Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
J Virol ; 90(8): 3873-3889, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26819314

RESUMEN

UNLABELLED: Latent membrane protein 1 (LMP1) is a major oncogene essential for primary B cell transformation by Epstein-Barr virus (EBV). Previous studies suggested that some transcription factors, such as PU.1, RBP-Jκ, NF-κB, and STAT, are involved in this expression, but the underlying mechanism is unclear. Here, we identified binding sites for PAX5, AP-2, and EBF in the proximal LMP1 promoter (ED-L1p). We first confirmed the significance of PU.1 and POU domain transcription factor binding for activation of the promoter in latency III. We then focused on the transcription factors AP-2 and early B cell factor (EBF). Interestingly, among the three AP-2-binding sites in the LMP1 promoter, two motifs were also bound by EBF. Overexpression, knockdown, and mutagenesis in the context of the viral genome indicated that AP-2 plays an important role in LMP1 expression in latency II in epithelial cells. In latency III B cells, on the other hand, the B cell-specific transcription factor EBF binds to the ED-L1p and activates LMP1 transcription from the promoter. IMPORTANCE: Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) is crucial for B cell transformation and oncogenesis of other EBV-related malignancies, such as nasopharyngeal carcinoma and T/NK lymphoma. Its expression is largely dependent on the cell type or condition, and some transcription factors have been implicated in its regulation. However, these previous reports evaluated the significance of specific factors mostly by reporter assay. In this study, we prepared point-mutated EBV at the binding sites of such transcription factors and confirmed the importance of AP-2, EBF, PU.1, and POU domain factors. Our results will provide insight into the transcriptional regulation of the major oncogene LMP1.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Transactivadores/metabolismo , Factor de Transcripción AP-2/metabolismo , Proteínas de la Matriz Viral/genética , Sitios de Unión , Transformación Celular Viral , Células HEK293 , Células HeLa , Humanos , Mutación , Oncogenes , Factores del Dominio POU/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo
2.
J Virol ; 89(1): 652-68, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25355892

RESUMEN

UNLABELLED: Epstein-Barr virus (EBV), a type of oncogenic herpesvirus, is associated with human malignancies. Previous studies have shown that lytic reactivation of EBV in latently infected cells induces an ATM-dependent DNA damage response (DDR). The involvement of ATM activation has been implicated in inducing viral lytic gene transcription to promote lytic reactivation. Its contribution to the formation of a replication compartment during lytic reactivation of EBV remains poorly defined. In this study, the role of ATM in viral DNA replication was investigated in EBV-infected nasopharyngeal epithelial cells. We observed that induction of lytic infection of EBV triggers ATM activation and localization of DDR proteins at the viral replication compartments. Suppression of ATM activity using a small interfering RNA (siRNA) approach or a specific chemical inhibitor profoundly suppressed replication of EBV DNA and production of infectious virions in EBV-infected cells induced to undergo lytic reactivation. We further showed that phosphorylation of Sp1 at the serine-101 residue is essential in promoting the accretion of EBV replication proteins at the replication compartment, which is crucial for replication of viral DNA. Knockdown of Sp1 expression by siRNA effectively suppressed the replication of viral DNA and localization of EBV replication proteins to the replication compartments. Our study supports an important role of ATM activation in lytic reactivation of EBV in epithelial cells, and phosphorylation of Sp1 is an essential process downstream of ATM activation involved in the formation of viral replication compartments. Our study revealed an essential role of the ATM-dependent DDR pathway in lytic reactivation of EBV, suggesting a potential antiviral replication strategy using specific DDR inhibitors. IMPORTANCE: Epstein-Barr virus (EBV) is closely associated with human malignancies, including undifferentiated nasopharyngeal carcinoma (NPC), which has a high prevalence in southern China. EBV can establish either latent or lytic infection depending on the cellular context of infected host cells. Recent studies have highlighted the importance of the DNA damage response (DDR), a surveillance mechanism that evolves to maintain genome integrity, in regulating lytic EBV replication. However, the underlying molecular events are largely undefined. ATM is consistently activated in EBV-infected epithelial cells when they are induced to undergo lytic reactivation. Suppression of ATM inhibits replication of viral DNA. Furthermore, we observed that phosphorylation of Sp1 at the serine-101 residue, a downstream event of ATM activation, plays an essential role in the formation of viral replication compartments for replication of virus DNA. Our study provides new insights into the mechanism through which EBV utilizes the host cell machinery to promote replication of viral DNA upon lytic reactivation.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Células Epiteliales/virología , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Replicación Viral , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Línea Celular , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN , Técnicas de Silenciamiento del Gen , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo
3.
Rev Med Virol ; 24(3): 142-53, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24339346

RESUMEN

The EBV is a human gamma-herpesvirus that is associated with a variety of neoplasms. Upon primary infection, it transiently runs a short lytic program and then predominantly establishes latent infection. Only a small percentage of infected cells switch from the latent stage into the lytic cycle and produce progeny viruses. Although EBV in cancer cells is mostly in the latent state, the lytic cycle of the virus is also expected to play a pivotal role in development and maintenance of tumors because of its association with secretion of cytokines or growth factors. Moreover, if efficient artificial induction of lytic replication could somehow be achieved, development of oncolytic therapy for EBV-positive cancers would be conceivable. Thus, understanding the switching mechanism is of essential importance. Reactivation of the virus from latency is dependent on expression of the viral BZLF1 protein. The BZLF1 promoter (Zp) normally exhibits only low basal activity but is activated in response to chemical or biological inducers, such as 12-O-tetradecanoylphorbol-13-acetate, calcium ionophore, or histone deacetylase inhibitors. Transcription from the Zp is regulated by the balance between active and suppressive epigenetic histone marks, including histone acetylation, histone H3 Lysine 4 trimethylation and histone H3 lysine 27 trimethylation, being mediated by multiple transcription factors, such as myocyte enhancer factor 2, specificity protein 1, and zinc finger E-box binding homeobox. This review will focus on such molecular mechanisms by which the EBV lytic switch is controlled and discuss the physiological significance of the switching for oncogenesis.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Transactivadores/genética , Ionóforos de Calcio/farmacología , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 4/patogenicidad , Inhibidores de Histona Desacetilasas/farmacología , Histonas/genética , Histonas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transactivadores/metabolismo , Activación Viral/efectos de los fármacos , Latencia del Virus , Dedos de Zinc/genética
4.
J Biol Chem ; 288(33): 24189-99, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23836915

RESUMEN

The Epstein-Barr virus (EBV) genome is episomally maintained in latently infected cells. The viral protein EBNA1 is a bridging molecule that tethers EBV episomes to host mitotic chromosomes as well as to interphase chromatin. EBNA1 localizes to cellular chromosomes (chromatin) via its chromosome binding domains (CBDs), which are rich in glycine and arginine residues. However, the molecular mechanism by which the CBDs of EBNA1 attach to cellular chromatin is still under debate. Mutation analyses revealed that stepwise substitution of arginine residues within the CBD1 (amino acids 40-54) and CBD2 (amino acids 328-377) regions with alanines progressively impaired chromosome binding activity of EBNA1. The complete arginine-to-alanine substitutions within the CBD1 and -2 regions abolished the ability of EBNA1 to stably maintain EBV-derived oriP plasmids in dividing cells. Importantly, replacing the same arginines with lysines had minimal effect, if any, on chromosome binding of EBNA1 as well as on its ability to stably maintain oriP plasmids. Furthermore, a glycine-arginine-rich peptide derived from the CBD1 region bound to reconstituted nucleosome core particles in vitro, as did a glycine-lysine rich peptide, whereas a glycine-alanine rich peptide did not. These results support the idea that the chromosome binding of EBNA1 is mediated by electrostatic interactions between the basic amino acids within the CBDs and negatively charged cellular chromatin.


Asunto(s)
Aminoácidos Básicos/metabolismo , Cromatina/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/química , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Plásmidos/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Cromosomas Humanos/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Mutación/genética , Nucleosomas/metabolismo , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo
5.
J Virol ; 87(18): 10148-62, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23843637

RESUMEN

Reactivation of Epstein-Barr virus (EBV) from latency is dependent on expression of the viral transactivator BZLF1 protein, whose promoter (Zp) normally exhibits only low basal activity but is activated in response to chemical or biological inducers. Using a reporter assay system, we screened for factors that can activate Zp and isolated genes, including those encoding MEF2B, KLF4, and some cellular b-Zip family transcription factors. After confirming their importance and functional binding sites in reporter assays, we prepared recombinant EBV-BAC, in which the binding sites were mutated. Interestingly, the MEF2 mutant virus produced very low levels of BRLF1, another transactivator of EBV, in addition to BZLF1 in HEK293 cells. The virus failed to induce a subset of early genes, such as that encoding BALF5, upon lytic induction, and accordingly, could not replicate to produce progeny viruses in HEK293 cells, but this restriction could be completely lifted by exogenous supply of BRLF1, together with BZLF1. In B cells, induction of BZLF1 by chemical inducers was inhibited by point mutations in the ZII or the three SP1/KLF binding sites of EBV-BAC Zp, while leaky BZLF1 expression was less affected. Mutation of MEF2 sites severely impaired both spontaneous and induced expression of not only BZLF1, but also BRLF1 in comparison to wild-type or revertant virus cases. We also observed that MEF2 mutant EBV featured relatively high repressive histone methylation, such as H3K27me3, but CpG DNA methylation levels were comparable around Zp and the BRLF1 promoter (Rp). These findings shed light on BZLF1 expression and EBV reactivation from latency.


Asunto(s)
Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Factores Reguladores Miogénicos/metabolismo , Transactivadores/biosíntesis , Activación Viral , Línea Celular , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción MEF2 , Replicación Viral
6.
J Virol ; 87(7): 4060-70, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23365429

RESUMEN

Epstein-Barr virus (EBV), a human oncogenic herpesvirus that establishes a lifelong latent infection in the host, occasionally enters lytic infection to produce progeny viruses. The EBV oncogene latent membrane protein 1 (LMP1), which is expressed in both latent and lytic infection, constitutively activates the canonical NF-κB (p65) pathway. Such LMP1-mediated NF-κB activation is necessary for proliferation of latently infected cells and inhibition of viral lytic cycle progression. Actually, canonical NF-κB target gene expression was suppressed upon the onset of lytic infection. TRAF6, which is activated by conjugation of polyubiquitin chains, associates with LMP1 to mediate NF-κB signal transduction. We have found that EBV-encoded BPLF1 interacts with and deubiquitinates TRAF6 to inhibit NF-κB signaling during lytic infection. HEK293 cells with BPLF1-deficient recombinant EBV exhibited poor viral DNA replication compared with the wild type. Furthermore, exogenous expression of BPLF1 or p65 knockdown in cells restored DNA replication of BPLF1-deficient viruses, indicating that EBV BPLF1 deubiquitinates TRAF6 to inhibit NF-κB signal transduction, leading to promotion of viral lytic DNA replication.


Asunto(s)
Herpesvirus Humano 4/enzimología , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/metabolismo , Proteínas de la Matriz Viral/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral/fisiología , Análisis de Varianza , Cromosomas Artificiales Bacterianos , Cartilla de ADN/genética , Células HEK293 , Herpesvirus Humano 4/fisiología , Humanos , Immunoblotting , Inmunoprecipitación , Luciferasas , Mutagénesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Ubiquitinación
7.
J Virol ; 87(4): 2120-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221557

RESUMEN

Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) protein is known as a regulator which recognizes phosphorylated Ser/Thr-Pro motifs and increases the rate of cis and trans amide isomer interconversion, thereby altering the conformation of its substrates. We found that Pin1 knockdown using short hairpin RNA (shRNA) technology resulted in strong suppression of productive Epstein-Barr virus (EBV) DNA replication. We further identified the EBV DNA polymerase catalytic subunit, BALF5, as a Pin1 substrate in glutathione S-transferase (GST) pulldown and immunoprecipitation assays. Lambda protein phosphatase treatment abolished the binding of BALF5 to Pin1, and mutation analysis of BALF5 revealed that replacement of the Thr178 residue by Ala (BALF5 T178A) disrupted the interaction with Pin1. To further test the effects of Pin1 in the context of virus infection, we constructed a BALF5-deficient recombinant virus. Exogenous supply of wild-type BALF5 in HEK293 cells with knockout recombinant EBV allowed efficient synthesis of viral genome DNA, but BALF5 T178A could not provide support as efficiently as wild-type BALF5. In conclusion, we found that EBV DNA polymerase BALF5 subunit interacts with Pin1 through BALF5 Thr178 in a phosphorylation-dependent manner. Pin1 might modulate EBV DNA polymerase conformation for efficient, productive viral DNA replication.


Asunto(s)
ADN Viral/biosíntesis , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Isomerasa de Peptidilprolil/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Línea Celular , Centrifugación , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Técnicas de Silenciamiento del Gen , Herpesvirus Humano 4/enzimología , Herpesvirus Humano 4/genética , Humanos , Inmunoprecipitación , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/genética , Fosforilación , Mapeo de Interacción de Proteínas , Treonina/metabolismo , Proteínas Virales/genética
8.
J Virol ; 87(11): 6482-91, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23552409

RESUMEN

Epstein-Barr virus (EBV) replication proteins are transported into the nucleus to synthesize viral genomes. We here report molecular mechanisms for nuclear transport of EBV DNA polymerase. The EBV DNA polymerase catalytic subunit BALF5 was found to accumulate in the cytoplasm when expressed alone, while the EBV DNA polymerase processivity factor BMRF1 moved into the nucleus by itself. Coexpression of both proteins, however, resulted in efficient nuclear transport of BALF5. Deletion of the nuclear localization signal of BMRF1 diminished the proteins' nuclear transport, although both proteins can still interact. These results suggest that BALF5 interacts with BMRF1 to effect transport into the nucleus. Interestingly, we found that Hsp90 inhibitors or knockdown of Hsp90ß with short hairpin RNA prevented the BALF5 nuclear transport, even in the presence of BMRF1, both in transfection assays and in the context of lytic replication. Immunoprecipitation analyses suggested that the molecular chaperone Hsp90 interacts with BALF5. Treatment with Hsp90 inhibitors blocked viral DNA replication almost completely during lytic infection, and knockdown of Hsp90ß reduced viral genome synthesis. Collectively, we speculate that Hsp90 interacts with BALF5 in the cytoplasm to assist complex formation with BMRF1, leading to nuclear transport. Hsp90 inhibitors may be useful for therapy for EBV-associated diseases in the future.


Asunto(s)
Antígenos Virales/metabolismo , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Infecciones por Virus de Epstein-Barr/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Herpesvirus Humano 4/enzimología , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Antígenos Virales/genética , Núcleo Celular/genética , Núcleo Celular/virología , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Infecciones por Virus de Epstein-Barr/virología , Proteínas HSP90 de Choque Térmico/genética , Células HeLa , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Unión Proteica , Proteínas Virales/genética
9.
J Virol ; 87(12): 6693-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23552415

RESUMEN

Productive replication of the Epstein-Barr virus (EBV) occurs in discrete sites in nuclei, called replication compartments, where viral genome DNA synthesis and transcription take place. The replication compartments include subnuclear domains, designated BMRF1 cores, which are highly enriched in the BMRF1 protein. During viral lytic replication, newly synthesized viral DNA genomes are organized around and then stored inside BMRF1 cores. Here, we examined spatial distribution of viral early and late gene mRNAs within replication compartments using confocal laser scanning microscopy and three-dimensional surface reconstruction imaging. EBV early mRNAs were mainly located outside the BMRF1 cores, while viral late mRNAs were identified inside, corresponding well with the fact that late gene transcription is dependent on viral DNA replication. From these results, we speculate that sites for viral early and late gene transcription are separated with reference to BMRF1 cores.


Asunto(s)
Núcleo Celular/ultraestructura , Núcleo Celular/virología , Herpesvirus Humano 4/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Animales , Antígenos Virales/genética , Antígenos Virales/metabolismo , Línea Celular , Replicación del ADN , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Imagenología Tridimensional , Hibridación Fluorescente in Situ , Microscopía Confocal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Virales/genética
10.
Mod Pathol ; 27(7): 922-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24336154

RESUMEN

Recent reports have indicated human cytomegalovirus (HCMV) to be associated with human glioblastoma carcinogenesis. In established examples of viral carcinogenesis, viral DNA and one or more of its products have been detected in most tumor cells of biopsies in the majority of cases. To test whether HCMV is associated with human glioblastoma based on this criterion, we measured the number of viral DNA molecules per cell in both frozen and paraffin-embedded tumor biopsies from 58 patients using real-time quantitative PCR (QPCR). Immunohistochemical and fluorescence in situ hybridization (FISH) to detect HCMV proteins and genome was performed in 10 cases using formalin-fixed paraffin-embedded glioblastoma tissues. Southern blotting using DNA extracted from four glioblastoma cell lines together with immunoblotting using the four cell lines and five glioblastoma tissue samples were also performed. We further confirmed the immunoblot bands using liquid chromatography-tandem mass spectrometry assay. As a result, HCMV DNA was not detected in the tumor cells from any of the glioblastoma cases by QPCR detecting two different HCMV genes, in clear contrast to samples from patients with HCMV infection. Southern blotting and immunoblotting of cell lines and FISH using paraffin sections were all negative. However, immunoblotting and immunohistochemistry using tissue samples were partly positive, but HCMV proteins were not detected by proteomic analysis, suggesting false positivity of the analyses. As our QPCR analysis could detect 10 copies of HCMV DNA mixed with DNA extracted from 10(4) HCMV-negative cells, we conclude that HCMV is not persistent, at least in the tumor cells, of developed human glioblastoma.


Asunto(s)
Neoplasias Encefálicas/virología , Citomegalovirus/aislamiento & purificación , ADN Viral/análisis , Glioblastoma/virología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/patología , Niño , Femenino , Glioblastoma/patología , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Rev Med Virol ; 23(4): 213-20, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23255396

RESUMEN

Because virus infections elicit various cellular responses that inhibit viral replication and growth, viruses must intervene to attenuate antiviral measures in order to thrive. The genome guardian p53 plays a central part not only in DNA damage responses, inducing cell cycle arrest or apoptosis, but also in the innate host immune control of viral infections by orchestrating diverse signaling pathways originating from many different cellular receptors and sensors. Many viruses have acquired sophisticated mechanisms to regulate p53 functions by deploying subversive proteins and modulating its post-transcriptional status. In this review, we overview the mechanisms by which DNA and RNA viruses manage p53 signaling in favor of their continued survival.


Asunto(s)
Virus ADN/inmunología , Interacciones Huésped-Patógeno , Virus ARN/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Virosis/inmunología , Animales , Humanos , Evasión Inmune , Transducción de Señal , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
12.
J Virol ; 86(9): 4752-61, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22357272

RESUMEN

The Epstein-Barr virus (EBV) predominantly establishes latent infection in B cells, and the reactivation of the virus from latency is dependent on the expression of the viral BZLF1 protein. The BZLF1 promoter (Zp) normally exhibits only low basal activity but is activated in response to chemical or biological inducers, such as 12-O-tetradecanoylphorbol-13-acetate (TPA), calcium ionophores, or histone deacetylase (HDAC) inhibitors. In some cell lines latently infected with EBV, an HDAC inhibitor alone can induce BZLF1 transcription, while the treatment does not enhance expression in other cell lines, such as B95-8 or Raji cells, suggesting unknown suppressive mechanisms besides histone deacetylation in those cells. Here, we found the epigenetic modification of the BZLF1 promoter in latent Raji cells by histone H3 lysine 27 trimethylation (H3K27me3), H3K9me2/me3, and H4K20me3. Levels of active markers such as histone acetylation and H3K4me3 were low in latent cells but increased upon reactivation. Treatment with 3-deazaneplanocin A (DZNep), an inhibitor of H3K27me3 and H4K20me3, significantly enhanced the BZLF1 transcription in Raji cells when in combination with an HDAC inhibitor, trichostatin A (TSA). The knockdown of Ezh2 or Suv420h1, histone methyltransferases for H3K27me3 or H4K20me3, respectively, further proved the suppression of Zp by the methylations. Taken together, the results indicate that H3K27 methylation and H4K20 methylation are involved, at least partly, in the maintenance of latency, and histone acetylation and H3K4 methylation correlate with the reactivation of the virus in Raji cells.


Asunto(s)
Epigénesis Genética , Herpesvirus Humano 4/genética , Histonas/metabolismo , Regiones Promotoras Genéticas , Transactivadores/genética , Latencia del Virus/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular , Metilasas de Modificación del ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Decitabina , Proteína Potenciadora del Homólogo Zeste 2 , Regulación Viral de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Inhibidores de Histona Desacetilasas/farmacología , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
J Biol Chem ; 286(25): 22007-16, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21525011

RESUMEN

Reactivation of the Epstein-Barr virus from latency is dependent on expression of the BZLF1 viral immediate-early protein. The BZLF1 promoter (Zp) normally exhibits only low basal activity but is activated in response to chemical inducers such as 12-O-tetradecanoylphorbol-13-acetate and calcium ionophore. We found that Jun dimerization protein 2 (JDP2) plays a significant role in suppressing Zp activity. Reporter, EMSA, and ChIP assays of a Zp mutant virus revealed JDP2 association with Zp at the ZII cis-element, a binding site for CREB/ATF/AP-1. Suppression of Zp activity by JDP2 correlated with HDAC3 association and reduced levels of histone acetylation. Although introduction of point mutations into the ZII element of the viral genome did not increase the level of BZLF1 production, silencing of endogenous JDP2 gene expression by RNA interference increased the levels of viral early gene products and viral DNA replication. These results indicate that JDP2 plays a role as a repressor of Zp and that its replacement by CREB/ATF/AP-1 at ZII is crucial to triggering reactivation from latency to lytic replication.


Asunto(s)
Herpesvirus Humano 4/fisiología , Proteínas Represoras/metabolismo , Latencia del Virus , Silenciador del Gen , Células HEK293 , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Regiones Promotoras Genéticas/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transactivadores/genética , Transcripción Genética , Activación Viral
14.
J Biol Chem ; 286(49): 42524-42533, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22013073

RESUMEN

Epstein-Barr virus LMP1, a major oncoprotein expressed in latent infection, is critical for primary B cell transformation, functioning as a TNFR family member by aggregation in the plasma membrane resulting in constitutive activation of cellular signals, such as NF-κB, MAPK, JAK/STAT, and AKT. Although transcription of LMP1 in latent type III cells is generally under the control of the viral coactivator EBNA2, little is known about EBNA2-independent LMP1 expression in type II latency. We thus screened a cDNA library for factors that can activate the LMP1 promoter in an EBNA2-independent manner, using a reporter assay system. So far, we have screened >20,000 clones, and here identified C/EBPε as a new transcriptional activator. Exogenous expression of C/EBPα, -ß, or -ε efficiently augmented LMP1 mRNA and protein levels in EBV-positive cell lines, whereas other members of the C/EBP family exhibited modest or little activity. It has been demonstrated that LMP1 gene transcription depends on two promoter regions: proximal (ED-L1) and distal (TR-L1). Interestingly, although we first used the proximal promoter for screening, we found that C/EBP increased transcription from both promoters in latent EBV-positive cells. Mutagenesis in reporter assays and EMSA identified only one functional C/EBP binding site, through which activation of both proximal and distal promoters is mediated. Introduction of point mutations into the identified C/EBP site in EBV-BAC caused reduced LMP1 transcription from both LMP1 promoters in epithelial cells. In conclusion, C/EBP is a newly identified transcriptional activator of the LMP1 gene, independent of the EBNA2 coactivator.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica , Activación Transcripcional , Proteínas de la Matriz Viral/genética , Secuencias de Aminoácidos , Sitios de Unión , Línea Celular , Biblioteca de Genes , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Modelos Genéticos , Oncogenes , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Transcripción Genética
15.
J Virol ; 85(13): 6629-44, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21507978

RESUMEN

The regulation of human cytomegalovirus (HCMV) late gene expression by viral proteins is poorly understood, and these viral proteins could be targets for novel antivirals. HCMV open reading frames (ORFs) UL79, -87, and -95 encode proteins with homology to late gene transcription factors of murine gammaherpesvirus 68 ORFs 18, 24, and 34, respectively. To determine whether these HCMV proteins are also essential for late gene transcription of a betaherpesvirus, we mutated HCMV ORFs UL79, -87, and -95. Cells were infected with the recombinant viruses at high and low multiplicities of infection (MOIs). While viral DNA was detected with the recombinant viruses, infectious virus was not detected unless the wild-type viral proteins were expressed in trans. At a high MOI, mutation of ORF UL79, -87, or -95 had no effect on the level of major immediate-early (MIE) gene expression or viral DNA replication, but late viral gene expression from the UL44, -75, and -99 ORFs was not detected. At a low MOI, preexpression of UL79 or -87, but not UL95, in human fibroblast cells negatively affected the level of MIE viral gene expression and viral DNA replication. The products of ORFs UL79, -87, and -95 were expressed as early viral proteins and recruited to prereplication complexes (pre-RCs), along with UL44, before the initiation of viral DNA replication. All three HCMV ORFs are indispensable for late viral gene expression and viral growth. The roles of UL79, -87, and -95 in pre-RCs for late viral gene expression are discussed.


Asunto(s)
Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/metabolismo , Replicación del ADN/fisiología , Regulación Viral de la Expresión Génica , Proteínas Inmediatas-Precoces/metabolismo , Sistemas de Lectura Abierta/fisiología , Proteínas Virales/metabolismo , Células Cultivadas , Citomegalovirus/genética , ADN Viral/genética , ADN Viral/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/virología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/fisiología , Sistemas de Lectura Abierta/genética , Proteínas Virales/genética , Replicación Viral
16.
J Virol ; 85(13): 6127-35, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21490093

RESUMEN

Productive replication of Epstein-Barr virus occurs in discrete sites in nuclei, called replication compartments, where viral DNA replication proteins and host homologous recombinational repair (HRR) and mismatch repair (MMR) factors are recruited. Three-dimensional (3D) surface reconstruction imaging clarified the spatial arrangements of these factors within the replication compartments. Subnuclear domains, designated BMRF1 cores, which were highly enriched in viral polymerase processivity factor BMRF1 could be identified inside the replication compartments. Pulse-chase experiments revealed that newly synthesized viral genomes organized around the BMRF1 cores were transferred inward. HRR factors could be demonstrated mainly outside BMRF1 cores, where de novo synthesis of viral DNA was ongoing, whereas MMR factors were found predominantly inside. These results imply that de novo synthesis of viral DNA is coupled with HRR outside the cores, followed by MMR inside cores for quality control of replicated viral genomes. Thus, our approach unveiled a viral genome manufacturing plant.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Reparación del ADN , Herpesvirus Humano 4/metabolismo , Recombinación Genética , Replicación Viral , Animales , Línea Celular , Núcleo Celular/ultraestructura , Núcleo Celular/virología , Replicación del ADN , ADN Viral/genética , ADN Viral/metabolismo , Genoma Viral , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/ultraestructura , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía Confocal , Proteínas Virales/genética , Proteínas Virales/metabolismo
17.
PLoS Pathog ; 6(12): e1001158, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-21187893

RESUMEN

Productive replication of DNA viruses elicits host cell DNA damage responses, which cause both beneficial and detrimental effects on viral replication. In response to the viral productive replication, host cells attempt to attenuate the S-phase cyclin-dependent kinase (CDK) activities to inhibit viral replication. However, accumulating evidence regarding interactions between viral factors and cellular signaling molecules indicate that viruses utilize them and selectively block the downstream signaling pathways that lead to attenuation of the high S-phase CDK activities required for viral replication. In this review, we describe the sophisticated strategy of Epstein-Barr virus to cancel such "noisy" host defense signals in order to hijack the cellular environment.


Asunto(s)
Replicación del ADN/inmunología , ADN Viral/fisiología , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno/inmunología , Quinasas Ciclina-Dependientes , Genoma Viral , Herpesvirus Humano 4/genética , Transducción de Señal
18.
J Biol Chem ; 285(31): 23925-35, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20516063

RESUMEN

The transition from latent to lytic phases of the Epstein-Barr virus life cycle is triggered by expression of a viral transactivator, BZLF1, that then induces expression of the viral immediate-early and early genes. The BZLF1 protein is post-translationally modified by a small ubiquitin-related modifier-1 (SUMO-1). Here we found that BZLF1 is conjugated at lysine 12 not only by SUMO-1 but also by SUMO-2 and 3. The K12R mutant of BZLF1, which no longer becomes sumoylated, exhibits stronger transactivation than the wild-type BZLF1 in a reporter assay system as well as in the context of virus genome with nucleosomal structures. Furthermore, exogenous supply of a SUMO-specific protease, SENP, caused de-sumoylation of BZLF1 and enhanced BZLF1-mediated transactivation. Immunoprecipitation experiments proved that histone deacetylase 3 preferentially associated with the sumoylated form of BZLF1. Levels of the sumoylated BZLF1 increased as lytic replication progressed. Based on these observations, we conclude that sumoylation of BZLF1 regulates its transcriptional activity through histone modification during Epstein-Barr virus productive replication.


Asunto(s)
Herpesvirus Humano 4/metabolismo , Histona Desacetilasas/metabolismo , Proteína SUMO-1/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Línea Celular , Línea Celular Tumoral , Humanos , Modelos Genéticos , Mutación , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Neoplasias Gástricas/metabolismo , Activación Transcripcional , Proteínas Virales/genética
19.
J Virol ; 84(24): 12589-98, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20926567

RESUMEN

The Epstein-Barr virus BMRF1 DNA polymerase processivity factor, which is essential for viral genome replication, exists mainly as a C-shaped head-to-head homodimer but partly forms a ring-shaped tetramer through tail-to-tail association. Based on its molecular structure, several BMRF1 mutant viruses were constructed to examine their influence on viral replication. The R256E virus, which has a severely impaired capacity for DNA binding and polymerase processivity, failed to form replication compartments, resulting in interference of viral replication, while the C95E mutation, which impairs head-to-head contact in vitro, unexpectedly hardly affected the viral replication. Also, surprisingly, replication of the C206E virus, which is expected to have impairment of tail-to-tail contact, was severely restricted, although the mutant protein possesses the same in vitro biochemical activities as the wild type. Since the tail-to-tail contact surface is smaller than that of the head-to-head contact area, its contribution to ring formation might be essential for viral replication.


Asunto(s)
Antígenos Virales/química , Antígenos Virales/metabolismo , Herpesvirus Humano 4/fisiología , Riñón/virología , Multimerización de Proteína , Replicación Viral , Antígenos Virales/genética , Núcleo Celular/metabolismo , Células Cultivadas , Cristalografía por Rayos X , ADN Viral/genética , Técnica del Anticuerpo Fluorescente , Humanos , Riñón/citología , Riñón/metabolismo , Mutación/genética , Plásmidos , Reacción en Cadena de la Polimerasa , Unión Proteica , Conformación Proteica
20.
PLoS Pathog ; 5(7): e1000530, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19649319

RESUMEN

p53-signaling is modulated by viruses to establish a host cellular environment advantageous for their propagation. The Epstein-Barr virus (EBV) lytic program induces phosphorylation of p53, which prevents interaction with MDM2. Here, we show that induction of EBV lytic program leads to degradation of p53 via an ubiquitin-proteasome pathway independent of MDM2. The BZLF1 protein directly functions as an adaptor component of the ECS (Elongin B/C-Cul2/5-SOCS-box protein) ubiquitin ligase complex targeting p53 for degradation. Intringuingly, C-terminal phosphorylation of p53 resulting from activated DNA damage response by viral lytic replication enhances its binding to BZLF1 protein. Purified BZLF1 protein-associated ECS could be shown to catalyze ubiquitination of phospho-mimetic p53 more efficiently than the wild-type in vitro. The compensation of p53 at middle and late stages of the lytic infection inhibits viral DNA replication and production during lytic infection, suggesting that the degradation of p53 is required for efficient viral propagation. Taken together, these findings demonstrate a role for the BZLF1 protein-associated ECS ligase complex in regulation of p53 phosphorylated by activated DNA damage signaling during viral lytic infection.


Asunto(s)
Herpesvirus Humano 4/metabolismo , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Línea Celular , Línea Celular Tumoral , Proteínas Cullin/metabolismo , Herpesvirus Humano 4/crecimiento & desarrollo , Humanos , Datos de Secuencia Molecular , Fosforilación , Ubiquitinación , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA