Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
Nat Commun ; 11(1): 877, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054837

ABSTRACT

Epstein-Barr virus (EBV) genomes persist in latently infected cells as extrachromosomal episomes that attach to host chromosomes through the tethering functions of EBNA1, a viral encoded sequence-specific DNA binding protein. Here we employ circular chromosome conformation capture (4C) analysis to identify genome-wide associations between EBV episomes and host chromosomes. We find that EBV episomes in Burkitt's lymphoma cells preferentially associate with cellular genomic sites containing EBNA1 binding sites enriched with B-cell factors EBF1 and RBP-jK, the repressive histone mark H3K9me3, and AT-rich flanking sequence. These attachment sites correspond to transcriptionally silenced genes with GO enrichment for neuronal function and protein kinase A pathways. Depletion of EBNA1 leads to a transcriptional de-repression of silenced genes and reduction in H3K9me3. EBV attachment sites in lymphoblastoid cells with different latency type show different correlations, suggesting that host chromosome attachment sites are functionally linked to latency type gene expression programs.


Subject(s)
Attachment Sites, Microbiological/genetics , Attachment Sites, Microbiological/physiology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Host Microbial Interactions/genetics , Burkitt Lymphoma/genetics , Burkitt Lymphoma/virology , Cell Line, Tumor , Chromosomes, Human/genetics , Chromosomes, Human/virology , Epigenesis, Genetic , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/pathogenicity , Host Microbial Interactions/physiology , Humans , Models, Biological , Plasmids/genetics , Virus Latency/genetics , Virus Latency/physiology
2.
Nucleic Acids Res ; 47(6): 2807-2821, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30649516

ABSTRACT

Epstein-Barr virus proteins EBNA3A, EBNA3B and EBNA3C control hundreds of host genes after infection. Changes in epigenetic marks around EBNA3-regulated genes suggest that they exert transcriptional control in collaboration with epigenetic factors. The roles of polycomb repressive complex (PRC)2 subunit SUZ12 and of PRC1 subunit BMI1 were assessed for their importance in EBNA3-mediated repression and activation. ChIP-seq experiments for SUZ12 and BMI1 were performed to determine their global localization on chromatin and analysis offered further insight into polycomb protein distribution in differentiated cells. Their localization was compared to that of each EBNA3 to resolve longstanding questions about the EBNA3-polycomb relationship. SUZ12 did not co-localize with any EBNA3, whereas EBNA3C co-localized significantly and co-immunoprecipitated with BMI1. In cells expressing a conditional EBNA3C, BMI1 was sequestered to EBNA3C-binding sites after EBNA3C activation. When SUZ12 or BMI1 was knocked down in the same cells, SUZ12 did not contribute to EBNA3C-mediated regulation. Surprisingly, after BMI1 knockdown, EBNA3C repressed equally efficiently but host gene activation by EBNA3C was impaired. This overturns previous assumptions about BMI1/PRC1 functions during EBNA3C-mediated regulation, for the first time identifies directly a host factor involved in EBNA3-mediated activation and provides a new insight into how PRC1 can be involved in gene activation.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/physiology , Host-Pathogen Interactions/genetics , Polycomb Repressive Complex 1/physiology , Transcriptional Activation , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Herpesvirus 4, Human/physiology , Humans , Polycomb Repressive Complex 1/metabolism , Protein Binding
4.
Cancer ; 120(3): 363-72, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24190575

ABSTRACT

BACKGROUND: The Epstein-Barr virus (EBV)-encoded EB nuclear antigen 1 (EBNA1) protein is required for maintenance and transmission of the viral episome in EBV-infected cells. The objective of this study was to investigate the role of EBNA1 protein in nasopharyngeal carcinoma (NPC). METHODS: Tissue samples from 48 patients with NPC and 12 patients with chronic nasopharyngitis were subjected to immunohistochemical analysis of EBNA1 expression. EBNA1 combinational DNA was used to overexpress EBNA1 protein in NPC cell lines to assess tumor cell epithelial-mesenchymal transition (EMT), colony formation, migration and invasion, and gene expression. RESULTS: EBNA1 protein was highly expressed in NPC tissue specimens, and its expression was associated with NPC lymph node metastasis. EBNA1 expression affected NPC cell morphology and the expression of EMT markers in vitro. Furthermore, overexpression of EBNA1 inhibited the expression of microRNA 200a (miR-200a) and miR-200b and, in turn, up-regulated expression of their target genes, zinc finger E-box binding homeobox 1 ( ZEB1) and ZEB2, which are well known mediators of EMT. In addition, EBNA1-regulated miR-200a and miR-200b expression was mediated by transforming growth factor-ß1. CONCLUSIONS: The current findings provided novel insight into the vital role of EBNA1 in manipulating a molecular switch of EMT in EBV-positive NPC cells.


Subject(s)
Epithelial-Mesenchymal Transition , Epstein-Barr Virus Nuclear Antigens/physiology , Nasopharyngeal Neoplasms/pathology , Carcinoma , Cell Line, Tumor , Cell Movement , Homeodomain Proteins/physiology , Humans , MicroRNAs/physiology , Nasopharyngeal Carcinoma , Neoplasm Invasiveness , Signal Transduction , Transcription Factors/physiology , Transforming Growth Factor beta1/physiology , Zinc Finger E-box-Binding Homeobox 1
5.
Proc Natl Acad Sci U S A ; 111(1): 243-8, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24344309

ABSTRACT

Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1) is essential for EBV episome maintenance, replication, and transcription. These effects are mediated by EBNA1 binding to cognate oriP DNA, which comprise 20 imperfect copies of a 30-bp dyad symmetry enhancer and an origin for DNA replication. To identify cell proteins essential for these EBNA1 functions, EBNA1 associated cell proteins were immune precipitated and analyzed by liquid chromatography-tandem mass spectrometry. Nucleolin (NCL) was identified to be EBNA1 associated. EBNA1's N-terminal 100 aa and NCL's RNA-binding domains were critical for EBNA1/NCL interaction. Lentivirus shRNA-mediated NCL depletion substantially reduced EBNA1 recruitment to oriP DNA, EBNA1-dependent transcription of an EBV oriP luciferase reporter, and EBV genome maintenance in lymphoblastoid cell lines. NCL RNA-binding domain K429 was critical for ATP and EBNA1 binding. NCL overexpression increased EBNA1 binding to oriP and transcription, whereas NCL K429A was deficient. Moreover, NCL silencing impaired lymphoblastoid cell line growth. These experiments reveal a surprisingly critical role for NCL K429 in EBNA1 episome maintenance and transcription, which may be a target for therapeutic intervention.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/physiology , Phosphoproteins/chemistry , Phosphoproteins/physiology , Plasmids/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Transcription, Genetic , Adenosine Triphosphate/chemistry , Binding Sites , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatography, Liquid , DNA Replication , Epitopes/chemistry , Gene Expression Regulation , Gene Silencing , Genome , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , Luciferases/metabolism , Mass Spectrometry , Microscopy, Confocal , Protein Binding , Protein Structure, Tertiary , Replication Origin , Virus Replication , Nucleolin
6.
Cancer Lett ; 330(2): 141-9, 2013 Apr 28.
Article in English | MEDLINE | ID: mdl-23220625

ABSTRACT

Nasopharyngeal carcinoma (NPC), an endemic head and neck cancer found in Southeast Asia, is etiologically associated with the infection of an oncogenic virus, the Epstein-Barr virus (EBV). Here, we review literature findings on the potential/putative role of STAT3 signaling in its pathogenesis and discuss anti-STAT3 targeting as a therapeutic and possibly prevention strategy for NPC. Lastly, the potential involvement of STAT3 in other oncovirus-associated (e.g. the human papillomavirus) head and neck cancers have not been investigated and we hope that findings in EBV-associated NPC can prompt future investigations in these cancers, which are of increasing impact in the Western countries.


Subject(s)
Epstein-Barr Virus Infections/complications , Nasopharyngeal Neoplasms/etiology , STAT3 Transcription Factor/physiology , Animals , Epstein-Barr Virus Nuclear Antigens/physiology , ErbB Receptors/physiology , Humans , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/virology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Viral Matrix Proteins/physiology , Viral Proteins/physiology
7.
Leukemia ; 26(11): 2343-52, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22614176

ABSTRACT

The discovery of microRNA (miR) represents a novel paradigm in RNA-based regulation of gene expression and their dysregulation has become a hallmark of many a tumor. In virally associated cancers, the host-pathogen interaction could involve alteration in miR expression. Epstein-Barr virus (EBV)-encoded EBNA2 is indispensable for the capacity of the virus to transform B cells in vitro. Here, we studied how it affects cellular miRs. Extensive miR profiling of the virus-infected and EBNA2-transfected B lymphoma cells revealed that oncomiR miR-21 is positively regulated by this viral protein. Conversely, Burkitt's lymphoma (BL) cell lines infected with EBNA2 lacking P3HR1 strain did not show any increase in miR-21. EBNA2 increased phosphorylation of AKT and this was directly correlated with increased miR-21. In contrast, miR-146a was downregulated by EBNA2 in B lymphoma cells. Low miR-146a expression correlates with an elevated level of IRAK1 and type I interferon in EBNA2 transfectants. Taken together, the present data suggest that EBNA2 might contribute to EBV-induced B-cell transformation by altering miR expression and in particular by increasing oncomiR-like miR-21 and by affecting the antiviral responses of the innate immune system through downregulation of its key regulator miR-146a.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/genetics , MicroRNAs/physiology , Viral Proteins/physiology , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Knockdown Techniques , Humans , MicroRNAs/genetics , Viral Proteins/genetics
8.
J Clin Invest ; 122(4): 1487-502, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22406538

ABSTRACT

Epstein-Barr virus (EBV) persistently infects more than 90% of the human population and is etiologically linked to several B cell malignancies, including Burkitt lymphoma (BL), Hodgkin lymphoma (HL), and diffuse large B cell lymphoma (DLBCL). Despite its growth transforming properties, most immune-competent individuals control EBV infection throughout their lives. EBV encodes various oncogenes, and of the 6 latency-associated EBV-encoded nuclear antigens, only EBNA3B is completely dispensable for B cell transformation in vitro. Here, we report that infection with EBV lacking EBNA3B leads to aggressive, immune-evading monomorphic DLBCL-like tumors in NOD/SCID/γc-/- mice with reconstituted human immune system components. Infection with EBNA3B-knockout EBV (EBNA3BKO) induced expansion of EBV-specific T cells that failed to infiltrate the tumors. EBNA3BKO-infected B cells expanded more rapidly and secreted less T cell-chemoattractant CXCL10, reducing T cell recruitment in vitro and T cell-mediated killing in vivo. B cell lines from 2 EBV-positive human lymphomas encoding truncated EBNA3B exhibited gene expression profiles and phenotypic characteristics similar to those of tumor-derived lines from the humanized mice, including reduced CXCL10 secretion. Screening EBV-positive DLBCL, HL, and BL human samples identified additional EBNA3B mutations. Thus, EBNA3B is a virus-encoded tumor suppressor whose inactivation promotes immune evasion and virus-driven lymphomagenesis.


Subject(s)
Cell Transformation, Viral/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/physiology , Genes, Tumor Suppressor , Genes, Viral , Herpesvirus 4, Human/physiology , Lymphoma, B-Cell/virology , Lymphoproliferative Disorders/virology , Postoperative Complications/virology , Tumor Suppressor Proteins/physiology , Tumor Virus Infections/virology , Animals , Cell Line, Transformed/transplantation , Cell Line, Transformed/virology , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/deficiency , Chemokine CXCL10/genetics , Chimera , DNA Mutational Analysis , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Deletion , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lymphoma, B-Cell/genetics , Lymphoproliferative Disorders/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mutation , Postoperative Complications/genetics , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics , Tumor Virus Infections/genetics
9.
Semin Cancer Biol ; 22(2): 154-61, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22206863

ABSTRACT

EBNA1 is expressed in all NPC tumours and is the only Epstein-Barr virus protein needed for the stable persistence of EBV episomes. EBNA1 binds to specific sequences in the EBV genome to facilitate the initiation of DNA synthesis, ensure the even distribution of the viral episomes to daughter cells during mitosis and to activate the transcription of other viral latency genes important for cell immortalization. In addition, EBNA1 has been found to alter cellular pathways in multiple ways that likely contribute to cell immortalization and malignant transformation. This chapter discusses the known functions and cellular effects of EBNA1, especially as pertains to NPC.


Subject(s)
Cell Transformation, Neoplastic , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/physiology , Nasopharyngeal Neoplasms/virology , Carcinoma , Cell Proliferation , Cell Survival , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/metabolism , Host-Pathogen Interactions , Humans , Models, Biological , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Signal Transduction
10.
Leukemia ; 25(6): 1017-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21394098

ABSTRACT

The Epstein-Barr virus (EBV) nuclear antigen (EBNA)-1 promotes the accumulation of chromosomal aberrations in malignant B cells by inducing oxidative stress. Here we report that this phenotype is associated with telomere dysfunction. Stable or conditional expression of EBNA1 induced telomere abnormalities including loss or gain of telomere signals, telomere fusion and heterogeneous length of telomeres. This was accompanied by the accumulation of extrachromosomal telomeres, telomere dysfunction-induced foci (TIFs) containing phosphorylated histone H2AX and the DNA damage response protein 53BP1, telomere-associated promyelocytic leukemia nuclear bodies (APBs), telomeric-sister chromatid exchanges and displacement of the shelterin protein TRF2. The induction of TIFs and APBs was inhibited by treatment with scavengers of reactive oxygen species (ROS) that also promoted the relocalization of TRF2 at telomeres. These findings highlight a novel mechanism by which EBNA1 may promote malignant transformation and tumor progression.


Subject(s)
Cell Transformation, Viral , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/physiology , Oxidative Stress , Telomere/pathology , Cell Line, Tumor , Chromosome Aberrations , DNA Repair , Histones/metabolism , Humans , Phosphorylation , Reactive Oxygen Species
11.
Proc Natl Acad Sci U S A ; 108(5): 1919-24, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21245331

ABSTRACT

Epstein-Barr virus (EBV) nuclear antigen 3C (EBNA3C) and EBNA3A are each essential for EBV conversion of primary human B lymphocytes into continuously proliferating lymphoblast cell lines (LCLs) and for maintaining LCL growth. We now find that EBNA3C and EBNA3A's essential roles are to repress p16(INK4A) and p14(ARF). In the absence of EBNA3C or EBNA3A, p16(INK4A) and p14(ARF) expression increased and cell growth ceased. EBNA3C inactivation did not alter p16(INK4A) promoter CpG methylation, but reduced already low H3K27me3, relative to resting B cells, and increased H3K4me3 and H3-acetylation, linking EBNA3C inactivation to histone modifications associated with increased transcription. Importantly, knockdown of p16(INK4A) or p14(ARF) partially rescued LCLs from EBNA3C or EBNA3A inactivation-induced growth arrest and knockdown of both rescued LCL growth, confirming central roles for p16(INK4A) and p14(ARF) in LCL growth arrest following EBNA3C or EBNA3A inactivation. Moreover, blockade of p16(INK4A) and p14(ARF) effects on pRb and p53 by human papilloma virus type 16 E7 and E6 expression, sustained LCL growth after EBNA3C or EBNA3A inactivation. These data indicate that EBNA3C and EBNA3A joint repression of CDKN2A p16(INK4A) and p14(ARF) is essential for LCL growth.


Subject(s)
Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Epstein-Barr Virus Nuclear Antigens/physiology , Tumor Suppressor Protein p14ARF/physiology , Cell Line , CpG Islands , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Gene Knockdown Techniques , Humans , Promoter Regions, Genetic , Tumor Suppressor Protein p14ARF/genetics
12.
Cell Host Microbe ; 8(6): 510-22, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21147465

ABSTRACT

Epstein-Barr virus (EBV), an oncogenic herpesvirus that causes human malignancies, infects and immortalizes primary human B cells in vitro into indefinitely proliferating lymphoblastoid cell lines, which represent a model for EBV-induced tumorigenesis. The immortalization efficiency is very low, suggesting that an innate tumor suppressor mechanism is operative. We identify the DNA damage response (DDR) as a major component of the underlying tumor suppressor mechanism. EBV-induced DDR activation was not due to lytic viral replication, nor did the DDR marks colocalize with latent episomes. Rather, a transient period of EBV-induced hyperproliferation correlated with DDR activation. Inhibition of the DDR kinases ATM and Chk2 markedly increased transformation efficiency of primary B cells. Further, the viral latent oncoprotein EBNA3C was required to attenuate the EBV-induced DDR. We propose that heightened oncogenic activity in early cell divisions activates a growth-suppressive DDR that is attenuated by viral latency products to induce cell immortalization.


Subject(s)
B-Lymphocytes/virology , Cell Cycle Proteins/physiology , DNA Damage , DNA-Binding Proteins/physiology , Herpesvirus 4, Human/physiology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/pathology , Cell Proliferation , Cell Transformation, Viral , Cells, Cultured , Checkpoint Kinase 2 , Epstein-Barr Virus Nuclear Antigens/physiology , Humans , Signal Transduction
13.
PLoS One ; 5(11): e13979, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21085583

ABSTRACT

Epstein-Barr virus (EBV) is able to drive the transformation of B-cells, resulting in the generation of lymphoblastoid cell lines (LCLs) in vitro. EBV nuclear proteins EBNA3A and EBNA3C are necessary for efficient transformation, while EBNA3B is dispensable. We describe a transcriptome analysis of BL31 cells infected with a series of EBNA3-knockout EBVs, including one deleted for all three EBNA3 genes. Using Affymetrix Exon 1.0 ST microarrays analysed with the MMBGX algorithm, we have identified over 1000 genes whose regulation by EBV requires one of the EBNA3s. Remarkably, a third of the genes identified require more than one EBNA3 for their regulation, predominantly EBNA3C co-operating with either EBNA3B, EBNA3A or both. The microarray was validated by real-time PCR, while ChIP analysis of a selection of co-operatively repressed promoters indicates a role for polycomb group complexes. Targets include genes involved in apoptosis, cell migration and B-cell differentiation, and show a highly significant but subtle alteration in genes involved in mitosis. In order to assess the relevance of the BL31 system to LCLs, we analysed the transcriptome of a set of EBNA3B knockout (3BKO) LCLs. Around a third of the genes whose expression level in LCLs was altered in the absence of EBNA3B were also altered in 3BKO-BL31 cell lines.Among these are TERT and TCL1A, implying that EBV-induced changes in the expression of these genes are not required for B-cell transformation. We also identify 26 genes that require both EBNA3A and EBNA3B for their regulation in LCLs. Together, this shows the complexity of the interaction between EBV and its host, whereby multiple EBNA3 proteins co-operate to modulate the behaviour of the host cell.


Subject(s)
Antigens, Viral/genetics , Chromatin/metabolism , Gene Expression Profiling , Antigens, Viral/physiology , Cell Line, Tumor , Cluster Analysis , Epigenomics , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/physiology , Gene Knockout Techniques , HEK293 Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions/genetics , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
14.
PLoS Pathog ; 6(10): e1001151, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20976201

ABSTRACT

Viruses are known to employ different strategies to manipulate the major histocompatibility (MHC) class I antigen presentation pathway to avoid recognition of the infected host cell by the immune system. However, viral control of antigen presentation via the processes that supply and select antigenic peptide precursors is yet relatively unknown. The Epstein-Barr virus (EBV)-encoded EBNA1 is expressed in all EBV-infected cells, but the immune system fails to detect and destroy EBV-carrying host cells. This immune evasion has been attributed to the capacity of a Gly-Ala repeat (GAr) within EBNA1 to inhibit MHC class I restricted antigen presentation. Here we demonstrate that suppression of mRNA translation initiation by the GAr in cis is sufficient and necessary to prevent presentation of antigenic peptides from mRNAs to which it is fused. Furthermore, we demonstrate a direct correlation between the rate of translation initiation and MHC class I antigen presentation from a certain mRNA. These results support the idea that mRNAs, and not the encoded full length proteins, are used for MHC class I restricted immune surveillance. This offers an additional view on the role of virus-mediated control of mRNA translation initiation and of the mechanisms that control MHC class I restricted antigen presentation in general.


Subject(s)
Antigen Presentation/genetics , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/immunology , Histocompatibility Antigens Class I/metabolism , Immune Evasion/genetics , RNA, Messenger/metabolism , Animals , Antigen Presentation/immunology , Base Sequence , Cell Line, Tumor , Dipeptides/chemistry , Dipeptides/immunology , Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/genetics , Eukaryotic Initiation Factors/metabolism , Eukaryotic Initiation Factors/physiology , Herpesvirus 4, Human/genetics , Histocompatibility Antigens Class I/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune Evasion/immunology , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/immunology , Mice , Models, Biological , Nucleic Acid Conformation , Protein Biosynthesis/genetics , Protein Biosynthesis/immunology , RNA, Messenger/genetics , Repetitive Sequences, Amino Acid/immunology , Repetitive Sequences, Amino Acid/physiology
15.
J Virol ; 84(20): 10488-500, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20686042

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of primary effusion lymphoma (PEL). All PEL cell lines are infected with KSHV, and 70% are coinfected with Epstein-Barr virus (EBV). KSHV reactivation from latency requires promoter-specific transactivation by the KSHV Rta protein through interactions with RBP-Jk (CSL), the cellular DNA-binding component of the Notch signal transduction pathway. EBV transformation of primary B cells requires EBV nuclear antigen 2 (EBNA-2) to interact with RBP-Jk to direct the latent viral and cellular gene expression program. Although KSHV Rta and EBV EBNA-2 both require RBP-Jk for transactivation, previous studies have suggested that RBP-Jk-dependent transactivators do not function identically. We have found that the EBV latent protein LMP-1 is expressed in less than 5% of KSHV(+)/EBV(+) PEL cells but is induced in an Rta-dependent fashion when KSHV reactivates. KSHV Rta transactivates the EBV latency promoters in an RBP-Jk-dependent fashion and forms a ternary complex with RBP-Jk on the promoters. In B cells that are conditionally transformed by EBV alone, we show that KSHV Rta complements a short-term EBNA-2 growth deficiency in an autocrine/paracrine manner. Complementation of EBNA-2 deficiency by Rta depends on RBP-Jk and LMP-1, and Rta transactivation is required for optimal growth of KSHV(+)/EBV(+) PEL lines. Our data suggest that Rta can contribute to EBV-driven cellular growth by transactivating RBP-Jk-dependent EBV latency genes. However, our data also suggest that EBNA-2 and Rta induce distinct alterations in the cellular proteomes that contribute to the growth of infected cells.


Subject(s)
Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Receptors, Notch/physiology , Animals , Base Sequence , Cell Line , Cell Proliferation , Culture Media, Conditioned , DNA Primers/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/physiology , Gene Expression , Genes, Viral , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Host-Pathogen Interactions/physiology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Mice , Mice, Knockout , Models, Biological , Promoter Regions, Genetic , Signal Transduction , Trans-Activators/genetics , Trans-Activators/physiology , Transfection , Viral Matrix Proteins/genetics , Viral Matrix Proteins/physiology , Viral Proteins/genetics , Viral Proteins/physiology , Virus Activation/physiology
16.
J Virol ; 84(21): 11113-23, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20719947

ABSTRACT

Latent Epstein-Barr virus (EBV) infection is an important causative factor in the development of several cancers, including nasopharyngeal carcinoma (NPC). The one EBV protein expressed in the nucleus of NPC cells, EBNA1, has been shown to disrupt promyelocitic leukemia (PML) nuclear bodies (NBs) by inducing the degradation of PML proteins, leading to impaired DNA repair and increased cell survival. Although EBNA1-mediated PML disruption is likely to be an important factor in the development of NPC, little is known about its mechanism. We now show that an interaction between EBNA1 and the host CK2 kinase is crucial for EBNA1 to disrupt PML bodies and degrade PML proteins. EBNA1 increases the association of CK2 with PML proteins, thereby increasing the phosphorylation of PML proteins by CK2, a modification that is known to trigger the polyubiquitylation and degradation of PML. The interaction between EBNA1 and CK2 is direct and occurs through the ß regulatory subunit of CK2 and EBNA1 amino acids 387 to 394. The binding of EBNA1 to the host ubiquitin specific protease USP7 has also been shown to be important for EBNA1-mediated PML disruption. We show that EBNA1 also increases the occupancy of USP7 at PML NBs and that CK2 and USP7 bind independently and simultaneously to EBNA1 to form a ternary complex. The combined results indicate that EBNA1 usurps two independent cellular pathways to trigger the loss of PML NBs.


Subject(s)
Casein Kinase II/metabolism , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/pathogenicity , Intranuclear Inclusion Bodies/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Binding Sites , Cell Line, Tumor , Host-Pathogen Interactions , Humans , Nasopharyngeal Neoplasms/etiology , Phosphorylation , Promyelocytic Leukemia Protein , Protein Binding , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Peptidase 7
17.
J Virol ; 84(10): 4988-97, 2010 May.
Article in English | MEDLINE | ID: mdl-20219917

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) is an abundant, chromatin-associated, NAD-dependent enzyme that functions in multiple chromosomal processes, including DNA replication and chromatin remodeling. The Epstein-Barr virus (EBV) origin of plasmid replication (OriP) is a dynamic genetic element that confers stable episome maintenance, DNA replication initiation, and chromatin organization functions. OriP function depends on the EBV-encoded origin binding protein EBNA1. We have previously shown that EBNA1 is subject to negative regulation by poly(ADP-ribosyl)ation (PARylation). We now show that PARP1 physically associates with OriP in latently EBV-infected B cells. Short hairpin RNA depletion of PARP1 enhances OriP replication activity and increases EBNA1, origin recognition complex 2 (ORC2), and minichromosome maintenance complex (MCM) association with OriP. Pharmacological inhibitors of PARP1 enhance OriP plasmid maintenance and increase EBNA1, ORC2, and MCM3 occupancy at OriP. PARylation in vitro inhibits ORC2 recruitment and remodels telomere repeat factor (TRF) binding at the dyad symmetry (DS) element of OriP. Purified PARP1 can ribosylate EBNA1 at multiple sites throughout its amino terminus but not in the carboxy-terminal DNA binding domain. We also show that EBNA1 linking regions (LR1 and LR2) can bind directly to oligomers of PAR. We propose that PARP1-dependent PARylation of EBNA1 and adjacently bound TRF2 induces structural changes at the DS element that reduce EBNA1 DNA binding affinity and functional recruitment of ORC.


Subject(s)
B-Lymphocytes/virology , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Poly(ADP-ribose) Polymerases/physiology , Virus Replication , Cell Line , Gene Knockdown Techniques , Humans , Plasmids , Poly (ADP-Ribose) Polymerase-1 , RNA, Small Interfering/genetics
18.
Cancer Sci ; 101(4): 876-81, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20132216

ABSTRACT

The oncogenic human herpes virus, the Epstein-Barr virus (EBV), expresses EBNA1 in almost all forms of viral latency. EBNA1 plays a major role in the maintenance of the viral genome and in the transactivation of viral transforming genes, including EBNA2 and latent membrane protein (LMP-1). However, it is unknown whether inhibition of EBNA1 from the onset of EBV infection disrupts the establishment of EBV's latency and transactivation of the viral oncogenes. To address this, we measured EBV infection kinetics in the B cell lines BALL-1 and BJAB, which stably express a dominant-negative EBNA1 (dnE1) fused to green fluorescent protein (GFP). The EBV genome was surprisingly unstable 1 week post-infection: the average loss rate of EBV DNA from GFP- and GFP-dnE1-expressing cells was 53.4% and 41.0% per cell generation, respectively, which was substantially higher than that of an 'established'oriP replicon (2-4%). GFP-dnE1 did not accelerate loss of the EBV genome, suggesting that EBNA1-dependent licensing of the EBV genome occurs infrequently during the acute phase of EBV infection. In the subacute phase, establishment of EBV latency was completely blocked in GFP-dnE1-expressing cells. In contrast, C/W promoter-driven transcription was strongly restricted in GFP-dnE1-expressing cells at 2 days post-infection. These data suggest that inhibition of EBNA1 from the onset of EBV infection is effective in blocking the positive feedback loop in the transactivation of viral transforming genes, and in eradicating the EBV genome during the subacute phase. Our results suggest that gene transduction of GFP-dnE1 could be a promising therapeutic and prophylactic approach toward EBV-associated malignancies.


Subject(s)
Cell Transformation, Viral , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens/physiology , Gene Expression Profiling , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Viral Proteins/pharmacology , Acute Disease , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Burkitt Lymphoma/virology , Cell Line, Tumor , Epstein-Barr Virus Infections/virology , Gene Expression , Genes, Viral , Genome, Viral , Herpesvirus 4, Human/immunology , Humans , Oncogenes , Transcriptional Activation , Virus Latency/genetics , Virus Latency/immunology
19.
J Gen Virol ; 91(Pt 4): 829-46, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20089802

ABSTRACT

Viruses that establish lifelong latent infections must ensure that the viral genome is maintained within the latently infected cell throughout the life of the host, yet at the same time must also be capable of avoiding elimination by the immune surveillance system. Gammaherpesviruses, which include the human viruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus, establish latent infections in lymphocytes. Infection of this dynamic host-cell population requires that the viruses have appropriate strategies for enabling the viral genome to persist while these cells go through rounds of mitosis, but at the same time must avoid detection by host CD8(+) cytotoxic T lymphocytes (CTLs). The majority of gammaherpesviruses studied have been found to encode a specific protein that is critical for maintenance of the viral genome within latently infected cells. This protein is termed the genome maintenance protein (GMP). Due to its vital role in long-term latency, this offers the immune system a crucial target for detection and elimination of virus-infected cells. GMPs from different gammaherpesviruses have evolved related strategies that allow the protein to be present within latently infected cells, but to remain effectively hidden from circulating CD8(+) CTLs. In this review, I will summarize the role of the GMPs and highlight the available data describing the immune-evasion properties of these proteins.


Subject(s)
Gammaherpesvirinae/immunology , Genome, Viral , Immune Evasion , Viral Proteins/physiology , Animals , Antigens, Viral/physiology , Epstein-Barr Virus Nuclear Antigens/immunology , Epstein-Barr Virus Nuclear Antigens/physiology , Gammaherpesvirinae/classification , Humans , Nuclear Proteins/physiology , T-Lymphocytes, Cytotoxic/immunology , Virus Latency
20.
PLoS Pathog ; 5(10): e1000624, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19834552

ABSTRACT

The EBNA1 protein of Epstein-Barr virus (EBV) plays essential roles in enabling the replication and persistence of EBV genomes in latently infected cells and activating EBV latent gene expression, in all cases by binding to specific recognition sites in the latent origin of replication, oriP. Here we show that EBNA1 binding to its recognition sites in vitro is greatly stimulated by binding to the cellular deubiquitylating enzyme, USP7, and that USP7 can form a ternary complex with DNA-bound EBNA1. Consistent with the in vitro effects, the assembly of EBNA1 on oriP elements in human cells was decreased by USP7 silencing, whereas assembly of an EBNA1 mutant defective in USP7 binding was unaffected. USP7 affinity column profiling identified a complex between USP7 and human GMP synthetase (GMPS), which was shown to stimulate the ability of USP7 to cleave monoubiquitin from histone H2B in vitro. Accordingly, silencing of USP7 in human cells resulted in a consistent increase in the level of monoubquitylated H2B. The USP7-GMPS complex formed a quaternary complex with DNA-bound EBNA1 in vitro and, in EBV infected cells, was preferentially detected at the oriP functional element, FR, along with EBNA1. Down-regulation of USP7 reduced the level of GMPS at the FR, increased the level of monoubiquitylated H2B in this region of the origin and decreased the ability of EBNA1, but not an EBNA1 USP7-binding mutant, to activate transcription from the FR. The results indicate that USP7 can stimulate EBNA1-DNA interactions and that EBNA1 can alter histone modification at oriP through recruitment of USP7.


Subject(s)
DNA Replication/physiology , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/genetics , Histones/metabolism , Replication Origin , Animals , Cells, Cultured , DNA-Directed DNA Polymerase/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , HeLa Cells , Herpesvirus 4, Human/metabolism , Histones/physiology , Humans , Multienzyme Complexes/metabolism , Multiprotein Complexes/metabolism , Protein Binding , Protein Processing, Post-Translational , Spodoptera , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/physiology , Ubiquitin-Specific Peptidase 7 , Ubiquitination/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...