Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32641480

RESUMEN

We previously reported that the cellular transcription factor hypoxia-inducible factor 1α (HIF-1α) binds a hypoxia response element (HRE) located within the promoter of Epstein-Barr virus's (EBV's) latent-lytic switch BZLF1 gene, Zp, inducing viral reactivation. In this study, EBV-infected cell lines derived from gastric cancers and Burkitt lymphomas were incubated with HIF-1α-stabilizing drugs: the iron chelator deferoxamine (Desferal [DFO]), a neddylation inhibitor (pevonedistat [MLN-4924]), and a prolyl hydroxylase inhibitor (roxadustat [FG-4592]). DFO and MLN-4924, but not FG-4592, induced accumulation of both lytic EBV proteins and phosphorylated p53 in cell lines that contain a wild-type p53 gene. FG-4592 also failed to activate transcription from Zp in a reporter assay despite inducing accumulation of HIF-1α and transcription from another HRE-containing promoter. Unexpectedly, DFO failed to induce EBV reactivation in cell lines that express mutant or no p53 or when p53 expression was knocked down with short hairpin RNAs (shRNAs). Likewise, HIF-1α failed to activate transcription from Zp when p53 was knocked out by CRISPR-Cas9. Importantly, DFO induced binding of p53 as well as HIF-1α to Zp in chromatin immunoprecipitation (ChIP) assays, but only when the HRE was present. Nutlin-3, a drug known to induce accumulation of phosphorylated p53, synergized with DFO and MLN-4924 in inducing EBV reactivation. Conversely, KU-55933, a drug that inhibits ataxia telangiectasia mutated, thereby preventing p53 phosphorylation, inhibited DFO-induced EBV reactivation. Lastly, activation of Zp transcription by DFO and MLN-4924 mapped to its HRE. Thus, we conclude that induction of BZLF1 gene expression by HIF-1α requires phosphorylated, wild-type p53 as a coactivator, with HIF-1α binding recruiting p53 to Zp.IMPORTANCE EBV, a human herpesvirus, is latently present in most nasopharyngeal carcinomas, Burkitt lymphomas, and some gastric cancers. To develop a lytic-induction therapy for treating patients with EBV-associated cancers, we need a way to efficiently reactivate EBV into lytic replication. EBV's BZLF1 gene product, Zta, usually controls this reactivation switch. We previously showed that HIF-1α binds the BZLF1 gene promoter, inducing Zta synthesis, and HIF-1α-stabilizing drugs can induce EBV reactivation. In this study, we determined which EBV-positive cell lines are reactivated by classes of HIF-1α-stabilizing drugs. We found, unexpectedly, that HIF-1α-stabilizing drugs only induce reactivation when they also induce accumulation of phosphorylated, wild-type p53. Fortunately, p53 phosphorylation can also be provided by drugs such as nutlin-3, leading to synergistic reactivation of EBV. These findings indicate that some HIF-1α-stabilizing drugs may be helpful as part of a lytic-induction therapy for treating patients with EBV-positive malignancies that contain wild-type p53.


Asunto(s)
Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Transactivadores/genética , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Ciclopentanos/farmacología , Deferoxamina/farmacología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/virología , Regulación de la Expresión Génica , Glicina/análogos & derivados , Glicina/farmacología , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/agonistas , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Imidazoles/farmacología , Quelantes del Hierro/farmacología , Isoquinolinas/farmacología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Linfocitos/virología , Morfolinas/farmacología , Piperazinas/farmacología , Inhibidores de Prolil-Hidroxilasa/farmacología , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Pirimidinas/farmacología , Pironas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Elementos de Respuesta , Transducción de Señal , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Activación Viral/efectos de los fármacos
2.
PLoS Pathog ; 13(6): e1006404, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28617871

RESUMEN

When confronted with poor oxygenation, cells adapt by activating survival signaling pathways, including the oxygen-sensitive transcriptional regulators called hypoxia-inducible factor alphas (HIF-αs). We report here that HIF-1α also regulates the life cycle of Epstein-Barr virus (EBV). Incubation of EBV-positive gastric carcinoma AGS-Akata and SNU-719 and Burkitt lymphoma Sal and KemIII cell lines with a prolyl hydroxylase inhibitor, L-mimosine or deferoxamine, or the NEDDylation inhibitor MLN4924 promoted rapid and sustained accumulation of both HIF-1α and lytic EBV antigens. ShRNA knockdown of HIF-1α significantly reduced deferoxamine-mediated lytic reactivation. HIF-1α directly bound the promoter of the EBV primary latent-lytic switch BZLF1 gene, Zp, activating transcription via a consensus hypoxia-response element (HRE) located at nt -83 through -76 relative to the transcription initiation site. HIF-1α did not activate transcription from the other EBV immediate-early gene, BRLF1. Importantly, expression of HIF-1α induced EBV lytic-gene expression in cells harboring wild-type EBV, but not in cells infected with variants containing base-pair substitution mutations within this HRE. Human oral keratinocyte (NOK) and gingival epithelial (hGET) cells induced to differentiate by incubation with either methyl cellulose or growth in organotypic culture accumulated both HIF-1α and Blimp-1α, another cellular factor implicated in lytic reactivation. HIF-1α activity also accumulated along with Blimp-1α during B-cell differentiation into plasma cells. Furthermore, most BZLF1-expressing cells observed in lymphomas induced by EBV in NSG mice with a humanized immune system were located distal to blood vessels in hypoxic regions of the tumors. Thus, we conclude that HIF-1α plays central roles in both EBV's natural life cycle and EBV-associated tumorigenesis. We propose that drugs that induce HIF-1α protein accumulation are good candidates for development of a lytic-induction therapy for treating some EBV-associated malignancies.


Asunto(s)
Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Linfoma/metabolismo , Transactivadores/genética , Animales , Linfocitos B/metabolismo , Linfocitos B/virología , Carcinogénesis , Línea Celular Tumoral , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Linfoma/genética , Linfoma/virología , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Transactivadores/metabolismo , Activación Viral
3.
J Virol ; 86(15): 8086-96, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623769

RESUMEN

The BZLF1 gene controls the switch between latent and lytic infection by Epstein-Barr virus (EBV). We previously reported that both the ZV and ZIIR elements within the BZLF1 promoter, Zp, are potent transcription silencers within the context of an intact EBV genome. We report here identification of another sequence element, ZV', which synergized with ZV in repressing Zp via binding ZEB1 or ZEB2. We then determined the phenotype of a variant of EBV strain B95.8 in which the ZV, ZV', and ZIIR elements were concurrently mutated. HEK293 cell lines infected with this triple mutant (tmt) virus spontaneously synthesized 6- to 10-fold more viral BZLF1, BRLF1, BMRF1, and BLLF1 RNAs, 3- to 6-fold more viral Zta, Rta, and EAD proteins, 3- to 5-fold more viral DNA, and 7- to 9-fold more infectious virus than did 293 cell lines latently infected with either the ZV ZV' double mutant (dmt) or ZIIR mutant (mt) virus. While ZV ZV' ZIIR tmt EBV efficiently infected human primary blood B cells in vitro, it was highly defective in immortalizing them. Instead of the nearly complete silencing of BZLF1 gene expression that occurs within 4 days after primary infection with wild-type EBV, the ZV ZV' ZIIR tmt-infected cells continued to synthesize BZLF1 RNA, with 90% of them dying within 9 days postinfection. BL41 cells infected with this "superlytic" virus also exhibited increased synthesis of BZLF1 and BMRF1 RNAs. Thus, we conclude that the ZV, ZV', and ZIIR silencing elements act synergistically to repress transcription from Zp, thereby tightly controlling BZLF1 gene expression, which is crucial for establishing and maintaining EBV latency.


Asunto(s)
Linfocitos B/metabolismo , Transformación Celular Viral , Infecciones por Virus de Epstein-Barr/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 4/fisiología , Transactivadores/biosíntesis , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Linfocitos B/patología , Linfocitos B/virología , Línea Celular Tumoral , Infecciones por Virus de Epstein-Barr/genética , Genoma Viral/fisiología , Células HeLa , Humanos , Mutación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Viral/biosíntesis , ARN Viral/genética , Elementos Silenciadores Transcripcionales/fisiología , Factores de Tiempo , Transactivadores/genética , Transcripción Genética/fisiología , Latencia del Virus/fisiología
4.
J Virol ; 86(15): 7976-87, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623780

RESUMEN

Immunosuppressed patients are at risk for developing Epstein-Barr Virus (EBV)-positive lymphomas that express the major EBV oncoprotein, LMP1. Although increasing evidence suggests that a small number of lytically infected cells may promote EBV-positive lymphomas, the impact of enhanced lytic gene expression on the ability of EBV to induce lymphomas is unclear. Here we have used immune-deficient mice, engrafted with human fetal hematopoietic stem cells and thymus and liver tissue, to compare lymphoma formation following infection with wild-type (WT) EBV versus infection with a "superlytic" (SL) mutant with enhanced BZLF1 (Z) expression. The same proportions (2/6) of the WT and SL virus-infected animals developed B-cell lymphomas by day 60 postinfection; the remainder of the animals had persistent tumor-free viral latency. In contrast, all WT and SL virus-infected animals treated with the OKT3 anti-CD3 antibody (which inhibits T-cell function) developed lymphomas by day 29. Lymphomas in OKT3-treated animals (in contrast to lymphomas in the untreated animals) contained many LMP1-expressing cells. The SL virus-infected lymphomas in both OKT3-treated and untreated animals contained many more Z-expressing cells (up to 30%) than the WT virus-infected lymphomas, but did not express late viral proteins and thus had an abortive lytic form of EBV infection. LMP1 and BMRF1 (an early lytic viral protein) were never coexpressed in the same cell, suggesting that LMP1 expression is incompatible with lytic viral reactivation. These results show that the SL mutant induces an "abortive" lytic infection in humanized mice that is compatible with continued cell growth and at least partially resistant to T-cell killing.


Asunto(s)
Infecciones por Virus de Epstein-Barr/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/metabolismo , Linfoma/metabolismo , Linfoma/virología , Mutación , Transactivadores/biosíntesis , Animales , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Humanos , Linfoma/genética , Linfoma/patología , Ratones , Ratones Mutantes , Linfocitos T/metabolismo , Linfocitos T/patología , Linfocitos T/virología , Transactivadores/genética , Latencia del Virus/genética
5.
J Virol ; 85(10): 5081-90, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21389123

RESUMEN

The Epstein-Barr virus (EBV) BZLF1 gene encodes the immediate-early (IE) protein Zta, which plays a central role in regulating the switch between viral latency and lytic replication. A silencing element, ZIIR, is located between the ZID and ZII positive regulatory elements in the BZLF1 promoter Zp. We report here the phenotypes of variants of EBV strain B95.8 containing base substitution mutations in this ZIIR element. HEK293 cells infected with ZIIR mutant (ZIIRmt) virus produced at least 20-fold more viral IE Zta and Rta and early (E) EAD protein than did cells infected with the parental wild-type (WT) virus, leading to viral DNA replication and production of infectious virus. However, ZIIR mutant virus was 1/10 as efficient as WT virus in establishing proliferating B-cell clones following infection of human primary blood B cells. The ZIIRmt-infected lymphoblastoid cell lines (LCLs) that did grow out exhibited a phenotype similar to the one observed in 293 cells, including marked overproduction of IE and E gene products relative to WT-infected LCLs and lytic replication of the viral genome. Incubation of the ZIIRmt-infected LCLs with the chemical inducer 12-O-tetradecanoyl-phorbol-13-acetate (TPA) led to much greater activation of Zp than did the same treatment of WT- or ZVmt-infected LCLs. Furthermore, a protein kinase C (PKC) inhibitor, bis-indolylmaleimide, eliminated this activation by TPA. Thus, we conclude that ZIIR is a potent silencing element of Zp; it plays a key role in establishment and maintenance of EBV latency by inhibiting activation of Zp through the PKC signal transduction pathway.


Asunto(s)
Herpesvirus Humano 4/fisiología , Regiones Promotoras Genéticas , Transactivadores/genética , Latencia del Virus , Linfocitos B/virología , Línea Celular , Herpesvirus Humano 4/genética , Humanos , Mutagénesis Sitio-Dirigida , Transactivadores/metabolismo
6.
Breast Cancer Res Treat ; 128(2): 527-34, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21293922

RESUMEN

Activated Leukocyte Cell Adhesion Molecules (ALCAM, also called CD166, MEMD) are cell surface immunoglobulins that are considered to be prognostic markers for breast cancer. CD166/ALCAM has gained increasing attention because of its significant association with tumor progression and the metastatic spread of breast cancer. Two polymorphisms have been identified in the CD166/ALCAM gene: 5'UTR C/T (rs6437585) and 3'UTR A/G (rs11559013). We analyzed the genotypes of 1033 individuals with breast cancer, and 1116 controls; odds ratios (ORs) and 95% confidence intervals (CIs) were estimated using logistic regression. The effects and functions of polymorphisms were examined using luciferase gene expression assays and real-time PCR analyses. Our data demonstrated that individuals with the rs6437585 CT + TT genotype had an OR of 1.38 (95% CI, 1.11-1.72) for developing breast cancer, compared to those with the CC genotype. The T allele increased the risk of breast cancer in a dose-dependent manner (P (trend) < 0.001). However, there were no significant differences found between cases and controls at the rs11559013 A/G site. Additional experiments that we performed, which focused on reporter gene expression driven by CD166/ALCAM promoters, demonstrated that the presence of an rs6437585 T allele led to greater transcriptional activity than the rs6437585 C allele. This was consistent with the increased cancer risk that we observed in our case-control analysis.


Asunto(s)
Antígenos CD/genética , Pueblo Asiatico/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Moléculas de Adhesión Celular Neuronal/genética , Proteínas Fetales/genética , Polimorfismo Genético/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/epidemiología , Carcinoma Ductal de Mama/secundario , Estudios de Casos y Controles , China/epidemiología , ADN de Neoplasias/genética , Femenino , Genotipo , Humanos , Luciferasas/metabolismo , Persona de Mediana Edad , Oportunidad Relativa , Reacción en Cadena de la Polimerasa , Pronóstico , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Neoplásico/genética , Factores de Riesgo , Adulto Joven
7.
Breast Cancer Res Treat ; 128(2): 467-72, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21221763

RESUMEN

Rare alleles at the HRAS1 variable number of tandem repeats (VNTRs) locus have been implicated in breast cancer risk. Although many studies have showed that rare HRAS1 alleles may be associated with breast cancer risk, this relationship remains controversial. A meta-analysis was conducted to investigate the potential association between rare HRAS1 alleles and breast cancer risk. A database search found a total of 13 studies involving 1926 breast cancer cases and 2800 controls. Crude odds ratios (OR) and 95% confidence intervals (CI) were used to test the strength of association. When all the studies were combined into the meta-analysis, we found that breast cancer cases had a significantly higher frequency of rare alleles (OR = 2.03, 95% CI = 1.34, 3.10). In the subgroup analysis by race, we found that breast cancer cases had a significantly higher frequency of rare alleles (OR = 2.14, 95% CI = 1.37, 3.36) among Caucasians. In the subgroup analysis by study design, we found that breast cancer cases had a significantly higher frequency of rare alleles (OR = 2.47, 95% CI = 1.62, 3.79) among groups with hospital-based controls. In conclusion, this meta-analysis suggested that rare alleles at the HRAS1 VNTRs may contribute to breast cancer susceptibility. More population-based case-control studies were needed especially in Asians in the future.


Asunto(s)
Neoplasias de la Mama/genética , Predisposición Genética a la Enfermedad , Repeticiones de Minisatélite/genética , Polimorfismo Genético/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Estudios de Casos y Controles , Femenino , Humanos , Pronóstico , Factores de Riesgo
8.
Breast Cancer Res Treat ; 127(3): 813-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21161369

RESUMEN

The matrix metalloproteinases (MMP) can degrade various components of the extracellular matrix and its functional genetic polymorphisms may be associated with breast cancer risk. However, this relationship remains controversial. A meta-analysis was conducted in order to investigate the potential association between four polymorphisms in the MMP gene and breast cancer risk. A database search yielded a total of 9 studies involving 2,597 cases and 2,618 controls. Four polymorphisms were included in the meta-analysis: MMP-1 -1607 2G/1G (rs1799750), MMP-2 -1306 C/T (rs243865), MMP-3 -1171 6A/5A (rs3025058) and MMP-9 -1562 C/T (rs3918242). Crude odds ratios (OR) with 95% confidence intervals (CI) were used to assess the strength of association. When all the studies were pooled into the meta-analysis, we found that breast cancer cases had a significantly higher frequency of CC genotype (OR = 1.27, 95% CI = 1.10, 1.47; P = 0.001) and lower frequency of CT genotype (OR = 0.78, 95% CI = 0.67, 0.91; P = 0.001) of MMP-2. No significant difference was found in any genotype of MMP-1, MMP-3 or MMP-9. In conclusion, this meta-analysis suggested that MMP-2 -1306 C/T polymorphism may contribute to breast cancer susceptibility. More studies were needed especially in Asians in the future.


Asunto(s)
Neoplasias de la Mama/genética , Predisposición Genética a la Enfermedad , Metaloproteinasas de la Matriz/genética , Polimorfismo de Nucleótido Simple , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Oportunidad Relativa , Regiones Promotoras Genéticas , Riesgo
9.
J Virol ; 84(12): 6139-52, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20375168

RESUMEN

We previously reported that the cellular protein ZEB1 can repress expression of the Epstein-Barr virus (EBV) BZLF1 gene in transient transfection assays by directly binding its promoter, Zp. We also reported that EBV containing a 2-bp substitution mutation in the ZEB-binding ZV element of Zp spontaneously reactivated out of latency into lytic replication at a higher frequency than did wild-type EBV. Here, using small interfering RNA (siRNA) and short hairpin RNA (shRNA) technologies, we definitively show that ZEB1 is, indeed, a key player in maintaining EBV latency in some epithelial and B-lymphocytic cell lines. However, in other EBV-positive epithelial and B-cell lines, another zinc finger E-box-binding protein, ZEB2/SIP1, is the key player. Both ZEB1 and ZEB2 can bind Zp via the ZV element. In EBV-positive cells containing only ZEB1, knockdown of ZEB1 led to viral reactivation out of latency, with synthesis of EBV immediate-early and early lytic gene products. However, in EBV-positive cells containing both ZEBs, ZEB2, not ZEB1, was the primary ZEB family member bound to Zp. Knockdown of ZEB2, but not ZEB1, led to EBV lytic reactivation. Thus, we conclude that either ZEB1 or ZEB2 can play a central role in the maintenance of EBV latency, doing so in a cell-type-dependent manner.


Asunto(s)
Infecciones por Virus de Epstein-Barr/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/fisiología , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Latencia del Virus , Replicación Viral , Secuencia de Bases , Línea Celular , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/genética , Proteínas de Homeodominio/genética , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Represoras/genética , Especificidad de la Especie , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Activación Viral , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
10.
J Virol ; 84(9): 4383-94, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181712

RESUMEN

The Epstein-Barr virus (EBV) immediate-early protein BZLF1 (Z) mediates the switch between latent and lytic EBV infection. Z not only activates early lytic viral gene transcription but also plays a direct role in lytic viral genome replication. Although a small fraction of Z is known to be sumoylated, the effects of this posttranslational modification on various different Z functions have not been well defined. In this report, we show that only the lysine at amino acid residue 12 is required for the sumoylation of Z, and that Z can be sumoylated by SUMO isoforms 1, 2, and 3. We also demonstrate that the sumo-defective Z mutants ZK12A and ZK12R have enhanced transcriptional activity. The sumoylated and nonsumoylated forms of Z were found to have a similar cellular location, both being localized primarily within the nuclear matrix. The Z sumo-defective mutants were, however, partially defective for disrupting promyelocytic leukemia (PML) bodies compared to the ability of wild-type Z. In addition, we show that lytic viral genome replication does not require the sumoylation of Z, although a Z mutant altered at both amino acids 12 and 13 is replication defective. Furthermore, we show that the sumoylation of Z is greatly increased (from less than 1 to about 11%) in lytically induced 293 cells infected with an EBV mutant virus deleted for the EBV-encoded protein kinase (EBV-PK) compared to that of 293 cells infected with wild-type EBV, and that the overexpression of EBV-PK leads to the reduced sumoylation of Z in EBV-negative cells. Our results suggest that the sumoylation of Z helps to promote viral latency, and that EBV-PK inhibits Z sumoylation during viral reactivation.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/fisiología , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Transactivadores/metabolismo , Transcripción Genética , Proteínas Virales/metabolismo , Linfocitos B/virología , Línea Celular , Células Epiteliales/virología , Humanos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Activación Viral , Latencia del Virus , Replicación Viral
11.
PLoS Pathog ; 3(12): e194, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18085824

RESUMEN

The immediate-early (IE) BZLF1 gene of Epstein-Barr virus (EBV) regulates the switch between latent and lytic infection by EBV. We previously showed that the cellular transcription factor ZEB1 binds to a sequence element, ZV, located at nt -17 to -12 relative to the transcription initiation site of the BZLF1 promoter, Zp, repressing transcription from Zp in a transient transfection assay. Here, we report the phenotype in the context of a whole EBV genome of a variant of EBV strain B95.8 containing a 2-bp substitution mutation in the ZV element of Zp that reduced, but did not eliminate, ZEB1 binding to Zp. Strikingly, epithelial 293 cells latently infected with the EBV ZV mutant spontaneously produced IE-, early-, and late-gene products and infectious virus, while wild-type (WT)-infected 293 cells did not and have never been reported to do so. Furthermore, treatment with the chemical inducers sodium butyrate and 12-O-tetradecanoyl-phorbol-13-acetate (TPA) led to an additional order-of-magnitude production of infectious virus in the ZV mutant-infected 293 cells, but still no virus in the WT-infected 293 cells. Similarly, ZV mutant-infected Burkitt's lymphoma BJAB cells accumulated at least 10-fold more EBV IE mRNAs than did WT-infected BJAB cells, with TPA or sodium butyrate treatment leading to an additional 5- to 10-fold accumulation of EBV IE mRNAs in the ZV mutant-infected cells. Thus, we conclude that ZEB1 binding to Zp plays a central role in regulating the latent-lytic switch in EBV-infected epithelial and B cells, suggesting ZEB1 as a target for lytic-induction therapies in EBV-associated malignancies.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Proteínas de Homeodominio/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción/metabolismo , Ácido Butírico/farmacología , Proteínas Portadoras , Línea Celular , Línea Celular Transformada , ADN Viral/metabolismo , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Herpesvirus Humano 4/patogenicidad , Proteínas de Homeodominio/genética , Factores de Transcripción de Tipo Kruppel/genética , Mutación , ARN Mensajero/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factores de Transcripción/genética , Transcripción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
12.
J Virol ; 81(18): 10113-22, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17626078

RESUMEN

The induction of lytic infection has been proposed as a therapeutic strategy for treating Epstein-Barr virus (EBV)-positive malignancies. To succeed, efficient methods are needed for activating the EBV immediate-early (IE) promoters, Zp and Rp. Here we compared factors which regulate Zp and Rp in AGS gastric carcinoma cells that support a remarkably high level of persistently lytic EBV infection with HeLa cervical cells that permit only tightly latent infection. We found that the level of Zp activity assayed by transient transfection assays with reporter plasmids was high in AGS cells but low in HeLa cells. The level of Rp activity was low in both cell types. Mutational analysis indicated that sequences within Zp located between -70 and +27 relative to the transcription initiation site were sufficient to confer a high level of Zp activity in AGS cells. The Zp CRE motif was necessary for this constitutive activity, while the ZIA and ZIB MEF2D motifs were not. Consistent with these findings, immunoblot analysis indicated that phosphorylated c-Jun, which activates Zp through the CRE motif, was expressed at a much higher level in EBV-infected AGS cells than in EBV-infected HeLa cells. In contrast, ZEB1, which represses Zp via the ZV motif located near the transcription initiation site, was abundant in HeLa cells, while it was absent from AGS cells. Exogenous addition of ZEB1 led to the repression of Zp in AGS cells. We conclude that the unusually high Zp activity level in AGS cells is due to the high abundance of positively acting transcription factors such as c-Jun combined with the low abundance of negatively acting factors such as ZEB1.


Asunto(s)
Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas de Homeodominio/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Represoras/metabolismo , Neoplasias Gástricas/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Infecciones por Virus de Epstein-Barr/genética , Células HeLa , Herpesvirus Humano 4/genética , Proteínas de Homeodominio/genética , Humanos , Proteínas de Dominio MADS/genética , Proteínas de Dominio MADS/metabolismo , Factores de Transcripción MEF2 , Mutación , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Represoras/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/terapia , Neoplasias Gástricas/virología , Factores de Transcripción/genética , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
13.
J Virol ; 77(4): 2489-99, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12551987

RESUMEN

To study the effects of the nuclear receptors (NRs) HNF4alpha and COUP-TF1 on the life cycle of hepatitis B virus (HBV), the human hepatoma cell line Huh7 was transiently cotransfected with plasmids containing the HBV genome and encoding these two NRs. Overexpression of HNF4alpha and COUP-TF1 led to a 9-fold increase and a 7- to 10-fold decrease, respectively, in viral DNA synthesis. These two NRs also exhibited distinct modes of regulation of viral transcription. Overexpression of HNF4alpha led to a more-than-10-fold increase in synthesis of the pregenomic RNA but to only a 2- to 3-fold increase in synthesis of the pre-C and S RNAs. Moreover, the NR response element within the pre-C promoter, NRRE(preC,) played the major role in activation of pregenomic RNA synthesis by HNF4alpha. On the other hand, overexpression of COUP-TF1 led to an over-10-fold repression of synthesis of both pre-C and pregenomic RNAs mediated through either NRRE(preC) or NRRE(enhI). HNF4alpha and COUP-TF1 antagonized each other's effects on synthesis of pregenomic RNA and viral DNA when they were co-overexpressed. A naturally occurring HBV variant which allows for binding by HNF4alpha but not COUP-TF1 in its NRRE(preC) exhibited significantly higher levels of synthesis of pregenomic RNA and viral DNA than wild-type HBV in coexpression experiments. Last, deletion analysis revealed that non-NRRE sequences located within both the C and pre-S1 regions are also essential for maximum activation of the pregenomic promoter by HNF4alpha but not for repression by COUP-TF1. Thus, HNF4alpha and COUP-TF1 function through different mechanisms to regulate expression of the HBV genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Factor de Transcripción COUP I , ADN Viral/biosíntesis , Proteínas de Unión al ADN/genética , Virus de la Hepatitis B/genética , Factor Nuclear 4 del Hepatocito , Humanos , Mutación , Fosfoproteínas/genética , ARN Viral/biosíntesis , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Elementos de Respuesta/genética , Factores de Transcripción/genética , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA