Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 16(12): e1009166, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33370402

RESUMO

Herpes simplex virus 1 (HSV-1) infects skin and mucosal epithelial cells and then travels along axons to establish latency in the neurones of sensory ganglia. Although viral gene expression is restricted during latency, the latency-associated transcript (LAT) locus encodes many RNAs, including a 2 kb intron known as the hallmark of HSV-1 latency. Here, we studied HSV-1 infection and the role of the LAT locus in human skin xenografts in vivo and in cultured explants. We sequenced the genomes of our stock of HSV-1 strain 17syn+ and seven derived viruses and found nonsynonymous mutations in many viral proteins that had no impact on skin infection. In contrast, deletions in the LAT locus severely impaired HSV-1 replication and lesion formation in skin. However, skin replication was not affected by impaired intron splicing. Moreover, although the LAT locus has been implicated in regulating gene expression in neurones, we observed only small changes in transcript levels that were unrelated to the growth defect in skin, suggesting that its functions in skin may be different from those in neurones. Thus, although the LAT locus was previously thought to be dispensable for lytic infection, we show that it is a determinant of HSV-1 virulence during lytic infection of human skin.


Assuntos
Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , MicroRNAs/genética , Pele/virologia , Virulência/genética , Animais , Xenoenxertos , Humanos , Camundongos , Fatores de Virulência/genética
2.
Viruses ; 11(6)2019 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-31159224

RESUMO

Varicella-zoster virus (VZV), an exclusively human herpesvirus, causes chickenpox and establishes a latent infection in ganglia, reactivating decades later to produce zoster and associated neurological complications. An understanding of VZV neurotropism in humans has long been hampered by the lack of an adequate animal model. For example, experimental inoculation of VZV in small animals including guinea pigs and cotton rats results in the infection of ganglia but not a rash. The severe combined immune deficient human (SCID-hu) model allows the study of VZV neurotropism for human neural sub-populations. Simian varicella virus (SVV) infection of rhesus macaques (RM) closely resembles both human primary VZV infection and reactivation, with analyses at early times after infection providing valuable information about the extent of viral replication and the host immune responses. Indeed, a critical role for CD4 T-cell immunity during acute SVV infection as well as reactivation has emerged based on studies using RM. Herein we discuss the results of efforts from different groups to establish an animal model of VZV neurotropism.


Assuntos
Modelos Animais de Doenças , Gânglios/virologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 3/patogenicidade , Tropismo Viral , Animais , Varicela/virologia , Cobaias , Herpes Zoster/virologia , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/imunologia , Macaca mulatta , Sigmodontinae , Carga Viral , Replicação Viral
3.
Virology ; 523: 110-120, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30119012

RESUMO

Knowledge about the function of varicella-zoster virus glycoprotein M is limited; the requirement of gM for skin and neural tropism are unknown. VZV gM contains two predicted YXXΦ trafficking motifs and a dileucine motif in the carboxyl-terminus. We constructed a recombinant VZV with gM truncated from the first YXXΦ and five additional viruses with YXXΦ tyrosine substitutions, alone and in combination with dileucine substitution. All recombinant viruses grew to high titer but mutation of the membrane-proximal YXXΦ motif reduced plaque size in cultured cells and altered gM localization. C-terminus truncation had a pronounced effect on virion morphogenesis and plaque size, but not on overall replication kinetics in vitro. Mutation of gM trafficking motifs and truncation attenuated replication in human skin xenografts in vivo; gM truncation did not alter neurotropism. Our results demonstrate that the gM C-terminus is dispensable for virus replication in cultured cells but is important for skin pathogenesis.


Assuntos
Gânglios Espinais/virologia , Herpes Zoster/virologia , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/patogenicidade , Pele/virologia , Proteínas da Matriz Viral/química , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Modelos Animais de Doenças , Gânglios Espinais/patologia , Herpes Zoster/patologia , Herpesvirus Humano 3/metabolismo , Xenoenxertos , Humanos , Masculino , Camundongos , Domínios Proteicos , Transporte Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pele/patologia , Carga Viral , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Ensaio de Placa Viral , Tropismo Viral , Virulência , Replicação Viral
4.
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
5.
PLoS Pathog ; 11(6): e1004989, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26090802

RESUMO

Varicella zoster virus (VZV), a human alphaherpesvirus, causes varicella during primary infection. VZV reactivation from neuronal latency may cause herpes zoster, post herpetic neuralgia (PHN) and other neurologic syndromes. To investigate VZV neuropathogenesis, we developed a model using human dorsal root ganglia (DRG) xenografts in immunodeficient (SCID) mice. The SCID DRG model provides an opportunity to examine characteristics of VZV infection that occur in the context of the specialized architecture of DRG, in which nerve cell bodies are ensheathed by satellite glial cells (SGC) which support neuronal homeostasis. We hypothesized that VZV exhibits neuron-subtype specific tropism and that VZV tropism for SGC contributes to VZV-related ganglionopathy. Based on quantitative analyses of viral and cell protein expression in DRG tissue sections, we demonstrated that, whereas DRG neurons had an immature neuronal phenotype prior to implantation, subtype heterogeneity was observed within 20 weeks and SGC retained the capacity to maintain neuronal homeostasis longterm. Profiling VZV protein expression in DRG neurons showed that VZV enters peripherin+ nociceptive and RT97+ mechanoreceptive neurons by both axonal transport and contiguous spread from SGC, but replication in RT97+ neurons is blocked. Restriction occurs even when the SGC surrounding the neuronal cell body were infected and after entry and ORF61 expression, but before IE62 or IE63 protein expression. Notably, although contiguous VZV spread with loss of SGC support would be predicted to affect survival of both nociceptive and mechanoreceptive neurons, RT97+ neurons showed selective loss relative to peripherin+ neurons at later times in DRG infection. Profiling cell factors that were upregulated in VZV-infected DRG indicated that VZV infection induced marked pro-inflammatory responses, as well as proteins of the interferon pathway and neuroprotective responses. These neuropathologic changes observed in sensory ganglia infected with VZV may help to explain the neurologic sequelae often associated with zoster and PHN.


Assuntos
Varicela/virologia , Gânglios Espinais/virologia , Herpes Zoster/virologia , Herpesvirus Humano 3/patogenicidade , Neurônios/virologia , Animais , Varicela/patologia , Herpes Zoster/patologia , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Células Satélites Perineuronais/virologia , Virulência/fisiologia
6.
Proc Natl Acad Sci U S A ; 112(1): 256-61, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25535384

RESUMO

Autophagy is a process by which misfolded and damaged proteins are sequestered into autophagosomes, before degradation in and recycling from lysosomes. We have extensively studied the role of autophagy in varicella-zoster virus (VZV) infection, and have observed that vesicular cells are filled with >100 autophagosomes that are easily detectable after immunolabeling for the LC3 protein. To confirm our hypothesis that increased autophagosome formation was not secondary to a block, we examined all conditions of VZV infection as well as carrying out two assessments of autophagic flux. We first investigated autophagy in human skin xenografts in the severe combined immunodeficiency (SCID) mouse model of VZV pathogenesis, and observed that autophagosomes were abundant in infected human skin tissues. We next investigated autophagy following infection with sonically prepared cell-free virus in cultured cells. Under these conditions, autophagy was detected in a majority of infected cells, but was much less than that seen after an infected-cell inoculum. In other words, inoculation with lower-titered cell-free virus did not reflect the level of stress to the VZV-infected cell that was seen after inoculation of human skin in the SCID mouse model or monolayers with higher-titered infected cells. Finally, we investigated VZV-induced autophagic flux by two different methods (radiolabeling proteins and a dual-colored LC3 plasmid); both showed no evidence of a block in autophagy. Overall, therefore, autophagy within a VZV-infected cell was remarkably different from autophagy within an HSV-infected cell, whose genome contains two modifiers of autophagy, ICP34.5 and US11, not present in VZV.


Assuntos
Autofagia , Herpes Simples/patologia , Herpes Simples/virologia , Herpes Zoster/patologia , Herpes Zoster/virologia , Herpesvirus Humano 3/fisiologia , Simplexvirus/fisiologia , Animais , Linhagem Celular , Sistema Livre de Células , Modelos Animais de Doenças , Fibroblastos/patologia , Fibroblastos/virologia , Proteínas de Fluorescência Verde/metabolismo , Xenoenxertos , Humanos , Camundongos SCID , Proteínas Associadas aos Microtúbulos/metabolismo , Fagossomos/metabolismo , Plasmídeos/metabolismo , Pele/patologia , Pele/virologia
7.
Nat Rev Microbiol ; 12(3): 197-210, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24509782

RESUMO

Varicella zoster virus (VZV) is the causative agent of varicella (chickenpox) and zoster (shingles). Investigating VZV pathogenesis is challenging as VZV is a human-specific virus and infection does not occur, or is highly restricted, in other species. However, the use of human tissue xenografts in mice with severe combined immunodeficiency (SCID) enables the analysis of VZV infection in differentiated human cells in their typical tissue microenvironment. Xenografts of human skin, dorsal root ganglia or foetal thymus that contains T cells can be infected with mutant viruses or in the presence of inhibitors of viral or cellular functions to assess the molecular mechanisms of VZV-host interactions. In this Review, we discuss how these models have improved our understanding of VZV pathogenesis.


Assuntos
Varicela/virologia , Herpes Zoster/virologia , Herpesvirus Humano 3/patogenicidade , Interações Hospedeiro-Patógeno , Animais , Varicela/patologia , Modelos Animais de Doenças , Herpes Zoster/patologia , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiologia , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Mutação , Tropismo Viral
8.
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
9.
J Virol ; 87(5): 2791-802, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23269807

RESUMO

The tropism of herpes simplex virus (HSV-1) for human sensory neurons infected in vivo was examined using dorsal root ganglion (DRG) xenografts maintained in mice with severe combined immunodeficiency (SCID). In contrast to the HSV-1 lytic infectious cycle in vitro, replication of the HSV-1 F strain was restricted in human DRG neurons despite the absence of adaptive immune responses in SCID mice, allowing the establishment of neuronal latency. At 12 days after DRG inoculation, 26.2% of human neurons expressed HSV-1 protein and 13.1% expressed latency-associated transcripts (LAT). Some infected neurons showed cytopathic changes, but HSV-1, unlike varicella-zoster virus (VZV), only rarely infected satellite cells and did not induce fusion of neuronal and satellite cell plasma membranes. Cell-free enveloped HSV-1 virions were observed, indicating productive infection. A recombinant HSV-1-expressing luciferase exhibited less virulence than HSV-1 F in the SCID mouse host, enabling analysis of infection in human DRG xenografts for a 61-day interval. At 12 days after inoculation, 4.2% of neurons expressed HSV-1 proteins; frequencies increased to 32.1% at 33 days but declined to 20.8% by 61 days. Frequencies of LAT-positive neurons were 1.2% at 12 days and increased to 40.2% at 33 days. LAT expression remained at 37% at 61 days, in contrast to the decline in neurons expressing viral proteins. These observations show that the progression of HSV-1 infection is highly restricted in human DRG, and HSV-1 genome silencing occurs in human neurons infected in vivo as a consequence of virus-host cell interactions and does not require adaptive immune control.


Assuntos
Gânglios Espinais/virologia , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Imunodeficiência Combinada Severa/virologia , Tropismo Viral , Aciclovir/administração & dosagem , Aciclovir/análogos & derivados , Aciclovir/farmacologia , Animais , Gânglios Espinais/patologia , Expressão Gênica , Herpes Simples/tratamento farmacológico , Herpes Simples/metabolismo , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 3 , Humanos , Luciferases/biossíntese , Camundongos , Camundongos SCID , Células Satélites Perineuronais/virologia , Transplante Heterólogo , Valaciclovir , Valina/administração & dosagem , Valina/análogos & derivados , Valina/farmacologia , Proteínas Virais/metabolismo , Latência Viral , Replicação Viral
10.
J Virol ; 86(1): 578-83, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22013055

RESUMO

Analyses of varicella-zoster virus (VZV) protein expression during latency have been discordant, with rare to many positive neurons detected. We show that ascites-derived murine and rabbit antibodies specific for VZV proteins in vitro contain endogenous antibodies that react with human blood type A antigens in neurons. Apparent VZV neuronal staining and blood type A were strongly associated (by a χ² test, α = 0.0003). Adsorption of ascites-derived monoclonal antibodies or antiserum with type A erythrocytes or the use of in vitro-derived VZV monoclonal antibodies eliminated apparent VZV staining. Animal-derived antibodies must be screened for anti-blood type A reactivity to avoid misidentification of viral proteins in the neurons of the 30 to 40% of individuals who are blood type A.


Assuntos
Sistema ABO de Grupos Sanguíneos/imunologia , Anticorpos Antivirais/imunologia , Anticorpos/imunologia , Herpes Zoster/virologia , Herpesvirus Humano 3/genética , Células Receptoras Sensoriais/virologia , Proteínas Virais/genética , Latência Viral , Animais , Anticorpos/análise , Anticorpos Antivirais/análise , Reações Cruzadas , Regulação Viral da Expressão Gênica , Herpes Zoster/imunologia , Herpesvirus Humano 3/química , Herpesvirus Humano 3/imunologia , Herpesvirus Humano 3/fisiologia , Humanos , Imuno-Histoquímica , Camundongos , Coelhos , Células Receptoras Sensoriais/química , Células Receptoras Sensoriais/imunologia , Proteínas Virais/análise , Proteínas Virais/imunologia
11.
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
12.
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
13.
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
14.
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
15.
Curr Top Microbiol Immunol ; 342: 189-209, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20397071

RESUMO

Varicella-zoster virus (VZV) is a medically important human alphaherpesvirus that causes varicella and zoster. VZV initiates primary infection by inoculation of the respiratory mucosa. In the course of primary infection, VZV establishes a life-long persistence in sensory ganglia; VZV reactivation from latency may result in zoster in healthy and immunocompromised patients. The VZV genome has at least 70 known or predicted open reading frames (ORFs), but understanding how these gene products function in virulence is difficult because VZV is a highly human-specific pathogen. We have addressed this obstacle by investigating VZV infection of human tissue xenografts in the severe combined immunodeficiency mouse model. In studies relevant to the pathogenesis of primary VZV infection, we have examined VZV infection of human T cell (thymus/liver) and skin xenografts. This work supports a new paradigm for VZV pathogenesis in which VZV T cell tropism provides a mechanism for delivering the virus to skin. We have also shown that VZV-infected T cells transfer VZV to neurons in sensory ganglia. The construction of infectious VZV recombinants that have deletions or targeted mutations of viral genes or their promoters and the evaluation of VZV mutants in T cell and skin xenografts has revealed determinants of VZV virulence that are important for T cell and skin tropism in vivo.


Assuntos
Varicela/imunologia , Varicela/virologia , Herpes Zoster/virologia , Herpesvirus Humano 3/imunologia , Tecido Linfoide/imunologia , Dermatopatias Infecciosas/virologia , Linfócitos T/imunologia , Animais , Herpes Zoster/imunologia , Humanos , Tecido Linfoide/virologia , Camundongos , Camundongos SCID , Pele/imunologia , Pele/virologia , Dermatopatias Infecciosas/imunologia , Linfócitos T/virologia
16.
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
17.
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
18.
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
19.
J Virol ; 84(1): 141-52, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19828615

RESUMO

Varicella-zoster virus (VZV) infection is usually mild in healthy individuals but can cause severe disease in immunocompromised patients. Prophylaxis with varicella-zoster immunoglobulin can reduce the severity of VZV if given shortly after exposure. Glycoprotein H (gH) is a highly conserved herpesvirus protein with functions in virus entry and cell-cell spread and is a target of neutralizing antibodies. The anti-gH monoclonal antibody (MAb) 206 neutralizes VZV in vitro. To determine the requirement for gH in VZV pathogenesis in vivo, MAb 206 was administered to SCID mice with human skin xenografts inoculated with VZV. Anti-gH antibody given at 6 h postinfection significantly reduced the frequency of skin xenograft infection by 42%. Virus titers, genome copies, and lesion size were decreased in xenografts that became infected. In contrast, administering anti-gH antibody at 4 days postinfection suppressed VZV replication but did not reduce the frequency of infection. The neutralizing anti-gH MAb 206 blocked virus entry, cell fusion, or both in skin in vivo. In vitro, MAb 206 bound to plasma membranes and to surface virus particles. Antibody was internalized into vacuoles within infected cells, associated with intracellular virus particles, and colocalized with markers for early endosomes and multivesicular bodies but not the trans-Golgi network. MAb 206 blocked spread, altered intracellular trafficking of gH, and bound to surface VZV particles, which might facilitate their uptake and targeting for degradation. As a consequence, antibody interference with gH function would likely prevent or significantly reduce VZV replication in skin during primary or recurrent infection.


Assuntos
Anticorpos Monoclonais/farmacocinética , Anticorpos Antivirais/uso terapêutico , Herpesvirus Humano 3/patogenicidade , Glicoproteínas de Membrana/imunologia , Pele/virologia , Proteínas Virais/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Transporte Biológico , Herpesvirus Humano 3/efeitos dos fármacos , Herpesvirus Humano 3/imunologia , Humanos , Imunoglobulinas/uso terapêutico , Camundongos , Camundongos SCID , Pele/imunologia , Transplante de Pele , Transplante Heterólogo
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...