Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
mSphere ; 3(1)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29435490

RESUMO

Increasing evidence shows that exosomes are key regulators in cancer cell-to-cell communication. Several reports on Epstein-Barr virus (EBV)-related malignancies demonstrate that latent membrane protein 1 (LMP1) secreted by exosomes derived from EBV- or LMP1-positive cells can promote cancer progression and metastasis. However, the mechanism by which LMP1 is loaded into exosomes is still poorly understood. Here, we examined whether the process of LMP1 loading into exosomes is linked to the multifunctional molecule of the ubiquitin system-ubiquitin C-terminal hydrolase-L1 (UCH-L1). For the first time, we demonstrate that LMP1 is physically associated with UCH-L1 and that directing of LMP1 to exosomes is mediated by C-terminal farnesylation of UCH-L1. Additionally, we found that the FTI-277 farnesyltransferase inhibitor reduces motility- and anchorage-independent growth of EBV-positive cells in functional assays. On the basis of our results, we conclude that C-terminal farnesylation of UCH-L1 is one of the key mechanisms by which LMP1 is sorted to exosomes. We hypothesize that inhibition of farnesylation with specific small-molecule inhibitors blocks exosome-mediated transfer of prometastatic molecules such as LMP1 during cancer cell-to-cell communications and thereby impedes the process of cancer invasion. IMPORTANCE Exosomes are small vesicles that cells secrete into the extracellular space, and there is increasing evidence that they have pivotal roles in cell-to-cell communication in malignancy. It is reported also that EBV-associated malignant cells, including those derived from nasopharyngeal carcinoma (NPC) and B-cell lymphoma, secrete exosomes. These EBV-related exosomes may contain viral products such as latent membrane protein 1 (LMP1) and may contribute to cancer progression. The aim of this study was to investigate the mechanism by which those viral products are loaded in exosomes. In this study, we show for the first time that ubiquitin C-terminal hydrolase-L1 (UCH-L1) and its C-terminal farnesylation, a posttranslational lipid modification, contribute to this mechanism. Our results also suggest that inhibition of UCH-L1 farnesylation is a potential therapeutic target against cancer metastasis and invasion.

2.
Oncogene ; 33(37): 4613-22, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-24662828

RESUMO

It has emerged recently that exosomes are potential carriers of pro-tumorigenic factors that participate in oncogenesis. However, whether oncogenic transcription factors are transduced by exosomes is unknown. Hypoxia-inducible factor-1α (HIF1α) transcriptionally regulates numerous key aspects of tumor development and progression by promoting a more aggressive tumor phenotype, characterized by increased proliferation and invasiveness coupled with neoangiogenesis. It has been shown that the principal oncoprotein of Epstein-Barr virus (EBV), latent membrane protein 1 (LMP1), drives oncogenic processes and tumor progression of the highly invasive EBV malignancy, nasopharyngeal carcinoma (NPC). We now demonstrate that endogenous HIF1α is detectable in exosomes and that LMP1 significantly increases levels of HIF1α in exosomes. HIF1 recovered from exosomes retains DNA-binding activity and is transcriptionally active in recipient cells after exosome uptake. We also show that treatment of EBV-negative cells with LMP1-exosomes increases migration and invasiveness of NP cell lines in functional assays, which correlates with the phenotype associated with epithelial-mesenchymal transition (EMT). In addition, we provide evidence that HIF1α itself participates in exosome-mediated pro-metastatic effects in recipient cells, as exosome-mediated delivery of active and inactive forms of HIF1α results in reciprocal changes in the expression of E- and N-cadherins associated with EMT. Further, immunohistochemical analysis of NPC tumor tissues revealed direct correlation between protein levels of LMP1 and of the endosome/exosome marker tetraspanin, CD63, which suggests an increase in exosome formation in this EBV-positive malignancy. We hypothesize that exosome-mediated transfer of functional pro-metastatic factors by LMP1-positive NPC cells to surrounding tumor cells promotes cancer progression.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteínas da Matriz Viral/metabolismo , Carcinoma , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica , DNA/química , Transição Epitelial-Mesenquimal , Exossomos/metabolismo , Células HEK293 , Herpesvirus Humano 4/metabolismo , Humanos , Carcinoma Nasofaríngeo , Invasividade Neoplásica , Metástase Neoplásica , Fenótipo , Ligação Proteica , Tetraspanina 30/metabolismo , Cicatrização
3.
Genes Immun ; 12(6): 399-414, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21490621

RESUMO

Interferon regulatory factor 7 (IRF7) was originally identified in the context of Epstein-Barr virus (EBV) infection, and has since emerged as the crucial regulator of type I interferons (IFNs) against pathogenic infections, which activate IRF7 by triggering signaling cascades from pathogen recognition receptors (PRRs) that recognize pathogenic nucleic acids. Moreover, IRF7 is a multifunctional transcription factor, underscored by the fact that it is associated with EBV latency, in which IRF7 is induced as well as activated by the EBV principal oncoprotein latent membrane protein-1 (LMP1). Aberrant production of type I IFNs is associated with many types of diseases such as cancers and autoimmune disorders. Thus, tight regulation of IRF7 expression and activity is imperative in dictating appropriate type I IFN production for normal IFN-mediated physiological functions. Posttranslational modifications have important roles in regulation of IRF7 activity, exemplified by phosphorylation, which is indicative of its activation. Furthermore, mounting evidence has shed light on the importance of regulatory ubiquitination in activation of IRF7. Albeit these exciting findings have been made in the past decade since its discovery, many questions related to IRF7 remain to be addressed.


