (pOka66S) impaired the growth of the parent Oka (pOka) strain in T cells in SCID-hu T-cell xenografts in vivo and reduced formation of VZV virions. The lack of ORF66 protein also increased the susceptibility of infected T cells to apoptosis and reduced the capacity of the virus to interfere with induction of the interferon (IFN) signaling pathway following exposure to IFN-gamma. However, preventing ORF66 protein expression only slightly reduced growth in melanoma cells in culture and did not diminish virion formation in these cells. The pOka66S virus showed only a slight defect in growth in SCID-hu skin implants compared with intact pOka. These observations suggest that the ORF66 kinase plays a unique role during infection of T cells and supports VZV T-cell tropism by contributing to immune evasion and enhancing survival of infected T cells.


Assuntos
Herpes Zoster/virologia , Herpesvirus Humano 3/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Células Cultivadas , Herpesvirus Humano 3/patogenicidade , Humanos , Masculino , Camundongos , Camundongos SCID , Fases de Leitura Aberta , Linfócitos T/virologia , Virulência , Replicação Viral
18.
Proc Natl Acad Sci U S A ; 102(18): 6490-5, 2005 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-15851670

RESUMO

Varicella-zoster virus (VZV) causes varicella and establishes latency in sensory ganglia. VZV reactivation results in herpes zoster. We developed a model using human dorsal root ganglion (DRG) xenografts in severe combined immunodeficient (SCID) mice to investigate VZV infection of differentiated neurons and satellite cells in vivo. DRG engrafted under the kidney capsule and contained neurons and satellite cells within a typical DRG architecture. VZV clinical isolates infected the neurons within DRG. At 14 days postinfection, VZ virions were detected by electron microscopy in neuronal cell nuclei and cytoplasm but not in satellite cells. The VZV genome copy number was 7.1 x 10(7) to 8.0 x 10(8) copies per 10(5) cells, and infectious virus was recovered. This initial phase of viral replication was followed within 4-8 weeks by a transition to VZV latency, characterized by the absence of infectious virus release, the cessation of virion assembly, and a reduction in VZV genome copies to 3.7 x 10(5) to 4.7 x 10(6) per 10(5) cells. VZV persistence in DRG was achieved without any requirement for VZV-specific adaptive immunity and was associated with continued transcription of the ORF63 regulatory gene. The live attenuated varicella vaccine virus exhibited the same pattern of short-term replication, persistence of viral DNA, and prominent ORF63 transcription as the clinical isolates. VZV-infected T cells transferred virus from the circulation into DRG, suggesting that VZV lymphotropism facilitates its neurotropism. DRG xenografts may be useful for investigating neuropathogenic mechanisms of other human viruses.


Assuntos
Varicela/patologia , Gânglios Espinais/virologia , Genoma Viral , Herpesvirus Humano 3/genética , Animais , Primers do DNA , Gânglios Espinais/patologia , Gânglios Espinais/transplante , Herpesvirus Humano 3/fisiologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos SCID , Microscopia Eletrônica de Transmissão , Neurônios/ultraestrutura , Neurônios/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Satélites Perineuronais/ultraestrutura , Células Satélites Perineuronais/virologia , Linfócitos T/virologia , Transplante Heterólogo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vírion/fisiologia , Vírion/ultraestrutura , Replicação Viral/fisiologia
19.
J Virol ; 79(8): 4819-27, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15795267

RESUMO

Although genes related to varicella-zoster virus (VZV) open reading frame 35 (ORF35) are conserved in the herpesviruses, information about their contributions to viral replication and pathogenesis is limited. Using a VZV cosmid system, we deleted ORF35 to produce two null mutants, designated rOkaDelta35(#1) and rOkaDelta35(#2), and replaced ORF35 at a nonnative site, generating two rOkaDelta35/35@Avr mutants. ORF35 Flag-tagged recombinants were made by inserting ORF35-Flag at the nonnative Avr site as the only copy of ORF35, yielding rOkaDelta35/35Flag@Avr, or as a second copy, yielding rOka35Flag@Avr. Replication of rOkaDelta35 viruses was diminished in melanoma and Vero cells in a 6-day analysis of growth kinetics. Plaque sizes of rOkaDelta35 mutants were significantly smaller than those of rOka in melanoma cells. Infection of melanoma cells with rOkaDelta35 mutants was associated with disrupted cell fusion and polykaryocyte formation. The small plaque phenotype was not corrected by growth of rOkaDelta35 mutants in melanoma cells expressing the major VZV glycoprotein E, gE. The rOkaDelta35/35@Avr viruses displayed growth kinetics and plaque morphologies that were indistinguishable from those of rOka. Analysis with ORF35-Flag recombinants showed that the ORF35 gene product localized predominantly to the nuclei of infected cells. Evaluations in the SCIDhu mouse model demonstrated that ORF35 was required for efficient VZV infection of skin and T-cell xenografts, although the decrease in infectivity was most significant in skin. These mutagenesis experiments indicated that ORF35 was dispensable for VZV replication, but deleting ORF35 diminished growth in cultured cells and was associated with attenuated VZV infection of differentiated human skin and T cells in vivo.


Assuntos
Herpesvirus Humano 3/genética , Fases de Leitura Aberta/genética , Pele/citologia , Pele/virologia , Linfócitos T/virologia , Sequência de Bases , Diferenciação Celular , Sequência Conservada , Primers do DNA , Genes Reporter , Herpesviridae/genética , Humanos , Mutagênese , Deleção de Sequência , Proteínas Virais/genética
20.
J Virol ; 78(23): 13293-305, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15542680

RESUMO

The protein product of varicella-zoster virus (VZV) ORF47 is a serine/threonine protein kinase and tegument component. Evaluation of two recombinants of the Oka strain, rOka47DeltaC, with a C-terminal truncation of ORF47, and rOka47D-N, with a point mutation in the conserved kinase motif, showed that ORF47 kinase function was necessary for optimal VZV replication in human skin xenografts in SCID mice but not in cultured cells. We now demonstrate that rOka47DeltaC and rOka47D-N mutants do not infect human T-cell xenografts. Differences in the growth of kinase-defective ORF47 mutants allowed an examination of requirements for VZV pathogenesis in skin and T cells in vivo. Although virion assembly was reduced and no virion transport to cell surfaces was observed, epidermal cell fusion persisted, and VZV polykaryocytes were generated by rOka47DeltaC and rOka47D-N in skin. Virion assembly was also impaired in vitro, but VZV-induced cell fusion continued to cause syncytia in cultured cells infected with rOka47DeltaC or rOka47D-N. Intracellular trafficking of envelope glycoprotein E and the ORF47 and IE62 proteins, components of the tegument, was aberrant without ORF47 kinase activity. In summary, normal VZV virion assembly appears to require ORF47 kinase function. Cell fusion was induced by ORF47 mutants in skin, and cell-cell spread occurred even though virion formation was deficient. VZV-infected T cells do not undergo cell fusion, and impaired virion assembly by ORF47 mutants was associated with a complete elimination of T-cell infectivity. These observations suggest a differential requirement for cell fusion and virion formation in the pathogenesis of VZV infection in skin and T cells.


Assuntos
Herpesvirus Humano 3/patogenicidade , Fusão de Membrana , Pele/virologia , Linfócitos T/virologia , Vírion/fisiologia , Replicação Viral , Animais , Humanos , Masculino , Camundongos , Proteínas Quinases/fisiologia , Transplante de Pele , Transplante Heterólogo , Proteínas do Envelope Viral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA