Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Virology ; 485: 47-57, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26207799

RESUMO

The immediate early (IE) 62 protein is the major varicella-zoster virus (VZV) regulatory factor. Analysis of the VZV genome revealed 40 predicted GC-rich boxes within 36 promoters. We examined effects of ectopic expression of Sp1-Sp4 on IE62- mediated transactivation of three viral promoters. Ectopic expression of Sp3 and Sp4 enhanced IE62 activation of ORF3 and gI promoters while Sp3 reduced IE62 activation of ORF28/29 promoter and VZV DNA replication. Sp2 reduced IE62 transactivation of gI while Sp1 had no significant influence on IE62 activation with any of these viral promoters. Electrophoretic mobility shift assays (EMSA) confirmed binding of Sp1 and Sp3 but not Sp2 and Sp4 to the gI promoter. Sp1-4 bound to IE62 and amino acids 238-258 of IE62 were important for the interaction with Sp3 and Sp4 as well as Sp1. This work shows that Sp family members have differential effects on IE62-mediated transactivation in a promoter-dependent manner.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 3/genética , Proteínas Imediatamente Precoces/genética , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp2/genética , Fator de Transcrição Sp3/genética , Fator de Transcrição Sp4/genética , Transativadores/genética , Proteínas do Envelope Viral/genética , Composição de Bases , Sequência de Bases , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/virologia , Genoma Viral , Herpesvirus Humano 3/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Proteínas Imediatamente Precoces/metabolismo , Dados de Sequência Molecular , Fases de Leitura Aberta , Regiões Promotoras Genéticas , Ligação Proteica , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp2/metabolismo , Fator de Transcrição Sp3/metabolismo , Fator de Transcrição Sp4/metabolismo , Transativadores/metabolismo , Ativação Transcricional , Proteínas do Envelope Viral/metabolismo
2.
Virology ; 481: 179-86, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25795313

RESUMO

The VZV genome has two origins of DNA replication (oriS), each of which consists of an AT-rich sequence and three origin binding protein (OBP) sites called Box A, C and B. In these experiments, the mutation in the core sequence CGC of the Box A and C not only inhibited DNA replication but also inhibited both ORF62 and ORF63 expression in reporter gene assays. In contrast the Box B mutation did not influence DNA replication or flanking gene transcription. These results suggest that efficient DNA replication enhances ORF62 and ORF63 transcription. Recombinant viruses carrying these mutations in both sites and one with a deletion of the whole oriS were constructed. Surprisingly, the recombinant virus lacking both copies of oriS retained the capacity to replicate in melanoma and HELF cells suggesting that VZV has another origin of DNA replication.


Assuntos
Varicela/virologia , Replicação do DNA , DNA Viral/genética , Herpesvirus Humano 3/genética , Origem de Replicação , Sequência de Bases , Linhagem Celular , Genoma Viral , Herpesvirus Humano 3/fisiologia , Humanos , Dados de Sequência Molecular , Transcrição Gênica , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
3.
Virology ; 449: 244-53, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24418559

RESUMO

Several cellular transcription factors have been shown to be involved in IE62-mediated activation. The YY1 cellular transcription factor has activating and repressive effects on gene transcription. Analysis of the VZV genome revealed 19 postulated YY1 binding sites located within putative promoters of 16 VZV genes. Electrophoretic mobility shift assays (EMSA) confirmed the binding of YY1 to ORF10, ORF28/29 and gI promoters and the mutation of these binding sites inhibited YY1 binding and the promoter activation by IE62 alone or following VZV infection. Mutation of the ORF28/29 YY1 site in the VZV genome displayed insignificant influence on virus growth in melanoma cells; but it inhibited the virus replication significantly at day 5 and 6 post infection in HELF cells. This work suggests a novel role for the cellular factor YY1 in VZV replication through the mediation of IE62 activation of viral gene expression.


Assuntos
Regulação Viral da Expressão Gênica , Herpes Zoster/metabolismo , Herpesvirus Humano 3/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Transativadores/metabolismo , Ativação Transcricional , Proteínas do Envelope Viral/metabolismo , Fator de Transcrição YY1/metabolismo , Linhagem Celular Tumoral , Herpes Zoster/genética , Herpes Zoster/virologia , Herpesvirus Humano 3/genética , Humanos , Proteínas Imediatamente Precoces/genética , Regiões Promotoras Genéticas , Ligação Proteica , Transativadores/genética , Proteínas do Envelope Viral/genética , Fator de Transcrição YY1/genética
4.
Virology ; 440(2): 171-81, 2013 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23523134

RESUMO

The varicella zoster virus (VZV) immediate early 62 protein (IE62) activates most if not all identified promoters of VZV genes and also some minimum model promoters that contain only a TATA box element. Analysis of the DNA elements that function in IE62 activation of the VZV ORF3 promoter revealed that the 100 nucleotides before the translation start site of the ORF3 gene contains the promoter elements. This promoter lacks any functional TATA box element. Cellular transcription factors Sp1, Sp3 and YY1 bind to the promoter, and mutation of their binding sites inhibited ORF3 gene expression. VZV regulatory proteins, IE63 and ORF29, ORF61 and ORF10 proteins inhibited IE62-mediated activation of this promoter. Mutation of the Sp1/Sp3 binding site in the VZV genome did not alter VZV replication kinetics. This work suggests that Sp family proteins contribute to the activation of VZV promoters by IE62 in the absence of functional TATA box.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 3/patogenicidade , Proteínas Imediatamente Precoces/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp3/metabolismo , Transativadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Fator de Transcrição YY1/metabolismo , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos
5.
J Virol ; 86(23): 13070-80, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22933283

RESUMO

The distribution and orientation of origin-binding protein (OBP) sites are the main architectural contrasts between varicella-zoster virus (VZV) and herpes simplex virus (HSV) origins of DNA replication (oriS). One important difference is the absence of a downstream OBP site in VZV, raising the possibility that an alternative cis element may replace its function. Our previous work established that Sp1, Sp3, and YY1 bind to specific sites within the downstream region of VZV oriS; we hypothesize that one or both of these sites may be the alternative cis element(s). Here, we show that the mutation of the Sp1/Sp3 site decreases DNA replication and transcription from the adjacent ORF62 and ORF63 promoters following superinfection with VZV. In contrast, in the absence of DNA replication or in transfection experiments with ORF62, only ORF63 transcription is affected. YY1 site mutations had no significant effect on either process. Recombinant viruses containing these mutations were then constructed. The Sp1/Sp3 site mutant exhibited a significant decrease in virus growth in MeWo cells and in human skin xenografts, while the YY1 site mutant virus grew as well as the wild type in MeWo cells, even showing a late increase in VZV replication in skin xenografts following infection. These results suggest that the Sp1/Sp3 site plays an important role in both VZV origin-dependent DNA replication and ORF62 and ORF63 transcription and that, in contrast to HSV, these events are linked during virus replication.


Assuntos
Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Herpesvirus Humano 3/fisiologia , Pele/virologia , Transcrição Gênica/genética , Proteínas Virais/genética , Replicação Viral/genética , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , Primers do DNA/genética , Herpesvirus Humano 3/genética , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/fisiologia , Immunoblotting , Técnicas In Vitro , Camundongos , Camundongos SCID , Plasmídeos/genética , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp3/genética , Transativadores/genética , Transativadores/fisiologia , Transcrição Gênica/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Fator de Transcrição YY1/genética
6.
J Virol ; 85(23): 12188-200, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21937644

RESUMO

The architecture of the varicella-zoster virus (VZV) origin of DNA replication (OriS) differs significantly from that of the herpes simplex virus (HSV) DNA replication origin. Novel aspects of the VZV OriS include a GA-rich region, three binding sites for the VZV origin-binding protein (OBP) all on the same strand and oriented in the same direction, and a partial OBP binding site of unknown function. We have designated this partial binding site Box D and have investigated the role it plays in DNA replication and flanking gene expression. This has been done with a model system using a replication-competent plasmid containing OriS and a replication- and transcription-competent dual-luciferase reporter plasmid containing both the OriS and the intergenic region between VZV open reading frames (ORFs) 62 and 63. We have found that (i) Box D is a negative regulator of DNA replication independent of flanking gene expression, (ii) the mutation of Box D results in a decrease in flanking gene expression, thus a sequence within the VZV OriS affects transcription, which is in contrast to results reported for HSV-1, (iii) there is a specific Box D complex formed with infected cell extracts in electrophoretic mobility shift assay experiments, (iv) supershift assays show that this complex contains the VZV ORF29 single-strand DNA-binding protein, and (v) the formation of this complex is dependent on the presence of CGC motifs in Box D and its downstream flanking region. These findings show that the VZV ORF29 protein, while required for DNA replication, also plays a novel role in the suppression of that process.


Assuntos
Replicação do DNA , Herpesvirus Humano 3/genética , Melanoma/genética , Fases de Leitura Aberta/genética , Origem de Replicação/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/virologia , DNA Viral/genética , Ensaio de Desvio de Mobilidade Eletroforética , Genes Reporter , Herpesvirus Humano 3/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética , Melanoma/patologia , Melanoma/virologia , Plasmídeos , Transativadores/genética , Transcrição Gênica , Células Tumorais Cultivadas , Proteínas do Envelope Viral/genética , Latência Viral , Replicação Viral
7.
J Struct Funct Genomics ; 12(3): 159-66, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21785987

RESUMO

The solution NMR structure of protein MED25(391-543), comprising the activator interacting domain (ACID) of subunit 25 of the human mediator, is presented along with the measurement of polypeptide backbone heteronuclear 15N-{1H} NOEs to identify fast internal motional modes. This domain interacts with the acidic transactivation domains of Herpes simplex type 1 (HSV-1) protein VP16 and the Varicella-zoster virus (VZV) major transactivator protein IE62, which initiate transcription of viral genes. The structure is similar to the ß-barrel domains of the human protein Ku and the SPOC domain of human protein SHARP, and provides a starting point to understand the structural biology of initiation of HSV-1 and VZV gene activation. Homology models built for the two ACID domains of the prostate tumor overexpressed (PTOV1) protein using the structure of MED25(391-543) as a template suggest that differential biological activities of the ACID domains in MED25 and PTOV1 arise from modulation of quite similar protein-protein interactions by variable residues grouped around highly conserved charged surface areas.


Assuntos
Complexo Mediador/química , Sequência de Aminoácidos , Biomarcadores Tumorais/química , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Complexo Mediador/genética , Complexo Mediador/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias/química , Ressonância Magnética Nuclear Biomolecular , Regiões Promotoras Genéticas , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Transativadores/metabolismo , Proteínas do Envelope Viral/metabolismo
8.
Virus Res ; 155(1): 334-42, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21074584

RESUMO

The varicella-zoster virus (VZV) open reading frame (ORF) 66 encodes a serine/threonine kinase that phosphorylates the major viral transactivator protein, immediate-early (IE) 62, preventing its nuclear importation. Cytoplasmic sequestration of IE62 may alter viral gene transcription and could serve as a mechanism for maintaining VZV latency. We examined the regulation of expression of the ORF66 gene by mapping the promoter region, which was localized to within 150 bases of the start codon. The ORF66 promoter was activated by two viral regulatory proteins, IE62 and IE63. We evaluated the binding of viral regulatory proteins and cellular transcription factors based on recognized cellular transcription factor binding sites identified within the ORF66 promoter. These included Sp1 and TBP binding sites, several of which were essential for optimal promoter activity. Site-directed mutations in Sp1 and TBP binding sites led to varying degrees of impairment of ORF66 gene expression in the context of VZV infection. We also examined the effect of Sp1 and TBP mutations on IE62, Sp1, and TBP binding. These studies reveal that host cell-derived and viral factors contribute to and cooperate in the expression of this important viral kinase gene.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 3/enzimologia , Herpesvirus Humano 3/fisiologia , Fases de Leitura Aberta , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Virais/biossíntese , Sítios de Ligação , Humanos , Proteínas Imediatamente Precoces/metabolismo , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Ligação Proteica , Transativadores/metabolismo , Sítio de Iniciação de Transcrição , Proteínas do Envelope Viral/metabolismo
9.
J Virol ; 84(18): 9240-53, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20631144

RESUMO

Varicella-zoster virus (VZV) is an alphaherpesvirus that is restricted to humans. VZV infection of differentiated cells within the host and establishment of latency likely require evasion of innate immunity and limited secretion of antiviral cytokines. Since interferons (IFNs) severely limit VZV replication, we examined the ability of VZV to modulate the induction of the type I IFN response in primary human embryonic lung fibroblasts (HELF). IFN-beta production was not detected, and transcription of two interferon response factor 3 (IRF3)-dependent interferon-stimulated genes (ISGs), ISG54 and ISG56, in response to poly(I:C) stimulation was downregulated in VZV-infected HELF. Inhibition of IRF3 function did not require VZV replication; the viral immediate-early protein 62 (IE62) alone was sufficient to produce this effect. IE62 blocked TBK1-mediated IFN-beta secretion and IRF3 function, as shown in an IFN-stimulated response element (ISRE)-luciferase reporter assay. However, IRF3 function was preserved if constitutively active IRF3 (IRF3-5D) was expressed in VZV-infected or IE62-transfected cells, indicating that VZV interferes with IRF3 phosphorylation. IE62-mediated inhibition was mapped to blocking phosphorylation of at least three serine residues on IRF3. However, IE62 binding to TBK1 or IRF3 was not detected and IE62 did not perturb TBK1-IRF3 complex formation. IE62-mediated inhibition of IRF3 function was maintained even if IE62 transactivator activity was disrupted. Thus, IE62 has two critical but discrete roles following VZV entry: to induce expression of VZV genes and to disarm the IFN-dependent antiviral defense through a novel mechanism that prevents IRF3 phosphorylation.


Assuntos
Herpesvirus Humano 3/patogenicidade , Interações Hospedeiro-Patógeno , Proteínas Imediatamente Precoces/fisiologia , Evasão da Resposta Imune , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/imunologia , Transativadores/fisiologia , Proteínas do Envelope Viral/fisiologia , Fatores de Virulência/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Células Cultivadas , Regulação para Baixo , Fibroblastos/virologia , Perfilação da Expressão Gênica , Genes Reporter , Herpesvirus Humano 3/imunologia , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/antagonistas & inibidores , Luciferases/genética , Luciferases/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas de Ligação a RNA , Serina/metabolismo , Fatores de Transcrição/biossíntese
10.
Curr Top Microbiol Immunol ; 342: 43-65, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20490778

RESUMO

Varicella zoster virus (VZV) is the causative agent of chickenpox and shingles. During productive infection the complete VZV proteome consisting of some 68 unique gene products is expressed through interaction of a small number of viral transcriptional activators with the general transcription apparatus of the host cell. Recent work has shown that the major viral transactivator, commonly designated the IE62 protein, interacts with the human Mediator of transcription. This interaction requires direct contact between the MED25 subunit of Mediator and the acidic N-terminal transactivation domain of IE62. A second cellular factor, host cell factor-1, has been shown to be the common element in two mechanisms of activation of the promoter driving expression of the gene encoding IE62. Finally, the ubiquitous cellular transcription factors Sp1, Sp3, and YY1 have been shown to interact with sequences near the VZV origin of DNA replication and in the case of Sp1/Sp3 to influence replication efficiency.


Assuntos
Replicação do DNA/fisiologia , Herpesvirus Humano 3/fisiologia , Fatores de Transcrição/fisiologia , Replicação Viral/fisiologia , Humanos
11.
J Virol ; 84(8): 3767-79, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20130051

RESUMO

The varicella-zoster virus (VZV) IE62 protein is the major transcriptional activator. IE62 is capable of associating with DNA both nonspecifically and in a sequence-specific manner via a consensus binding site (5'-ATCGT-3'). However, the function of the consensus site is poorly understood, since IE62 efficiently transactivates promoter elements lacking this sequence. In the work presented here, sequence analysis of the VZV genome revealed the presence of 245 IE62 consensus sites throughout the genome. Some 54 sites were found to be present within putative VZV promoters. Electrophoretic mobility shift assay (EMSA) experiments using an IE62 fragment containing the IE62 DNA-binding domain and duplex oligonucleotides that did or did not contain the IE62 consensus binding sequence yielded K(D) (equilibrium dissociation constant) values in the nanomolar range. Further, the IE62 DNA binding domain was shown to have a 5-fold-increased affinity for its consensus site compared to nonconsensus sequences. The effect of consensus site presence and position on IE62-mediated activation of native VZV and model promoters was examined using site-specific mutagenesis and transfection and superinfection reporter assays. In all promoters examined, the consensus sequence functioned as a distance-dependent repressive element. Protein recruitment assays utilizing the VZV gI promoter indicated that the presence of the consensus site increased the recruitment of IE62 but not Sp1. These data suggest a model where the IE62 consensus site functions to down-modulate IE62 activation, and interaction of IE62 with this sequence may result in loss or decrease of the ability of IE62 to recruit cellular factors needed for full promoter activation.


Assuntos
DNA Viral/metabolismo , Herpesvirus Humano 3/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Transativadores/metabolismo , Ativação Transcricional , Proteínas do Envelope Viral/metabolismo , Sequência de Bases , Sítios de Ligação , Sequência Consenso , DNA Viral/genética , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Proteínas Imediatamente Precoces/genética , Cinética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Transativadores/genética , Proteínas do Envelope Viral/genética
12.
J Virol ; 83(12): 6300-5, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19357160

RESUMO

The varicella-zoster virus major transactivator, IE62, contains a potent N-terminal acidic transcriptional activation domain (TAD). Our experiments revealed that the minimal IE62 TAD encompasses amino acids (aa) 19 to 67. We showed that the minimal TAD interacts with the human Mediator complex. Site-specific mutations revealed residues throughout the minimal TAD that are important for both activation and Mediator interaction. The TAD interacts directly with aa 402 to 590 of the MED25 subunit, and site-specific TAD mutations abolished this interaction. Two-dimensional nuclear magnetic resonance spectroscopy revealed that the TAD is intrinsically unstructured. Our studies suggest that transactivation may involve the TAD adopting a defined structure upon binding MED25.


Assuntos
Herpesvirus Humano 3/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Transativadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Complexo Mediador , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Estrutura Secundária de Proteína , Ativação Transcricional
13.
J Virol ; 82(24): 12154-63, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18842726

RESUMO

The varicella-zoster virus (VZV) major transactivator, IE62, is involved in the expression of all kinetic classes of VZV genes and can also activate cellular promoters, promoters from heterologous viruses, and artificial promoters containing only TATA elements. A key component of the mechanism of IE62 transactivation is an acidic activation domain comprising the N-terminal 86 amino acids of IE62. However, the cellular target of this N-terminal acidic activation is unknown. In the work presented here, we show that the IE62 activation domain targets the human Mediator complex via the Med25 (ARC92) subunit and that this interaction appears to be fundamental for transactivation by the IE62 activation domain. In contrast, the Med23 subunit (Sur2/TRAP150beta/DRIP130/CRSP130) of the Mediator complex is not essential for IE62-mediated activation. Further, the IE62 activation domain appears to selectively interact with a form of the Mediator complex lacking CDK8. Chromatin immunoprecipitation experiments showed that IE62 stimulates recruitment of Mediator to an IE62-responsive model promoter. Finally, immunofluorescence microscopy of VZV-infected cells demonstrated intranuclear translocation of the Mediator complex to viral replication compartments. These studies suggest that Mediator is an essential component for efficient VZV gene expression.


Assuntos
Herpesvirus Humano 3/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Regiões Promotoras Genéticas/genética , Transativadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Linhagem Celular Tumoral , Quinase 8 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Herpesvirus Humano 3/genética , Humanos , Proteínas Imediatamente Precoces/genética , Complexo Mediador , Modelos Genéticos , Ligação Proteica , Transporte Proteico , Transativadores/genética , Ativação Transcricional/genética , Proteínas do Envelope Viral/genética
14.
J Virol ; 82(23): 11723-33, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18815296

RESUMO

The varicella-zoster virus (VZV) origin of DNA replication (oriS) contains a 46-bp AT-rich palindrome and three consensus binding sites for the VZV origin binding protein (OBP) encoded by VZV ORF51. All three OBP binding sites are upstream of the palindrome in contrast to the sequence of the herpes simplex virus oriS, which has required OBP binding sites upstream and downstream of the AT-rich region. We are investigating the roles that sequences downstream of the palindrome play in VZV oriS-dependent DNA replication. Computer analysis identified two GC boxes, GC box 1 and GC box 2, in the downstream region which were predicted to be binding sites for the cellular transcription factor Sp1. Electrophoretic mobility shift assay and supershift assays showed that two members of the Sp family (Sp1 and Sp3) stably bind to GC box 1, but not to GC box 2. A predicted binding site for the cellular factor Yin Yang 1 (YY1) that overlaps with GC box 2 was also identified. Supershift and mutational analyses confirmed the binding of YY1 to this site. Mutation of GC box 1 resulted in loss of Sp1 and Sp3 binding and an increase in origin-dependent replication efficiency in DpnI replication assays. In contrast, mutation of the YY1 site had a statistically insignificant effect. These results suggest a model where origin-dependent DNA replication and viral transcription are coupled by the binding of Sp1 and Sp3 to the downstream region of the VZV replication origin during lytic infection. They may also have implications regarding establishment or reactivation of viral latency.


Assuntos
Replicação do DNA , Herpesvirus Humano 3/genética , Origem de Replicação , Fator de Transcrição Sp1/fisiologia , Fator de Transcrição Sp3/fisiologia , Replicação Viral , Sítios de Ligação , Linhagem Celular Tumoral , Ensaio de Desvio de Mobilidade Eletroforética , Humanos
15.
Proc Natl Acad Sci U S A ; 104(26): 10835-40, 2007 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-17578910

RESUMO

Originally identified as an essential component of the herpes simplex virus immediate early (IE) gene enhancer complex, the transcriptional coactivator host cell factor-1 (HCF-1) has been implicated in a broad range of cellular regulatory circuits. The protein mediates activation through multiple interactions with transcriptional activators, coactivators, and chromatin remodeling complexes. However, the mechanisms involved in HCF-1-dependent transcriptional stimulation were undefined. By using a minimal HCF-1-dependent promoter and a model activator, the varicella zoster IE62 protein, it was determined that HCF-1 was not required for the assembly of the RNAPII basal complex, which depended solely on IE62 in conjunction with the cellular factor Sp1. In contrast, HCF-1 was required for recruitment of the histone methyltransferases Set1 and MLL1 (mixed-lineage leukemia 1), leading to histone H3K4 trimethylation and transcriptional activation. Similarly, in a varicella zoster virus lytic infection, HCF-1, Set1, and MLL1 were recruited to the viral genomic IE promoter, suggesting an essential role for HCF-1 in chromatin modification and remodeling during initiation of lytic infection. The results indicate that one biological rationale for the incorporation of the viral IE activators in the viral particle is to recruit HCF-1/histone methyltransferase complexes and promote assembly of the viral IE gene promoters into transcriptionally active chromatin. These studies also contribute to the model whereby the induced nuclear transport of HCF-1 in sensory neurons may be critical to the reactivation of latent herpesviruses by promoting the activation of chromatin modifications.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Herpes Simples/etiologia , Fator C1 de Célula Hospedeira/fisiologia , Proteína de Leucina Linfoide-Mieloide/metabolismo , Regiões Promotoras Genéticas , Simplexvirus/genética , Simplexvirus/patogenicidade , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular , Proteínas de Ligação a DNA , Regulação Viral da Expressão Gênica , Genes Precoces , Células HeLa , Chaperonas de Histonas , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Fator C1 de Célula Hospedeira/metabolismo , Humanos , Proteínas Imediatamente Precoces , Metilação , Transativadores , Ativação Transcricional , Proteínas do Envelope Viral
16.
J Neurovirol ; 13(1): 56-66, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17454449

RESUMO

Herpes simplex virus type 1 (HSV-1) primarily infects mucoepithelial tissues of the eye, the orofacial region, and to a lesser extent the genitalia. The virus is retrogradely transported through the axons of sensory and sympathetic neurons to their cell bodies to establishe a life-long latent infection. Throughout this latency period, the viral genome is transcriptionally silent except for a single region encoding the latency-associated transcript (LAT). The function of LAT is still largely unknown. To understand how HSV-1 latency might affect neurons, the authors transfected primary cultures of sympathetic neurons and trigeminal sensory neurons obtained from rat embryos with LAT-expressing plasmids. LAT increased the survival of both sympathetic and trigeminal neurons after induction of cell death by nerve growth factor (NGF) deprivation. Because HSV-1 is transported through axons both after initial infection and during reactivation, the authors considered the possibility that LAT may affect axonal growth. They found that LAT expression increased axonal regeneration by twofold in both types of neurons. Inhibition of the mitogen-activated protein kinase (MAPK) pathway reverses stimulation of both neuronal survival and axonal regeneration, which indicates that these effects are mediated through the MAPK pathway. These data provide evidence that HSV-1 LAT promotes survival of sympathetic as well as trigeminal neurons. The authors show for the first time that LAT stimulates axonal regeneration in both sympathetic and trigeminal neurons.


Assuntos
Axônios/fisiologia , Herpesvirus Humano 1/fisiologia , Neurônios/virologia , Gânglio Trigeminal/citologia , Proteínas Virais/fisiologia , Sobrevivência Celular , Células Cultivadas , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/fisiologia , Herpesvirus Humano 1/genética , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Virais/genética , Latência Viral/fisiologia
17.
Neurosci Lett ; 413(1): 31-5, 2007 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-17239535

RESUMO

Herpes simplex virus type-1 (HSV-1) primarily infects mucoepithelial tissues of the eye and the orofacial region. Subsequently, the virus is retrogradely transported through the axons of the trigeminal sensory neurons to their nuclei, where the virus establishes a life-long latent infection. During this latency period, the viral genome is transcriptionally silent except for a single region encoding the latency-associated transcript (LAT). To understand how HSV-1 latency might affect the expression of substance P in sensory neurons, we transfected primary cultures of trigeminal neurons obtained from rat embryos, with LAT expressing plasmids. The expression of LAT increased the percentage of substance P-immunoreactive neurons by two thirds. To examine the effect of bone morphogenetic protein-7 (BMP7) on the LAT-induced increase in substance P expression in trigeminal neurons, cultures transfected with LAT were treated with BMP7. Treatment with BMP7 reversed the effects of LAT on substance P expression in trigeminal neurons. Our data show for the first time that LAT increases substance P expression in trigeminal neurons and BMP7 can reverse these effects of LAT.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Neurônios/metabolismo , Substância P/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Gânglio Trigeminal/citologia , Animais , Proteína Morfogenética Óssea 7 , Células Cultivadas , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica/métodos , Neurônios/fisiologia , Ratos , Transcrição Gênica/fisiologia , Latência Viral
18.
J Virol ; 81(7): 3229-39, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17251302

RESUMO

Varicella-zoster virus (VZV) open reading frame 10 (ORF10) is a determinant of virulence in SCIDhu skin xenografts but not in human T cells in vivo. In this analysis of the regulation of ORF10 transcription, we have identified four ORF10-related transcripts, including a major 1.3-kb RNA spanning ORF10 only and three other read-through transcripts. Rapid-amplification-of-cDNA-ends experiments indicated that the 1.3-kb transcript of ORF10 has single initiation and termination sites. In transient expression assays, the ORF10 promoter was strongly stimulated by the major VZV transactivator, IE62. Deletion analyses revealed approximate boundaries for the full ORF10 promoter activity between -75 and -45 and between +5 and -8, relative to the ORF10 transcription start site. The recombinant virus POKA10-Deltapro, with the ORF10 promoter deletion, blocked transcription of ORF10 and also of ORF9A and ORF9 mRNAs, whereas expression of read-through ORF9A/9/10 and ORF9/10 transcripts was increased, compensating for the loss of the monocistronic mRNAs. The cellular factor USF bound specifically to its consensus site within the ORF10 promoter and was required for IE62 transactivation, whereas disrupting the predicted TATA boxes or Oct-1 binding elements had no effect. The USF binding site was disrupted in the recombinant virus, POKA10-proDeltaUSF, and no ORF10 protein was produced. Both ORF10 promoter mutants reduced VZV replication in SCIDhu skin xenografts. These observations provided further evidence of the contribution of the ORF10 protein to VZV pathogenesis in skin and demonstrated that VZV depends upon the cellular transcriptional factor USF to support its virulence in human skin in vivo.


Assuntos
Herpesvirus Humano 3/metabolismo , Herpesvirus Humano 3/patogenicidade , Fases de Leitura Aberta/genética , Regiões Promotoras Genéticas/genética , Transplante de Pele/patologia , Transplante Heterólogo/patologia , Fatores Estimuladores Upstream/metabolismo , Animais , Linhagem Celular , Regulação Viral da Expressão Gênica , Genes Reporter/genética , Herpesvirus Humano 3/genética , Humanos , Camundongos , Camundongos SCID , Mutação/genética , RNA/genética , Elementos de Resposta , Transativadores/genética , Transcrição Gênica/genética , Fatores Estimuladores Upstream/genética , Virulência , Replicação Viral
19.
J Virol ; 81(2): 761-74, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17079304

RESUMO

The varicella-zoster virus (VZV) ORF9 protein is a member of the herpesvirus UL49 gene family but shares limited identity and similarity with the UL49 prototype, herpes simplex virus type 1 VP22. ORF9 mRNA is the most abundantly expressed message during VZV infection; however, little is known concerning the functions of the ORF9 protein. We have found that the VZV major transactivator IE62 and the ORF9 protein can be coprecipitated from infected cells. Yeast two-hybrid analysis localized the region of the ORF9 protein required for interaction with IE62 to the middle third of the protein encompassing amino acids 117 to 186. Protein pull-down assays with GST-IE62 fusion proteins containing N-terminal IE62 sequences showed that amino acids 1 to 43 of the acidic transcriptional activation domain of IE62 can bind recombinant ORF9 protein. Confocal microscopy of transiently transfected cells showed that in the absence of other viral proteins, the ORF9 protein was localized in the cytoplasm while IE62 was localized in the nucleus. In VZV-infected cells, the ORF9 protein was localized to the cytoplasm whereas IE62 exhibited both nuclear and cytoplasmic localization. Cotransfection of plasmids expressing ORF9, IE62, and the viral ORF66 kinase resulted in significant colocalization of ORF9 and IE62 in the cytoplasm. Coimmunoprecipitation experiments with antitubulin antibodies indicate the presence of ORF9-IE62-tubulin complexes in infected cells. Colocalization of ORF9 and tubulin in transfected cells was visualized by confocal microscopy. These data suggest a model for ORF9 protein function involving complex formation with IE62 and possibly other tegument proteins in the cytoplasm at late times in infection.


Assuntos
Herpesvirus Humano 3/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Fases de Leitura Aberta/fisiologia , Transativadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/química , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Citoplasma/metabolismo , Herpesvirus Humano 3/genética , Humanos , Microscopia Confocal , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Alinhamento de Sequência , Tubulina (Proteína)/metabolismo , Células Vero , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo
20.
Neurobiol Dis ; 25(3): 553-60, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17184994

RESUMO

Herpes simplex virus type-1 (HSV-1) initially infects mucoepithelial tissues of the eye and the orofacial region. Subsequently, the virus is retrogradely transported through the axons of the trigeminal sensory neurons. HSV-1 establishes a life-long latent infection in these neurons, during which the transcription of the viral genome is silent, except for the sequences encoding the latency-associated transcript (LAT). To determine if HSV-1 latency might affect calcitonin gene-related peptide (CGRP) expression in trigeminal sensory neurons, we transfected primary neuronal cultures of trigeminal ganglia from rat embryos with plasmids expressing LAT. In the presence of Bone Morphogenetic Protein-7 (BMP7), CGRP was expressed in 49% of sensory neurons. However, this percentage was reduced to 19% in neurons transfected with LAT expressing plasmids. We also found that transfection of the IE63 gene of varicella-zoster virus (VZV) reduced the percentage of trigeminal neurons containing CGRP. However, the observed effect of IE63 in contrast to that of LAT was completely reversed by treatment of cultures with MgCl2, which indicates that the effect of IE63 was due to increased release of CGRP from trigeminal neurons. We provide here the first evidence that HSV-1 LAT decreases the level of CGRP in trigeminal neurons. These effects may be important for reducing the neuroinflammatory response, thus protecting host neuronal cells during HSV-1 latency in trigeminal neurons. In contrast, increased release of CGRP in the presence of IE63 protein may contribute to the neuralgias associated with VZV infection.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Proteínas Imediatamente Precoces/genética , Neurônios Aferentes/virologia , Proteínas do Envelope Viral/genética , Proteínas Virais/genética , Animais , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/farmacologia , Sobrevivência Celular/fisiologia , Células Cultivadas , DNA Viral/genética , DNA Viral/farmacologia , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Gânglios Espinais/virologia , Deleção de Genes , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/fisiologia , Herpes Simples/fisiopatologia , Proteínas Imediatamente Precoces/metabolismo , Cloreto de Magnésio/farmacologia , MicroRNAs , Neurônios Aferentes/citologia , Neurônios Aferentes/fisiologia , Ratos , Ratos Sprague-Dawley , Transfecção , Fator de Crescimento Transformador beta/farmacologia , Gânglio Trigeminal/citologia , Proteínas do Envelope Viral/metabolismo , Latência Viral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...