Assuntos
Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiologia , Humanos , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Latência Viral
4.
Br J Cancer ; 104(7): 1160-7, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21386845

RESUMO

BACKGROUND: Epstein-Barr Virus (EBV)-associated nasopharyngeal carcinoma (NPC) is distinctive among head-and-neck cancers in its undifferentiated histopathology and highly metastatic character. We have recently investigated the involvement of epithelial-mesenchymal transition (EMT) in NPC. In a previous study, we found a close association of expression of LMP1, the principal EBV oncoprotein, with expression of Twist and induction of EMT. METHODS: We analysed expression of Snail in 41 NPC tissues by immunohistochemistry. The role of Twist as well as Snail in EMT of NPC was investigated by using NP69SV40T human nasopharyngeal cells. RESULTS: In NPC tissues, overexpression of Snail is associated with expression of LMP1 in carcinomatous cells. In addition, expression of Snail positively correlated with metastasis and independently correlated inversely with expression of E-cadherin. Expression of Twist had no association with expression of E-cadherin. Further, in a human nasopharyngeal cell line, LMP1 induces EMT and its associated cellular motility and invasiveness. Expression of Snail is induced by LMP1 in these cells, and small hairpin RNA (shRNA) to Snail reversed the cellular changes. By contrast, Twist did not produce EMT in these nasopharyngeal cells. CONCLUSIONS: This study strengthens the association of EMT with the metastatic behaviour of NPC. These results suggest that induction of Snail by the EBV oncoprotein LMP1 has a pivotal role in EMT in NPC.


Assuntos
Transição Epitelial-Mesenquimal , Herpesvirus Humano 4/fisiologia , Fatores de Transcrição/fisiologia , Proteínas da Matriz Viral/fisiologia , Caderinas/análise , Carcinoma , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/etiologia , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/virologia , Metástase Neoplásica , Proteínas do Tecido Nervoso/análise , Proteínas de Ligação a RNA/análise , Fatores de Transcrição da Família Snail , Fatores de Transcrição/análise , Proteínas da Matriz Viral/análise
5.
Oncogene ; 28(14): 1725-35, 2009 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-19234486

RESUMO

Tumor metastasis is a complex phenomenon that is the culmination of effects of numerous cellular factors. We have shown that the Epstein-Barr virus (EBV) oncoprotein, latent membrane protein 1 (LMP1), is capable of inducing a wide range of such factors in cell culture, expression of which is also elevated in the LMP1-expressing tumor, nasopharyngeal carcinoma (NPC), a highly invasive neoplasm. Recently, the membrane crosslinker protein, ezrin, has been implicated in tumor cell metastasis and malignant progression. In this study, we evaluated the possible role of LMP1 and ezrin in the pathophysiology of NPC. We show that C-terminal phosphorylation of ezrin is increased by the expression of LMP1 in nasopharyngeal (NP) cells through a protein kinase C (PKC) pathway. LMP1 enhances the organization of a ternary complex of CD44, ezrin and F-actin, which is a prerequisite for ezrin phosphorylation. In NPC tissues, the expression of phosphoezrin and LMP1 is directly correlated. Silencing of endogenously expressed ezrin suppresses LMP1-induced cell motility and invasiveness. Moreover, the inhibition of ezrin phosphorylation by PKC inhibitor suppresses migration and invasion of NP cells. These data show that the phosphorylation of ezrin and its recruitment to the cell membrane linked to F-actin and CD44 is a process required for LMP1-stimulated cell motility and invasion of NP cells.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Neoplasias Nasofaríngeas/patologia , Proteínas da Matriz Viral/fisiologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Citosol/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Receptores de Hialuronatos/análise , Invasividade Neoplásica , Fosforilação , Proteína Quinase C/fisiologia , Transporte Proteico , Fatores de Transcrição/metabolismo , Quinases Associadas a rho/fisiologia
6.
J Virol ; 78(22): 12140-6, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15507600

RESUMO

The protein kinase (PK) encoded by the Epstein-Barr Virus (EBV) BGLF4 gene is the only EBV protein kinase. The expression pattern of EBV PK during the reactivation of the viral lytic cycle and the subcellular localization of the protein were analyzed with a polyclonal antiserum raised against a peptide corresponding to the N terminus of EBV PK. Based on previously published data (E. Gershburg and J. S. Pagano, J. Virol. 76:998-1003, 2002) and the expression pattern described here, we conclude that EBV PK is an early protein that requires viral-DNA replication for maximum expression. By biochemical fractionation, the protein could be detected mainly in the nuclear fraction 4 h after viral reactivation in Akata cells. Nuclear localization could be visualized by indirect immunofluorescence in HeLa cells transiently expressing EBV BGLF4 in the absence of other viral products. Transient expression of 3'-terminal deletion mutants of EBV BGLF4 resulted in cytoplasmic localization, confirming the presence of a nuclear localization site in the C-terminal region of the protein. In contrast to the wild-type EBV PK, all of the mutants were unable to hyperphosphorylate EA-D during coexpression or to phosphorylate ganciclovir, as measured by an in-cell activity assay. Thus, the results demonstrate that the nuclear localization, as well as the kinase activity, of BGFL4 is dependent on an intact C-terminal region.


Assuntos
Herpesvirus Humano 4/enzimologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Virais/genética , Transporte Ativo do Núcleo Celular , Linhagem Celular Tumoral , Herpesvirus Humano 4/genética , Humanos , Sinais de Localização Nuclear , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Ativação Viral
7.
Cancer Res ; 61(21): 7875-7, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11691806

RESUMO

Nasopharyngeal carcinoma (NPC) is universally associated with EBV infection. We have shown that the phosphonated nucleoside analog, (S)-1-[3-hydroxy-2-(phosphonylmethoxy)-propyl]cytosine (HPMPC) strongly inhibits growth of NPC xenografts in nude mice by causing apoptosis (J. Neyts et al., Cancer Res., 58, 384-388, 1998). We, therefore, tested two additional members of this drug family that have different degrees of antiviral activity, 9-[2-(phosphonomethoxy)ethyl]adenine (PMEA) and 9-2-(R)-(phosphonomethoxy)propyladenine (PMPA). Intratumoral injection of PMEA (75 microl of 2% solution) in C15 NPC xenografts, which are latently infected with EBV, slowed tumor growth moderately, whereas PMPA (75 microl of 2% solution) slowed tumor growth only marginally. Compared with the previous results showing complete regression of tumor, PMEA had less antitumoral effect than HPMPC, and PMPA had the least. After 4 weeks of preventive treatment, tumors formed in 12.5, 50, and 100% of mice treated with HPMPC, PMEA, and PMPA, respectively, in contrast to the development of tumors in all of the PBS-treated control mice. We also investigated the effect of each drug on the EBV-positive epithelial cell line NPC-KT in vitro. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed inhibition of growth of NPC-KT cells by HPMPC and PMEA, but not by PMPA, which correlates with the results observed in tumor xenografts. Growth inhibition was attributable to induction of apoptosis in NPC-KT cells as indicated by a DNA fragmentation assay. Cleavage of poly(ADP-ribose) polymerase after treatment of NPC-KT cells with HPMPC was observed, which suggested that the apoptosis may be mediated by caspase(s). The apoptotic effects of the drugs are independent of any effects on EBV DNA polymerase, which is not expressed in these latently infected NPCs. These results suggest that HPMPC as well as PMEA could provide an adjunctive treatment for NPC.


Assuntos
Adenina/análogos & derivados , Adenina/farmacologia , Antineoplásicos/farmacologia , Carcinoma/tratamento farmacológico , Citosina/análogos & derivados , Neoplasias Nasofaríngeas/tratamento farmacológico , Organofosfonatos , Compostos Organofosforados/farmacologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma/patologia , Carcinoma/prevenção & controle , Divisão Celular/efeitos dos fármacos , Cidofovir , Citosina/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Inibidores do Crescimento/farmacologia , Herpesvirus Humano 4 , Humanos , Camundongos , Camundongos Nus , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/prevenção & controle , Tenofovir , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Virol ; 75(24): 12393-401, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11711629

RESUMO

Epstein-Barr virus (EBV) latent membrane protein 1 (LMP-1) is the principal oncogenic protein in the EBV transformation process. LMP-1 induces the expression of interferon regulatory factor 7 (IRF-7) and activates IRF-7 protein by phosphorylation and nuclear translocation. LMP-1 is an integral membrane protein with two regions in its C terminus that initiate signaling processes, the C-terminal activator regions 1 (CTAR-1) and CTAR-2. Here, genetic analysis of LMP-1 has determined that the PXQXT motif that governs the interaction between LMP-1 CTAR-1 and tumor necrosis factor receptor-associated factors (TRAFs) is needed to induce the expression of IRF-7. Mutations in the PXQXT motif in CTAR-1 that disrupt the interaction between LMP-1 and TRAFs abolished the induction of IRF-7. Also, dominant-negative mutants of TRAFs inhibited the induction of IRF-7 by CTAR-1. The last three amino acids (YYD) of CTAR-2 are also important for the induction of IRF-7. When both PXQXT and YYD were mutated (LMP-DM), the LMP-1 mutant failed to induce IRF-7. Also, LMP-DM blocked the induction of IRF-7 by wild-type LMP-1. These data strongly suggest that both CTAR-1 and CTAR-2 of LMP-1 independently induce the expression of IRF-7. In addition, NF-kappaB is involved in the induction of IRF-7. A superrepressor of IkappaB (sr-IkappaB) could block the induction of IRF-7 by LMP-1, and overexpression of NF-kappaB (p65 plus p50) could induce the expression of IRF-7. In addition, we have found that human IRF-7 is a stable protein, and sodium butyrate, a modifier of chromatin structure, induces IRF-7.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Herpesvirus Humano 4/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Linhagem Celular , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/química , Meia-Vida , Humanos , Fator Regulador 7 de Interferon , Peptídeos e Proteínas de Sinalização Intracelular , Janus Quinase 3 , Proteínas com Domínio LIM , NF-kappa B/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas/fisiologia , Fator 1 Associado a Receptor de TNF , Fator 2 Associado a Receptor de TNF
9.
Semin Cancer Biol ; 11(6): 445-53, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11669606

RESUMO

Interferon regulatory factor 7 (IRF-7) was cloned within the biological context of Epstein-Barr virus (EBV) latency, and has an intimate relation with EBV. EBV latent membrane protein 1 (LMP-1) regulates IRF-7 both by inducing the expression of IRF-7 and by activating IRF-7 protein through phosphorylation and nuclear translocation in a post-translational manner. The activated IRF-7 then functions to regulate both EBV and cellular target genes involved in latency, transformation and immune regulation. IRF-7 appears to be a key cellular latency protein involved in both the pathogenesis and persistence of EBV infection.


Assuntos
Transformação Celular Viral/fisiologia , Proteínas de Ligação a DNA/metabolismo , Herpesvirus Humano 4/fisiologia , Proteínas da Matriz Viral/metabolismo , Latência Viral/fisiologia , Transformação Celular Viral/imunologia , Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/metabolismo , Humanos , Fator Regulador 7 de Interferon , Modelos Biológicos , Oncogenes/fisiologia , Regiões Promotoras Genéticas/genética , Processamento de Proteína Pós-Traducional , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética
10.
Proc Natl Acad Sci U S A ; 98(12): 6905-10, 2001 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-11381123

RESUMO

Cyclooxygenase-2 (COX-2) is an inducible form of COX and is overexpressed in diverse tumors, raising the possibility of a role for COX-2 in carcinogenesis. In addition, COX-2 contributes to angiogenesis. The Epstein-Barr virus (EBV) oncoprotein, latent membrane protein 1 (LMP1), is detected in at least 70% of nasopharyngeal carcinoma (NPC) and all EBV-infected preinvasive nasopharyngeal lesions. We found that in specimens of LMP1-positive NPC, COX-2 is frequently expressed, whereas LMP1-negative NPC rarely express the enzyme. We next found that expression of LMP1 in EBV-negative nasopharyngeal epithelial cells induced COX-2 expression. Coexpression of IkappaBalpha(S32A/S36A), which is not phosphorylated and prevents NF-kappaB activation, with LMP1 showed that NF-kappaB is essential for induction of COX-2 by LMP1. We also demonstrate that NF-kappaB is involved in LMP1-induced cox-2 promoter activity with the use of reporter assays. Two major regions of LMP1, designated CTAR1 and CTAR2, are signal-transducing domains of LMP1. Constructs expressing either CTAR1 or CTAR2 induce COX-2 but to a lesser extent than wild-type LMP1, consistent with the ability of both regions to activate NF-kappaB. Furthermore, we demonstrate that LMP1-induced COX-2 is functional because LMP1 increased production of prostaglandin E(2) in a COX-2-dependent manner. Finally, we demonstrate that LMP1 increased production of vascular endothelial growth factor (VEGF). Treatment of LMP1-expressing cells with the COX-2-specific inhibitor (NS-398) dramatically decreased production of VEGF, suggesting that LMP1-induced VEGF production is mediated, at least in part, by COX-2. These results suggest that COX-2 induction by LMP1 may play a role in angiogenesis in NPC.


Assuntos
Fatores de Crescimento Endotelial/biossíntese , Isoenzimas/biossíntese , Linfocinas/biossíntese , Neoplasias Nasofaríngeas/irrigação sanguínea , Neovascularização Patológica/etiologia , Prostaglandina-Endoperóxido Sintases/biossíntese , Proteínas da Matriz Viral/fisiologia , Ciclo-Oxigenase 2 , Dinoprostona/biossíntese , Indução Enzimática , Humanos , Isoenzimas/genética , Metaloproteinase 9 da Matriz/biossíntese , Proteínas de Membrana , NF-kappa B/fisiologia , Neoplasias Nasofaríngeas/enzimologia , Neoplasias Nasofaríngeas/virologia , Regiões Promotoras Genéticas , Prostaglandina-Endoperóxido Sintases/genética , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
11.
J Virol ; 75(1): 341-50, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11119603

RESUMO

Transporter associated with antigen processing 2 (Tap-2) is responsible for ATP-dependent transport of peptides from the cytosol to the endoplasmic reticulum, where peptides bind to newly synthesized human leukocyte antigen (HLA) class I molecules, which are essential for cellular immune responses. Epstein-Barr virus (EBV) latent membrane protein 1 (LMP-1) has been shown to induce the expression of Tap-2. In this study, the induction of endogenous Tap-2 by LMP-1 is shown to be associated with and requires the expression of interferon regulatory factor 7 (IRF-7). In DG75 Burkitt's lymphoma (BL) cells, in which LMP-1 induces the expression of IRF-7, LMP-1 induced endogenous Tap-2, and ectopic expression of IRF-7 could enhance the induction. In Akata BL cells, in which LMP-1 could not induce IRF-7, LMP-1 could not induce Tap-2. Addition of IRF-7, which complements the defect in Akata cells, could stimulate the expression of Tap-2. Furthermore, LMP-1 and IRF-7A but not other IRF-7 splicing variants could activate endogenous Tap-2. A Tap-2 promoter reporter construct could be activated by the overexpression of IRF-7A. The activation could be specifically enhanced by LMP-1 and was dependent on an intact interferon-stimulated response element (ISRE) present in the Tap-2 promoter. Also, IRF-7 can bind to the Tap-2 promoter under physiological conditions in vivo, as shown by formaldehyde cross-linking, as well as to the Tap-2 ISRE in vitro, as shown by gel mobility shift assays. Furthermore, LMP-1 facilitates the phosphorylation and nuclear translocation of IRF-7. These data point to the role of IRF-7 as a secondary mediator of LMP-1-activated signal transduction for Tap-2 as follows: LMP-1 stimulates the expression of IRF-7 and facilitates its phosphorylation and nuclear translocation, and then the activated IRF-7 mediates the activation of the cellular Tap-2 gene. The induction of Tap-2 by IRF-7 and LMP-1 may have an important implication for the immune response to EBV and its persistence in vivo.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Proteínas de Transporte/fisiologia , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/análise , Núcleo Celular/metabolismo , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/análise , Humanos , Fator Regulador 7 de Interferon , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas com Domínio LIM , Fosforilação , Regiões Promotoras Genéticas
12.
Cancer Res ; 60(9): 2555-61, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10811139

RESUMO

Matrix metalloproteinases (MMPs) are thought to play crucial roles in tumor invasion and metastasis. Because we have shown that EBV latent membrane protein 1 (LMP1) enhances MMP-9 expression by activation of nuclear factor (NF)-kappaB and activator protein (AP)-1 (T. Yoshizaki, et al., Proc. Natl. Acad. Sci. USA, 95: 3621-3626, 1998), we therefore tested whether up-regulation of MMP-9 by LMP1 could be correlated with enhanced invasiveness of tumor cells in vitro. Whether aspirin and sodium salicylate could reduce invasiveness and whether LMP1 could enhance MMP-9 expression in tumors grown in nude mice were also tested. C33A cells stably expressing LMP1 had increased expression of MMP-9 and showed greater invasion through reconstituted basement membrane compared with vector-transfected C33A cells (P < 0.02). Treatment with aspirin or sodium salicylate inhibited invasiveness of the LMP1-expressing C33A cells (P < 0.03) and suppressed both the LMP1-induced MMP-9 expression in zymographic analyses and LMP1-induced MMP-9 promoter activity in CAT reporter assays (P < 0.01). Endogenous MMP-2 levels were unaffected by either drug. Both drugs repressed the CAT activity of the truncated MMP-9 promoter construct, which only contained a binding site for AP-1, to the basal level (P < 0.05). Moreover, EMSA indicated that the effects of the salicylates were through the inhibition of not only NF-kappaB but also AP-1 binding activity. Inhibitory effect of salicylates could be reversed by p50/p65 subunits of NF-kappaB or c-Jun overexpression. The inhibitory effect of aspirin on NF-kappaB activity was attributable to the inhibition of IkappaB kinase activity. Finally, tumors derived from C33A cells stably expressing LMP1 grown in nude mice showed enhanced MMP-9 levels compared with tumors derived from vector-transfected C33A cells. This enhancement was inhibited by treatment of the mice with aspirin. These results suggest that aspirin may be able to suppress invasion and metastasis of EBV-associated tumors that express LMP1 by suppression of MMP-9.


Assuntos
Aspirina/farmacologia , Inibidores de Metaloproteinases de Matriz , Invasividade Neoplásica , Proteínas da Matriz Viral/farmacologia , Animais , Western Blotting , Núcleo Celular/metabolismo , Cloranfenicol O-Acetiltransferase/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Quinase I-kappa B , Proteínas I-kappa B/metabolismo , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Nus , NF-kappa B/antagonistas & inibidores , Metástase Neoplásica , Plasmídeos , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Salicilato de Sódio/farmacologia , Fator de Transcrição AP-1/antagonistas & inibidores , Transfecção , Células Tumorais Cultivadas
13.
J Virol ; 74(3): 1061-8, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10627515

RESUMO

Infection by Epstein-Barr virus (EBV) generates several types of latency with different profiles of gene expression but with expression of Epstein-Barr nuclear antigen 1 (EBNA-1) in common. The BamHI Q promoter (Qp) is used for the transcription of EBNA-1 mRNA in type I latency, which is an EBV infection state exemplified by Burkitt's lymphoma (BL). However, Qp is inactive in type III latency, and other promoters (C/Wp) are used for transcription of EBNA-1, which raises the question of how usage of these promoters is governed. Interferon (IFN) regulatory factor 7 (IRF-7) was identified first as a negative regulator of Qp. Expression of IRF-7 is associated with EBV type III latency, where Qp is inactive, but not with type I latency, raising the possibility that a viral gene product(s) expressed in type III latency might induce IRF-7 and repress Qp. Here, detailed analysis of the expression of IRF-7 revealed that it is associated with the expression of EBV latent membrane protein 1 (LMP-1) and that LMP-1 stimulates the expression of IRF-7 in type III latency in which Qp is inactive. In contrast, LMP-1 is not expressed in type I latency cells in which Qp is active. LMP-1 represses the constitutive activity of Qp reporter constructs. Mutational analysis of Qp reporter constructs revealed that the Qp IFN-stimulated response element (ISRE) is essential for the repression by LMP-1. Furthermore, LMP-1 reduced EBNA-1 mRNA derived from Qp only in type I cells in which IRF-7 could be induced. Finally, IFN-alpha, but not IFN-gamma, repressed endogenous Qp activity, which is consistent with the ability of IFN-alpha to induce IRF-7. Thus, IRF-7 may mediate repression of Qp by LMP-1. The induction of IRF-7 by LMP-1 may be relevant to the silencing of Qp in EBV type III latency.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/fisiologia , Proteínas da Matriz Viral/metabolismo , Latência Viral/genética , Western Blotting , Proteínas de Ligação a DNA/genética , Antígenos Nucleares do Vírus Epstein-Barr/genética , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Genes Reporter , Herpesvirus Humano 4/genética , Humanos , Fator Regulador 7 de Interferon , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Proteínas da Matriz Viral/genética
14.
Proc Assoc Am Physicians ; 111(6): 573-80, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10591086

RESUMO

The Epstein-Barr virus (EBV) is classically associated with three malignancies, Burkitt's lymphoma. B-cell lymphoproliferative syndromes, and nasopharyngeal carcinoma, and, more recently, with Hodgkin's disease, T-cell lymphomas, and gastric carcinoma, as well as being the causal agent for infectious mononucleosis. The relation of the virus to the malignancies varies from primary etiologic agent to necessary or contributory cofactor. Clonal EBV episomes are found in all the malignant conditions. EBV infects both epithelial and lymphoid cells, providing a pathobiological basis for these diverse associations. Most of the malignancies occur after years of viral dormancy and are accompanied or triggered by viral reactivation, in contrast to infectious mononucleosis, which results from primary infection with EBV. The EBV-associated malignancies offer insights into the causation and early detection of cancer. The molecular virology and pathobiology of EBV infection states provide the basis for the specific diagnosis of these diseases and a framework for new therapies.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4/patogenicidade , Neoplasias/virologia , Adolescente , Adulto , Antivirais/uso terapêutico , Linfoma de Burkitt/epidemiologia , Linfoma de Burkitt/etiologia , Linfoma de Burkitt/virologia , Carcinoma/epidemiologia , Carcinoma/etiologia , Carcinoma/virologia , Transformação Celular Neoplásica , Transformação Celular Viral , Criança , Pré-Escolar , Cocarcinogênese , Células Epiteliais/virologia , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Infecções por Vírus Epstein-Barr/epidemiologia , Infecções por Vírus Epstein-Barr/imunologia , Feminino , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/fisiologia , Humanos , Terapia de Imunossupressão/efeitos adversos , Lactente , Recém-Nascido , Mononucleose Infecciosa/virologia , Leucoplasia Oral/etiologia , Leucoplasia Oral/virologia , Linfócitos/virologia , Linfoma de Células B/epidemiologia , Linfoma de Células B/etiologia , Linfoma de Células B/virologia , Transtornos Linfoproliferativos/epidemiologia , Transtornos Linfoproliferativos/etiologia , Transtornos Linfoproliferativos/virologia , Masculino , Pessoa de Meia-Idade , Neoplasias Nasofaríngeas/epidemiologia , Neoplasias Nasofaríngeas/etiologia , Neoplasias Nasofaríngeas/virologia , Neoplasias/epidemiologia , Neoplasias/etiologia , Neoplasias/imunologia , Neoplasias/prevenção & controle , Lesões Pré-Cancerosas/etiologia , Lesões Pré-Cancerosas/terapia , Lesões Pré-Cancerosas/virologia
15.
J Virol ; 73(9): 7271-7, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10438815

RESUMO

Although a number of antiviral drugs inhibit replication of Epstein-Barr virus (EBV) in cell culture, and acyclovir (ACV) suppresses replication in vivo, currently available drugs have not proven effective for treatment of EBV-associated diseases other than oral hairy leukoplakia. Benzimidazole riboside compounds represent a new class of antiviral compounds that are potent inhibitors of human cytomegalovirus (HCMV) replication but not of other herpesviruses. Here we characterize the effects of two compounds in this class against lytic replication of EBV induced in a Burkitt lymphoma cell line latently infected with EBV. We analyzed linear forms of EBV genomes, indicative of lytic replication, and episomal forms present in latently infected cells by terminal probe analysis followed by Southern blot hybridization as well as the high-molecular-weight unprocessed viral DNA by pulsed-field gel electrophoresis. D-Ribofuranosyl benzimidazole compounds that act as inhibitors of HCMV DNA maturation, including BDCRB (5, 6-dichloro-2-bromo-1-beta-D-ribofuranosyl-1H-benzimidazole), did not affect the accumulation of high-molecular-weight or monomeric forms of EBV DNA in the induced cells. In contrast, the generation of linear EBV DNA as well as precursor viral DNA was sensitive to the L-riboside 1263W94 [5, 6-dichloro-2-(isopropylamino)-1-beta-L-ribofuranosyl-1H-benzimidazole]. The 50% inhibitory concentration range for 1263W94 was 0.15 to 1. 1 microM, compared with 10 microM for ACV. Thus, 1263W94 is a potent inhibitor of EBV. In addition, 1263W94 inhibited the phosphorylation and the accumulation of the essential EBV replicative cofactor, early antigen D.


Assuntos
Antivirais/farmacologia , Benzimidazóis/farmacologia , Diclororribofuranosilbenzimidazol/farmacologia , Herpesvirus Humano 4/efeitos dos fármacos , Ribonucleosídeos/farmacologia , Replicação Viral/efeitos dos fármacos , Aciclovir/farmacologia , Antígenos Virais/genética , Antígenos Virais/metabolismo , Linhagem Celular , Diclororribofuranosilbenzimidazol/química , Herpesvirus Humano 4/fisiologia , Humanos , Fosforilação/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
16.
J Virol ; 73(7): 5548-55, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10364303

RESUMO

Nasopharyngeal carcinoma (NPC), which is closely associated with the Epstein-Barr virus (EBV), is a highly metastatic malignant tumor. An important activity in tumor invasion and metastasis is that of the 92-kDa type IV collagenase or gelatinase, matrix metalloproteinase 9 (MMP-9), which mediates the degradation of the basement membrane and extracellular matrix. The expression of MMP-9 has been shown to be enhanced by the EBV oncoprotein, latent membrane protein 1 (LMP-1). LMP-1, which is expressed in NPC, has two essential signaling domains within the carboxy terminus, termed C-terminal activation regions 1 (CTAR-1) and CTAR-2. This study reveals that either signaling domain can activate the MMP-9 promoter and induce MMP-9 activity; however, LMP-1 deletion mutants lacking either CTAR-1 or CTAR-2 had a decreased ability to induce MMP-9 expression. The deletion of both activation regions completely abolished the induction of MMP-9 activity, while the cotransfection of both the CTAR-1 and CTAR-2 deletion mutants restored MMP-9 activity to levels produced by wild-type LMP-1. The NF-kappaB and activator protein 1 (AP-1) binding sites in the MMP-9 promoter were essential for the activation of MMP-9 gene expression by both CTAR-1 and CTAR-2. The induction of MMP-9 expression by LMP-1 and both CTAR-1 and CTAR-2 mutants was blocked by the overexpression of IkappaB. The tumor necrosis factor receptor-associated factor (TRAF) pathway also contributed to the activation of the MMP-9 promoter as shown by the use of TRAF-2 and TRAF-3 dominant-negative constructs. These data indicate that the activation of both the NF-kappaB and AP-1 pathways by LMP-1, CTAR-1, and CTAR-2 is necessary for the activation of MMP-9 expression. In NPC, LMP-1 may contribute to invasiveness and metastasis through the induction of MMP-9 transcription and enzymatic activity.


Assuntos
Colagenases/genética , Regulação Enzimológica da Expressão Gênica , Herpesvirus Humano 4/metabolismo , Proteínas da Matriz Viral/metabolismo , Sítios de Ligação , Colagenases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Viral da Expressão Gênica , Humanos , Proteínas I-kappa B , Metaloproteinase 9 da Matriz , Mutagênese , NF-kappa B/metabolismo , Peptídeos/genética , Regiões Promotoras Genéticas , Proteínas/metabolismo , Transdução de Sinais , Fator 2 Associado a Receptor de TNF , Fator 3 Associado a Receptor de TNF , Fator de Transcrição AP-1/metabolismo , Células Tumorais Cultivadas , Proteínas da Matriz Viral/genética
17.
J Virol ; 73(4): 3154-61, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10074167

RESUMO

Expression of EBNA-1 protein is required for the establishment and maintenance of the Epstein-Barr virus (EBV) genome during latent infection. During type I latency, the BamHI Q promoter (Qp) gives rise to EBNA-1 expression. The dominant regulatory mechanism for Qp appears to be mediated through the Q locus, located immediately downstream of the transcription start site. Binding of EBNA-1 to the Q locus represses Qp constitutive activity, and repression has been reported to be overcome by an E2F family member that binds to the Q locus and displaces EBNA-1 (N. S. Sung, J. Wilson, M. Davenport, N. D. Sista, and J. S. Pagano, Mol. Cell. Biol. 14:7144-7152, 1994). These data suggest that the final outcome of Qp activity is reciprocally controlled by EBNA-1 and E2F. Since E2F activity is cell cycle regulated, Qp activity and EBNA-1 expression are predicted to be regulated in a cell cycle-dependent manner. Proliferation of the type I latently infected cell line, Akata, was synchronized with the use of the G2/M blocking agent nocodazole. From 65 to 75% of cells could be made to peak in S phase without evidence of viral reactivation. Following release from G2/M block, EBNA-1 mRNA levels declined as the synchronized cells entered the G1 phase of the cell cycle. As cells proceeded into S phase, EBNA-1 mRNA levels increased parallel to the peak in cell numbers in S phase. However, EBNA-1 protein levels showed no detectable change during the cell cycle, most likely due to the protein's long half-life as estimated by inhibition of protein synthesis by cycloheximide. Finally, in Qp luciferase reporter assays, the activity of Qp was shown to be regulated by cell cycle and to be dependent on the E2F sites within the Q locus. These findings demonstrate that transcriptional activity of Qp is cell cycle regulated and indicated that E2F serves as the stimulus for this regulation.


Assuntos
Ciclo Celular/genética , Antígenos Nucleares do Vírus Epstein-Barr/genética , Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/fisiologia , Latência Viral , Animais , Linhagem Celular , Antígenos Nucleares do Vírus Epstein-Barr/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
18.
Mol Cell Biol ; 19(4): 3216-23, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10082588

RESUMO

Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA-1) is the essential protein for maintenance of the EBV episome and establishment of latency. The BamHI Q promoter (Qp) is used for the transcription of EBNA-1 mRNA in type I and type II latency, which are EBV infection states exemplified by Burkitt's lymphoma and nasopharyngeal carcinoma. However, Qp is inactive in type III latency, and other promoters (the BamHI C promoter and/or the BamHI W promoter) are used for EBNA-1. The involvement of interferon regulatory factors (IRFs) in the regulation of Qp is suggested by the presence of an essential interferon-stimulated response element (ISRE) in the promoter. In this work, expression of IRF-2 is shown to be inversely associated with Qp status, i.e., IRF-2 levels are high in type III latency (when Qp is inactive) and low in type I latency (when Qp is active). Also, IRF-2 is identified by electrophoretic mobility shift assay as the major protein binding to the Qp ISRE in type III latency. In transient transfection assays, IRF-2 represses the activity of Qp-reporter constructs. Overexpression of IRF-2 in a type I latency cell line did not activate the endogenous Qp but marginally reduced the EBNA-1 mRNA level. Switching from type III latency (Qp inactive) to type II latency (Qp active), as produced by cell fusion, is directly associated with greatly reduced expression of IRF-2. These data strongly suggest that IRF-2 is a negative regulator of Qp and may contribute to the silencing of Qp in type III latency.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/biossíntese , Herpesvirus Humano 4/fisiologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição , Latência Viral/genética , Regulação para Baixo , Regulação Viral da Expressão Gênica , Genes Reporter , Fator Regulador 2 de Interferon , Fator Regulador 7 de Interferon , Regiões Promotoras Genéticas , Elementos de Resposta
19.
Clin Exp Metastasis ; 17(5): 431-6, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10651310

RESUMO

Type IV collagenases, matrix metalloproteinase (MMP) 2 and MMP9 are implicated in tumor invasion and metastasis. In patients with nasopharyngeal carcinoma (NPC), poor prognosis due to development of local and distant metastasis has been reported to be predicted by antibody titers against the Z protein which is an AP-1 family transcription factor encoded by the EBV BZLF1 immediate-early gene. Here we report that in patients with NPC, expression of Z in tumor cells correlates with advanced cervical lymph node metastasis which may suggest that Z affects tumor invasion and metastasis. We therefore tested if Z would induce expression of type IV collagenases. Transfection of Z expression plasmid into the C33A epithelial cell line increased expression of MMP9, but MMP2 expression was unaltered. Mutational analysis of the Z protein revealed that, in addition to all three functional domains of Z (dimerization domain, DNA binding domain, and activation domain), the carboxyl terminal 17 amino acids which stabilize the Z protein were necessary for induction of MMP9 expression. Analysis of the MMP9 promoter demonstrated that only AP-1 site close to the transcriptional start-site was essential for transactivation by Z. Previously we reported that Epstein-Barr virus latent membrane protein 1 (LMP1) stimulates MMP9 expression (Yoshizaki et al. Proc. Natl. Acad. Sci. 1998; 95: 3621-6). Thus, Z together with LMP1 may contribute to invasion and metastasis of NPC by inducing expression of MMP9.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Metaloproteinase 9 da Matriz/genética , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Transativadores/genética , Proteínas Virais , Proteínas de Ligação a DNA/biossíntese , Humanos , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/biossíntese , Metástase Neoplásica/genética , Transativadores/biossíntese
20.
J Virol ; 72(11): 8485-92, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9765385

RESUMO

The Epstein-Barr virus (EBV) DNA polymerase (pol) mRNA, which contains a noncanonical polyadenylation signal, UAUAAA, is cleaved and polyadenylated inefficiently (S. C. S. Key and J. S. Pagano, Virology 234:147-159, 1997). We postulated that the EBV early proteins SM and M, which appear to act posttranscriptionally and are homologs of herpes simplex virus (HSV) ICP27, might compensate for the inefficient processing of pol pre-mRNA. Here we show that the SM and M proteins interact with each other in vitro. In addition, glutathione S-transferase-SM/M fusion proteins precipitate the heterogeneous ribonucleoprotein (hnRNP) C1 splicing protein. Further, the SM protein is coimmunoprecipitated from SM-expressing cell extracts with an antibody to the hnRNP A1/A2 proteins, which are splicing and nuclear shuttling proteins. Finally, the amount of processed EBV DNA polymerase mRNA was increased three- to fourfold in a HeLa cell line expressing SM; this increase was not due to enhanced transcription. Thus, inefficient processing of EBV pol RNA by cellular cleavage and polyadenylation factors appears to be compensated for and may be regulated by the early EBV protein, SM, perhaps via RNA 3'-end formation.


Assuntos
DNA Polimerase Dirigida por DNA/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B , Ribonucleoproteínas Nucleares Heterogêneas Grupo C , Fosfoproteínas/metabolismo , Precursores de RNA/metabolismo , RNA Viral/metabolismo , Transativadores/metabolismo , Proteínas Virais/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Viral da Expressão Gênica , Genes Virais , Genes pol , Células HeLa , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas , Humanos , Fosfoproteínas/genética , Precursores de RNA/genética , Processamento Pós-Transcricional do RNA , RNA Viral/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ribonucleoproteínas/metabolismo , Transativadores/genética , Transfecção , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA