Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
PLoS Pathog ; 15(1): e1007489, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30682185

RESUMO

The molecular basis for the formation of functional, higher-ordered macro-molecular domains is not completely known. The Kaposi's Sarcoma-Associated Herpesvirus (KSHV) genome forms a super-molecular domain structure during latent infection that is strictly dependent on the DNA binding of the viral nuclear antigen LANA to the viral terminal repeats (TR). LANA is known to form oligomeric structures that have been implicated in viral episome maintenance. In this study, we show that the LANA oligomerization interface is required for the formation of higher-order nuclear bodies that partially colocalize with DAXX, EZH2, H3K27me3, and ORC2 but not with PML. These nuclear bodies assemble at the periphery of condensed cellular chromosomes during mitotic cell division. We demonstrate that the LANA oligomerization interface contributes to the cooperative DNA binding at the viral TR and the recruitment of ORC to the viral episome. Oligomerization mutants failed to auto-regulate LANA/ORF73 transcription, and this correlated with the loss of a chromosome conformational DNA-loop between the TR and LANA promoter. Viral genomes with LANA oligomerization mutants were subject to genome rearrangements including the loss of subgenomic DNA. Our data suggests that LANA oligomerization drives stable binding to the TR and formation of an epigenetically stable chromatin architecture resulting in higher-order LANA nuclear bodies important for viral genome integrity and long-term episome persistence.


Assuntos
Antígenos Virais/metabolismo , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Antígenos Virais/genética , Linhagem Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromossomos/metabolismo , Proteínas Correpressoras , Replicação do DNA , DNA Viral/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Genoma Viral , Herpesvirus Humano 8/genética , Humanos , Corpos de Inclusão Intranuclear/metabolismo , Chaperonas Moleculares , Proteínas Nucleares/genética , Complexo de Reconhecimento de Origem , Sequências Repetidas Terminais , Latência Viral/genética
2.
PLoS Pathog ; 14(4): e1007027, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29694443

RESUMO

[This corrects the article DOI: 10.1371/journal.ppat.1006596.].

3.
PLoS Pathog ; 13(1): e1006100, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28107481

RESUMO

Kaposi's Sarcoma-associated Herpesvirus (KSHV) establishes stable latent infection in B-lymphocytes and pleural effusion lymphomas (PELs). During latency, the viral genome persists as an epigenetically constrained episome with restricted gene expression programs. To identify epigenetic regulators of KSHV latency, we screened a focused small molecule library containing known inhibitors of epigenetic factors. We identified JQ1, a Bromodomain and Extended Terminal (BET) protein inhibitor, as a potent activator of KSHV lytic reactivation from B-cells carrying episomal KSHV. We validated that JQ1 and other BET inhibitors efficiently stimulated reactivation of KSHV from latently infected PEL cells. We found that BET proteins BRD2 and BRD4 localize to several regions of the viral genome, including the LANA binding sites within the terminal repeats (TR), as well as at CTCF-cohesin sites in the latent and lytic control regions. JQ1 did not disrupt the interaction of BRD4 or BRD2 with LANA, but did reduce the binding of LANA with KSHV TR. We have previously demonstrated a cohesin-dependent DNA-loop interaction between the latent and lytic control regions that restrict expression of ORF50/RTA and ORF45 immediate early gene transcripts. JQ1 reduced binding of cohesin subunit Rad21 with the CTCF binding sites in the latency and lytic control regions. JQ1 also reduced DNA-loop interaction between latent and lytic control regions. These findings implicate BET proteins BRD2 and BRD4 in the maintenance of KSHV chromatin architecture during latency and reveal BET inhibitors as potent activators of KSHV reactivation from latency.


Assuntos
Azepinas/farmacologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Triazóis/farmacologia , Latência Viral/efeitos dos fármacos , Antígenos Virais/genética , Antígenos Virais/metabolismo , Linfócitos B/virologia , Sítios de Ligação/efeitos dos fármacos , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Regulação Viral da Expressão Gênica , Células HEK293 , Herpesvirus Humano 8/efeitos dos fármacos , Humanos , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Derrame Pleural Maligno/virologia , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Sarcoma de Kaposi/virologia , Transativadores/biossíntese , Transativadores/genética , Fatores de Transcrição/metabolismo , Ativação Viral/efeitos dos fármacos
4.
PLoS Pathog ; 13(8): e1006596, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28854249

RESUMO

Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) is a human gammaherpesvirus recognized as the principal causative agent of KS and primary effusion lymphoma (PEL). KSHV establishes persistent latent infection in B lymphocytes where viral gene expression is restricted, in part, by a cohesin-dependent chromosome conformation. Here, we show that endoplasmic reticulum (ER) stress induces a rapid, caspase-dependent cleavage of cohesin subunit RAD21. ER stress-induced cleavage of RAD21 correlated with a rapid and strong viral lytic transcriptional activation. This effect was observed in several KSHV positive PEL cells, but not in other B-cells or non-B-cell models of KSHV latency. The cleaved-RAD21 does not dissociate from viral genomes, nor disassemble from other components of the cohesin complex. However, RAD21 cleavage correlated with the disruption of the latency genome conformation as revealed by chromosome conformation capture (3C). Ectopic expression of C-terminal RAD21 cleaved form could partially induce KSHV lytic genes transcription in BCBLI cells, suggesting that ER-stress induced RAD21 cleavage was sufficient to induce KSHV reactivation from latency in PEL cells. Taken together our results reveal a novel aspect for control and maintenance of KSHV genome latency conformation mediated by stress-induced RAD21 cleavage. Our studies also suggest that RAD21 cleavage may be a general regulatory mechanism for rapid alteration of cellular chromosome conformation and cohesin-dependent transcription regulation.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Herpesvirus Humano 8/fisiologia , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Latência Viral/fisiologia , Western Blotting , Caspases/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA , Infecções por Herpesviridae/metabolismo , Humanos , Linfoma de Efusão Primária/virologia , Reação em Cadeia da Polimerase
5.
PLoS Pathog ; 12(1): e1005339, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26752713

RESUMO

Epstein-Barr Virus (EBV) transforms resting B-lymphocytes into proliferating lymphoblasts to establish latent infections that can give rise to malignancies. We show here that EBV-encoded transcriptional regulator EBNA2 drives the cooperative and combinatorial genome-wide binding of two master regulators of B-cell fate, namely EBF1 and RBP-jκ. Previous studies suggest that these B-cell factors are statically bound to target gene promoters. In contrast, we found that EBNA2 induces the formation of new binding for both RBP-jκ and EBF1, many of which are in close physical proximity in the cellular and viral genome. These newly induced binding sites co-occupied by EBNA2-EBF1-RBP-jκ correlate strongly with transcriptional activation of linked genes that are important for B-lymphoblast function. Conditional expression or repression of EBNA2 leads to a rapid alteration in RBP-jκ and EBF1 binding. Biochemical and shRNA depletion studies provide evidence for cooperative assembly at co-occupied sites. These findings reveal that EBNA2 facilitate combinatorial interactions to induce new patterns of transcription factor occupancy and gene programming necessary to drive B-lymphoblast growth and survival.


Assuntos
Linfócitos B/virologia , Transformação Celular Viral/genética , Antígenos Nucleares do Vírus Epstein-Barr/genética , Regulação Viral da Expressão Gênica/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Transativadores/genética , Proteínas Virais/genética , Western Blotting , Imunoprecipitação da Cromatina , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase
6.
EMBO J ; 31(21): 4165-78, 2012 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-23010778

RESUMO

The contribution of human subtelomeric DNA and chromatin organization to telomere integrity and chromosome end protection is not yet understood in molecular detail. Here, we show by ChIP-Seq that most human subtelomeres contain a CTCF- and cohesin-binding site within ∼1-2 kb of the TTAGGG repeat tract and adjacent to a CpG-islands implicated in TERRA transcription control. ChIP-Seq also revealed that RNA polymerase II (RNAPII) was enriched at sites adjacent to the CTCF sites and extending towards the telomere repeat tracts. Mutation of CTCF-binding sites in plasmid-borne promoters reduced transcriptional activity in an orientation-dependent manner. Depletion of CTCF by shRNA led to a decrease in TERRA transcription, and a loss of cohesin and RNAPII binding to the subtelomeres. Depletion of either CTCF or cohesin subunit Rad21 caused telomere-induced DNA damage foci (TIF) formation, and destabilized TRF1 and TRF2 binding to the TTAGGG proximal subtelomere DNA. These findings indicate that CTCF and cohesin are integral components of most human subtelomeres, and important for the regulation of TERRA transcription and telomere end protection.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Proteínas Repressoras/metabolismo , Telômero/genética , Fatores de Transcrição/genética , Transcrição Gênica , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular/genética , Células Cultivadas , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/genética , Ilhas de CpG/genética , Ensaio de Desvio de Mobilidade Eletroforética , Imunofluorescência , Humanos , Luciferases/metabolismo , Neoplasias/genética , Neoplasias/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas/genética , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Coesinas
7.
J Virol ; 88(3): 1703-13, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24257606

RESUMO

The chromatin regulatory factors CTCF and cohesin have been implicated in the coordinated control of multiple gene loci in Epstein-Barr virus (EBV) latency. We have found that CTCF and cohesin are highly enriched at the convergent and partially overlapping transcripts for the LMP1 and LMP2A genes, but it is not yet known how CTCF and cohesin may coordinately regulate these transcripts. We now show that genetic disruption of this CTCF binding site (EBVΔCTCF166) leads to a deregulation of LMP1, LMP2A, and LMP2B transcription in EBV-immortalized B lymphocytes. EBVΔCTCF166 virus-immortalized primary B lymphocytes showed a decrease in LMP1 and LMP2A mRNA and a corresponding increase in LMP2B mRNA. The reduction of LMP1 and LMP2A correlated with a loss of euchromatic histone modification H3K9ac and a corresponding increase in heterochromatic histone modification H3K9me3 at the LMP2A promoter region in EBVΔCTCF166. Chromosome conformation capture (3C) revealed that DNA loop formation with the origin of plasmid replication (OriP) enhancer was eliminated in EBVΔCTCF166. We also observed that the EBV episome copy number was elevated in EBVΔCTCF166 and that this was not due to increased lytic cycle activity. These findings suggest that a single CTCF binding site controls LMP2A and LMP1 promoter selection, chromatin boundary function, DNA loop formation, and episome copy number control during EBV latency.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Epigênese Genética , Infecções por Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/genética , Mutação , Proteínas Repressoras/metabolismo , Proteínas da Matriz Viral/genética , Sítios de Ligação , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/virologia , Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/química , Herpesvirus Humano 4/fisiologia , Histonas/genética , Histonas/metabolismo , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Repressoras/genética , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/metabolismo , Latência Viral , Coesinas
8.
PLoS Pathog ; 9(10): e1003672, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24146617

RESUMO

LANA is the KSHV-encoded terminal repeat binding protein essential for viral replication and episome maintenance during latency. We have determined the X-ray crystal structure of LANA C-terminal DNA binding domain (LANADBD) to reveal its capacity to form a decameric ring with an exterior DNA binding surface. The dimeric core is structurally similar to EBV EBNA1 with an N-terminal arm that regulates DNA binding and is required for replication function. The oligomeric interface between LANA dimers is dispensable for single site DNA binding, but is required for cooperative DNA binding, replication function, and episome maintenance. We also identify a basic patch opposite of the DNA binding surface that is responsible for the interaction with BRD proteins and contributes to episome maintenance function. The structural features of LANADBD suggest a novel mechanism of episome maintenance through DNA-binding induced oligomeric assembly.


Assuntos
Antígenos Virais/química , DNA Viral/química , Herpesvirus Humano 8/química , Proteínas Nucleares/química , Plasmídeos/química , Multimerização Proteica , Antígenos Virais/genética , Antígenos Virais/metabolismo , Linhagem Celular Tumoral , Replicação do DNA , DNA Viral/genética , DNA Viral/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
9.
J Virol ; 87(7): 3699-709, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23325691

RESUMO

Kaposi's Sarcoma-associated herpesvirus (KSHV) is maintained as a stable episome in latently infected pleural effusion lymphoma (PEL) cells. Episome maintenance is conferred by the binding of the KSHV-encoded LANA protein to the viral terminal repeats (TR). Here, we show that DNA replication in the KSHV TR is coupled with DNA recombination and mediated in part through the cellular replication fork protection factors Timeless (Tim) and Tipin. We show by two-dimensional (2D) agarose gel electrophoresis that replication forks naturally stall and form recombination-like structures at the TR during an unperturbed cell cycle. Chromatin immunoprecipitation (ChIP) assays revealed that Tim and Tipin are selectively enriched at the KSHV TR during S phase and in a LANA-dependent manner. Tim depletion inhibited LANA-dependent TR DNA replication and caused the loss of KSHV episomes from latently infected PEL cells. Tim depletion resulted in the aberrant accumulation of recombination structures and arrested MCM helicase at TR. Tim depletion did not induce the KSHV lytic cycle or apoptotic cell death. We propose that KSHV episome maintenance requires Tim-assisted replication fork protection at the viral terminal repeats and that Tim-dependent recombination-like structures form at TR to promote DNA repeat stability and viral genome maintenance.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Replicação do DNA/fisiologia , Instabilidade Genômica/fisiologia , Herpesvirus Humano 8/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Plasmídeos/fisiologia , Recombinação Genética/fisiologia , Antígenos Virais/metabolismo , Bromodesoxiuridina , Proteínas de Transporte/metabolismo , Imunoprecipitação da Cromatina , Primers do DNA/genética , Proteínas de Ligação a DNA , Eletroforese em Gel de Campo Pulsado , Eletroforese em Gel Bidimensional , Citometria de Fluxo , Instabilidade Genômica/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Proteínas Nucleares/metabolismo , Plasmídeos/genética , Sequências Repetidas Terminais/genética , Sequências Repetidas Terminais/fisiologia
10.
J Virol ; 86(17): 9454-64, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22740398

RESUMO

Chromatin-organizing factors such as CTCF and cohesins have been implicated in the control of complex viral regulatory programs. We investigated the role of CTCF and cohesins in the control of the switch from latency to the lytic cycle for Kaposi's sarcoma-associated herpesvirus (KSHV). We found that cohesin subunits but not CTCF are required for the repression of KSHV immediate early gene transcription. Depletion of the cohesin subunits Rad21, SMC1, and SMC3 resulted in lytic cycle gene transcription and viral DNA replication. In contrast, depletion of CTCF failed to induce lytic transcription or DNA replication. Chromatin immunoprecipitation with high-throughput sequencing (ChIP-Seq) revealed that cohesins and CTCF bound to several sites within the immediate early control region for ORF50 and to more distal 5' sites that also regulate the divergently transcribed ORF45-ORF46-ORF47 gene cluster. Rad21 depletion led to a robust increase in ORF45, ORF46, ORF47, and ORF50 transcripts, with similar kinetics to that observed with chemical induction by sodium butyrate. During latency, the chromatin between the ORF45 and ORF50 transcription start sites was enriched in histone H3K4me3, with elevated H3K9ac at the ORF45 promoter and elevated H3K27me3 at the ORF50 promoter. A paused form of RNA polymerase II (Pol II) was loosely associated with the ORF45 promoter region during latency but was converted to an active elongating form upon reactivation induced by Rad21 depletion. Butyrate treatment caused a rapid dissociation of cohesins and loss of CTCF binding at the immediate early gene locus, suggesting that cohesins may be a direct target of butyrate-mediated lytic induction. Our findings implicate cohesins as a major repressor of KSHV lytic gene activation and show that they function coordinately with CTCF to regulate the switch between latent and lytic gene activity.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Regulação para Baixo , Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/genética , Transativadores/genética , Transcrição Gênica , Latência Viral , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Regulação Viral da Expressão Gênica , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Proteínas Imediatamente Precoces/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Coesinas
11.
J Virol ; 86(10): 5752-62, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22419807

RESUMO

LANA is essential for tethering the Kaposi's sarcoma-associated herpesvirus (KSHV) genome to metaphase chromosomes and for modulating host-cell gene expression, but the binding sites in the host-chromosome remain unknown. Here, we use LANA-specific chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-Seq) to identify LANA binding sites in the viral and host-cell genomes of a latently infected pleural effusion lymphoma cell line BCBL1. LANA bound with high occupancy to the KSHV genome terminal repeats (TR) and to a few minor binding sites in the KSHV genome, including the LANA promoter region. We identified 256 putative LANA binding site peaks with P < 0.01 and overlap in two independent ChIP-Seq experiments. We validated several of the high-occupancy binding sites by conventional ChIP assays and quantitative PCR. Candidate cellular LANA binding motifs were identified and assayed for binding to purified recombinant LANA protein in vitro but bound with low affinity compared to the viral TR binding site. More than half of the LANA binding sites (170/256) could be mapped to within 2.5 kb of a cellular gene transcript. Pathways and Gene Ontogeny (GO) analysis revealed that LANA binds to genes within the p53 and tumor necrosis factor (TNF) regulatory network. Further analysis revealed partial overlap of LANA and STAT1 binding sites in several gamma interferon (IFN-γ)-regulated genes. We show that ectopic expression of LANA can downmodulate IFN-γ-mediated activation of a subset of genes, including the TAP1 peptide transporter and proteasome subunit beta type 9 (PSMB9), both of which are required for class I antigen presentation. Our data provide a potential mechanism through which LANA may regulate several host cell pathways by direct binding to gene regulatory elements.


Assuntos
Antígenos Virais/metabolismo , Cromossomos Humanos/virologia , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/metabolismo , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virologia , Antígenos Virais/química , Antígenos Virais/genética , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Cromossomos Humanos/química , Regulação da Expressão Gênica , Marcação de Genes , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Interações Hospedeiro-Patógeno , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica
12.
J Virol ; 82(2): 1053-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17989183

RESUMO

Epidemiological studies suggest that cigarette smoke carcinogens are cofactors which synergize with human papillomavirus (HPV) to increase the risk of cervical cancer progression. Benzo[a]pyrene (BaP), a major carcinogen in cigarette smoke, is detected in the cervical mucus and may interact with HPV. Exposure of cervical cells to high concentrations of BaP resulted in a 10-fold increase in HPV type 31 (HPV31) viral titers, whereas treatment with low concentrations of BaP resulted in an increased number of HPV genome copies but not an increase in virion morphogenesis. BaP exposure also increased HPV16 and HPV18 viral titers. Overall, BaP modulation of the HPV life cycle could potentially enhance viral persistence, host tissue carcinogenesis, and permissiveness for cancer progression.


Assuntos
Benzo(a)pireno/farmacologia , Carcinógenos/farmacologia , Papillomaviridae/efeitos dos fármacos , Papillomaviridae/crescimento & desenvolvimento , DNA Viral/biossíntese , Humanos
13.
J Chemother ; 18(2): 209-16, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16736891

RESUMO

The present study describes 31 clinical cases of neuroendocrine cervical carcinoma (NECC) treated at Mackay Memorial Hospital between January 1, 1991 and October 31, 2003. There are two cases of atypical carcinoid tumor (ACT), four cases of large-cell neuroendocrine carcinoma (LCNEC), and 25 cases of small-cell neuroendocrine carcinoma (SCNEC). Overall survival did not differ significantly in relation to surgery, tumor histology, age, FIGO stages, chemotherapeutic regimens or lymph node involvement. The specimens available did not permit HPV (human papillomavirus)-DNA analysis in 5 cases (5/31, 9.7%). The HPV viral infection was absent in 8 cases (8/31, 26%); 17 cases of HPV-18 (17/31); and 1 case of HPV-16 (1/31). The prognosis between mixed and pure type histologic patterns is not significant. The mean survival time for all patients was 32.3 months. The 2-year and 5-year survival rates were 54.8% and 31.5% for all patients. The results of this study reaffirm the biologically aggressive nature of this rare malignancy, its low survival rate, and its very unpredictable prognostic factors. Effective treatments of neuroendocrine cervical tumor still remain inconclusive. Further efforts are still required to identify prognostic factors for this uncommon disease.


Assuntos
Carcinoma Neuroendócrino/patologia , Neoplasias do Colo do Útero/patologia , Adulto , Tumor Carcinoide/patologia , Tumor Carcinoide/virologia , Carcinoma de Células Grandes/patologia , Carcinoma de Células Grandes/virologia , Carcinoma Neuroendócrino/virologia , Carcinoma de Células Pequenas/patologia , Carcinoma de Células Pequenas/virologia , Colo do Útero/patologia , Colo do Útero/virologia , Sondas de DNA de HPV , Feminino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/metabolismo , Humanos , Pessoa de Meia-Idade , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/complicações , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Neoplasias do Colo do Útero/virologia
14.
Curr Opin Virol ; 3(3): 251-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23601957

RESUMO

The gammaherpesviruses are unique for their capacity to establish a variety of gene expression programs during latent and lytic infection. This capacity enables the virus to control host-cell proliferation, prevent programmed cell death, elude immune cell detection, and ultimately adapt to a wide range of environmental and developmental changes in the host cell. This remarkable plasticity of gene expression results from the combined functionalities of viral and host factors that biochemically remodel and epigenetically modify the viral chromosome. These epigenetic modifications range from primary DNA methylations, to chromatin protein post-translational modifications, to higher-order chromosome conformations. In addition, gammaherpesviruses have acquired specialized tools to modulate the epigenetic processes that promote viral genome propagation and host-cell survival.


Assuntos
Epigênese Genética , Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/fisiologia , Herpesvirus Humano 8/fisiologia , Latência Viral , Metilação de DNA , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Histonas/metabolismo , Humanos , Processamento de Proteína Pós-Traducional
15.
Cell Host Microbe ; 12(2): 233-45, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22901543

RESUMO

Epstein-Barr virus (EBV), which is associated with multiple human tumors, persists as a minichromosome in the nucleus of B lymphocytes and induces malignancies through incompletely understood mechanisms. Here, we present a large-scale functional genomic analysis of EBV. Our experimentally generated nucleosome positioning maps and viral protein binding data were integrated with over 700 publicly available high-throughput sequencing data sets for human lymphoblastoid cell lines mapped to the EBV genome. We found that viral lytic genes are coexpressed with cellular cancer-associated pathways, suggesting that the lytic cycle may play an unexpected role in virus-mediated oncogenesis. Host regulators of viral oncogene expression and chromosome structure were identified and validated, revealing a role for the B cell-specific protein Pax5 in viral gene regulation and the cohesin complex in regulating higher order chromatin structure. Our findings provide a deeper understanding of latent viral persistence in oncogenesis and establish a valuable viral genomics resource for future exploration.


Assuntos
Infecções por Vírus Epstein-Barr/virologia , Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/genética , Interações Hospedeiro-Patógeno , Transcriptoma , Epigenômica , Infecções por Vírus Epstein-Barr/genética , Herpesvirus Humano 4/fisiologia , Humanos , Fator de Transcrição PAX5/genética , Fator de Transcrição PAX5/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
16.
Virology ; 412(2): 378-83, 2011 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-21329956

RESUMO

We studied a panel of mutant viruses containing wild-type and chimeric capsid HPV16 and HPV18 proteins. The mutant capsid protein expression, genome amplification, and episomal maintenance were comparable with the wild-type virus. However, the chimeric viruses varied in their titers from wild-type. We show that the intertypical mutant chimeric capsid viruses, that L2 affects the structure of L1 and that L1 affects the structure of L2 in the virion. These effects were measured using a panel of conformation-dependent neutralizing L1 MAbs and an L2 capsid surface peptide derived neutralizing antibody. These data suggest that variation of one capsid gene not only affects its own structure and antigenicity, but also affects the structure and antigenicity of the other capsid protein. Implications of our data suggest that for the continued effectiveness of a vaccine, variation in both capsid proteins need to be considered and not just the protein the vaccine is directed against.


Assuntos
Proteínas do Capsídeo/metabolismo , Papillomavirus Humano 16/fisiologia , Papillomavirus Humano 18/fisiologia , Montagem de Vírus , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/imunologia , Papillomavirus Humano 16/imunologia , Papillomavirus Humano 18/imunologia , Humanos , Recombinação Genética
17.
Virus Res ; 160(1-2): 246-55, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21762735

RESUMO

Papillomaviruses (PVs) comprise a large family of viruses infecting nearly all vertebrate species, with more than 100 human PVs identified. Our previous studies showed that a mutant chimera HPV18/16 genome, consisting of the upper regulatory region and early ORFs of HPV18 and the late ORFs of HPV16, was capable of producing infectious virus in organotypic raft cultures. We were interested in determining whether the ability of this chimeric genome to produce infectious virus was the result of HPV18 and HPV16 being similarly oncogenic, anogenital types and whether more disparate PV types could also interact functionally. To test this we created a series of HPV18 chimeric genomes where the ORFs for the HPV18 capsid genes were replaced with the capsid genes of HPV45, HPV39, HPV33, HPV31, HPV11, HPV6b, HPV1a, CRPV, and BPV1. All chimeras were able to produce infectious chimeric viral particles, although with lower infectivity than wild-type HPV18. Steps in the viral life cycle and characteristics of the viral particles were examined to identify potential causes for the decrease in infectivity.


Assuntos
Proteínas do Capsídeo/genética , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/patogenicidade , Proteínas Oncogênicas Virais/genética , Linhagem Celular , Humanos , Recombinação Genética
18.
Virology ; 405(2): 289-99, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20598725

RESUMO

Using the HPV18 genome as the backbone, we exchanged the HPV18 L2 or L1 genes with those of HPV16. The intertypical exchange of HPV18 L1 with the HPV16 L1 produced genomes that efficiently replicated and produced infectious virus. Genomes containing an intertypical exchange of HPV18 L2 for the HPV16 L2 failed to produce infectious virus in multiple independently derived cell lines. Using chimeric constructs of individual capsid proteins, we identified a type-specific domain at the N-terminus of the HPV18L1 capsid protein, which interferes with its ability to cooperate with the HPV16 L2 protein to form infectious viral particles. Deletion of this domain allows for the cooperation of the HPV18 L1 protein and HPV16 L2 protein and production of infectious progeny. In addition, cooperation of this N-terminal HPV18 L1 deletion mutant protein with the wild-type HPV18 L2 protein efficiently replicates infectious virus but changes occur in the viral structure.


Assuntos
Proteínas do Capsídeo/metabolismo , Papillomavirus Humano 16/patogenicidade , Papillomavirus Humano 18 , Recombinação Genética , Vírion , Montagem de Vírus , Sequência de Aminoácidos , Capsídeo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Células Cultivadas , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/metabolismo , Papillomavirus Humano 18/patogenicidade , Humanos , Queratinócitos , Dados de Sequência Molecular , Mutação , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Técnicas de Cultura de Órgãos , Proteínas Recombinantes de Fusão/metabolismo , Vírion/metabolismo , Vírion/patogenicidade
19.
J Virol ; 77(10): 6076-81, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12719601

RESUMO

Borna disease virus (BDV) infection of Lewis rats is the most studied animal model of Borna disease, an often fatal encephalomyelitis. In this experimental model, BDV-specific CD8(+) cytotoxic T lymphocytes (CTLs) play a prominent role in the immunopathogenesis of infection by the noncytolytic, persistent BDV. Of the six open reading frames of BDV, CTLs to BDV X (p10) and the L-polymerase have never been studied. In this study, we used plasmid immunization to investigate the CTL response to BDV X and N. Plasmid-based immunization was a potent CTL inducer in Lewis rats. Anti-X CTLs were primed by a single injection of the p10 cDNA. Two codominant p10 epitopes, M(1)SSDLRLTLL(10) and T(8)LLELVRRL(16), associated with the RT1.A(l) major histocompatibility complex class I molecules of the Lewis rats, were identified. In addition, immunization with a BDV p40-expressing plasmid confirmed the previously reported RT1.A(l)-restricted A(230)SYAQMTTY(238) peptide as the CTL target for BDV N. In contrast to the CTL responses, plasmid vaccination was a poor inducer of an antibody response to p10. Three injections of a recombinant eukaryotic expression plasmid of BDV p10 were needed to generate a weak anti-p10 immunoglobulin M response. However, the antibody response could be optimized by a protein boost after priming with cDNA.


Assuntos
Vírus da Doença de Borna/imunologia , Epitopos de Linfócito T/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/imunologia , Proteínas Virais/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/sangue , Doença de Borna/imunologia , Doença de Borna/prevenção & controle , Mapeamento de Epitopos , Epitopos de Linfócito T/química , Imunização , Dados de Sequência Molecular , Plasmídeos , Ratos , Ratos Endogâmicos Lew , Proteínas Virais/genética , Vacinas Virais/genética , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA