Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Biol ; 15(8): e2001992, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28771465

ABSTRACT

Mature human B cells infected by Epstein-Barr virus (EBV) become activated, grow, and proliferate. If the cells are infected ex vivo, they are transformed into continuously proliferating lymphoblastoid cell lines (LCLs) that carry EBV DNA as extra-chromosomal episomes, express 9 latency-associated EBV proteins, and phenotypically resemble antigen-activated B-blasts. In vivo similar B-blasts can differentiate to become memory B cells (MBC), in which EBV persistence is established. Three related latency-associated viral proteins EBNA3A, EBNA3B, and EBNA3C are transcription factors that regulate a multitude of cellular genes. EBNA3B is not necessary to establish LCLs, but EBNA3A and EBNA3C are required to sustain proliferation, in part, by repressing the expression of tumour suppressor genes. Here we show, using EBV-recombinants in which both EBNA3A and EBNA3C can be conditionally inactivated or using virus completely lacking the EBNA3 gene locus, that-after a phase of rapid proliferation-infected primary B cells express elevated levels of factors associated with plasma cell (PC) differentiation. These include the cyclin-dependent kinase inhibitor (CDKI) p18INK4c, the master transcriptional regulator of PC differentiation B lymphocyte-induced maturation protein-1 (BLIMP-1), and the cell surface antigens CD38 and CD138/Syndecan-1. Chromatin immunoprecipitation sequencing (ChIP-seq) and chromatin immunoprecipitation quantitative PCR (ChIP-qPCR) indicate that in LCLs inhibition of CDKN2C (p18INK4c) and PRDM1 (BLIMP-1) transcription results from direct binding of EBNA3A and EBNA3C to regulatory elements at these loci, producing stable reprogramming. Consistent with the binding of EBNA3A and/or EBNA3C leading to irreversible epigenetic changes, cells become committed to a B-blast fate <12 days post-infection and are unable to de-repress p18INK4c or BLIMP-1-in either newly infected cells or conditional LCLs-by inactivating EBNA3A and EBNA3C. In vitro, about 20 days after infection with EBV lacking functional EBNA3A and EBNA3C, cells develop a PC-like phenotype. Together, these data suggest that EBNA3A and EBNA3C have evolved to prevent differentiation to PCs after infection by EBV, thus favouring long-term latency in MBC and asymptomatic persistence.


Subject(s)
B-Lymphocytes/virology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/physiology , Viral Proteins/physiology , Virus Latency , B-Lymphocytes/physiology , Biomarkers/metabolism , Cell Differentiation , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Histone Code , Humans , Immunoglobulins/metabolism , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/metabolism
2.
Nucleic Acids Res ; 45(5): 2368-2383, 2017 03 17.
Article in English | MEDLINE | ID: mdl-27903901

ABSTRACT

ChIP-seq performed on lymphoblastoid cell lines (LCLs), expressing epitope-tagged EBNA3A, EBNA3B or EBNA3C from EBV-recombinants, revealed important principles of EBNA3 binding to chromatin. When combined with global chromatin looping data, EBNA3-bound loci were found to have a singular character, each directly associating with either EBNA3-repressed or EBNA3-activated genes, but not with both. EBNA3A and EBNA3C showed significant association with repressed and activated genes. Significant direct association for EBNA3B loci could only be shown with EBNA3B-repressed genes. A comparison of EBNA3 binding sites with known transcription factor binding sites in LCL GM12878 revealed substantial co-localization of EBNA3s with RUNX3-a protein induced by EBV during B cell transformation. The beta-subunit of core binding factor (CBFß), that heterodimerizes with RUNX3, could co-immunoprecipitate robustly EBNA3B and EBNA3C, but only weakly EBNA3A. Depletion of either RUNX3 or CBFß with lentivirus-delivered shRNA impaired epitope-tagged EBNA3B and EBNA3C binding at multiple regulated gene loci, indicating a requirement for CBF heterodimers in EBNA3 recruitment during target-gene regulation. ShRNA-mediated depletion of CBFß in an EBNA3C-conditional LCL confirmed the role of CBF in the regulation of EBNA3C-induced and -repressed genes. These results reveal an important role for RUNX3/CBF during B cell transformation and EBV latency that was hitherto unexplored.


Subject(s)
Core Binding Factors/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation , Binding Sites , Cell Line , Chromatin/chemistry , Chromatin/metabolism , Chromatin Immunoprecipitation , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/physiology , Core Binding Factor beta Subunit/metabolism , Core Binding Factors/physiology , Enhancer Elements, Genetic , Genome, Human , Humans , Transcription Factors/metabolism , Transcription Initiation Site
3.
PLoS Pathog ; 11(7): e1005031, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26153983

ABSTRACT

We show that two host-encoded primary RNAs (pri-miRs) and the corresponding microRNA (miR) clusters--widely reported to have cell transformation-associated activity--are regulated by EBNA3A and EBNA3C. Utilising a variety of EBV-transformed lymphoblastoid cell lines (LCLs) carrying knockout-, revertant- or conditional-EBV recombinants, it was possible to demonstrate unambiguously that EBNA3A and EBNA3C are both required for transactivation of the oncogenic miR-221/miR-222 cluster that is expressed at high levels in multiple human tumours--including lymphoma/leukemia. ChIP, ChIP-seq, and chromosome conformation capture analyses indicate that this activation results from direct targeting of both EBV proteins to chromatin at the miR-221/miR-222 genomic locus and activation via a long-range interaction between enhancer elements and the transcription start site of a long non-coding pri-miR located 28 kb upstream of the miR sequences. Reduced levels of miR-221/miR-222 produced by inactivation or deletion of EBNA3A or EBNA3C resulted in increased expression of the cyclin-dependent kinase inhibitor p57KIP2, a well-established target of miR-221/miR-222. MiR blocking experiments confirmed that miR-221/miR-222 target p57KIP2 expression in LCLs. In contrast, EBNA3A and EBNA3C are necessary to silence the tumour suppressor cluster miR-143/miR-145, but here ChIP-seq suggests that repression is probably indirect. This miR cluster is frequently down-regulated or deleted in human cancer, however, the targets in B cells are unknown. Together these data indicate that EBNA3A and EBNA3C contribute to B cell transformation by inhibiting multiple tumour suppressor proteins, not only by direct repression of protein-encoding genes, but also by the manipulation of host long non-coding pri-miRs and miRs.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , MicroRNAs/genetics , B-Lymphocytes/virology , Blotting, Western , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Epstein-Barr Virus Nuclear Antigens/metabolism , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunoprecipitation , MicroRNAs/biosynthesis , Oncogenes , Real-Time Polymerase Chain Reaction
4.
PLoS Pathog ; 9(2): e1003187, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23436997

ABSTRACT

To explore the role of p16(INK4a) as an intrinsic barrier to B cell transformation by EBV, we transformed primary B cells from an individual homozygous for a deletion in the CDKN2A locus encoding p16(INK4a) and p14(ARF). Using recombinant EBV-BAC viruses expressing conditional EBNA3C (3CHT), we developed a system that allows inactivation of EBNA3C in lymphoblastoid cell lines (LCLs) lacking active p16(INK4a) protein but expressing a functional 14(ARF)-fusion protein (p14/p16). The INK4a locus is epigenetically repressed by EBNA3C--in cooperation with EBNA3A--despite the absence of functional p16(INK4a). Although inactivation of EBNA3C in LCLs from normal B cells leads to an increase in p16(INK4a) and growth arrest, EBNA3C inactivation in the p16(INK4a)-null LCLs has no impact on the rate of proliferation, establishing that the repression of INK4a is a major function of EBNA3C in EBV-driven LCL proliferation. This conditional LCL system allowed us to use microarray analysis to identify and confirm genes regulated specifically by EBNA3C, independently of proliferation changes modulated by the p16(INK4a)-Rb-E2F axis. Infections of normal primary B cells with recombinant EBV-BAC virus from which EBNA3C is deleted or with 3CHT EBV in the absence of activating ligand 4-hydroxytamoxifen, revealed that EBNA3C is necessary to overcome an EBV-driven increase in p16(INK4a) expression and concomitant block to proliferation 2-4 weeks post-infection. If cells are p16(INK4a)-null, functional EBNA3C is dispensable for the outgrowth of LCLs.


Subject(s)
B-Lymphocytes/virology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Epigenetic Repression/genetics , Herpesvirus 4, Human/physiology , Lymphocyte Activation , Antigens, Viral/genetics , Antigens, Viral/metabolism , Cell Line , Cell Proliferation , Cell Survival , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Genetic Loci , Herpesvirus 4, Human/immunology , Humans , Oligonucleotide Array Sequence Analysis , Phosphorylation , Primary Cell Culture , Virus Latency
5.
Nucleic Acids Res ; 40(15): 7233-46, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22584624

ABSTRACT

Detailed analyses of the chromatin around the BIM promoter has revealed that latent Epstein-Barr virus (EBV) triggers the recruitment of polycomb repressive complex 2 (PRC2) core subunits and the trimethylation of histone H3 lysine 27 (H3K27me3) at this locus. The recruitment is absolutely dependent on nuclear proteins EBNA3A and EBNA3C; what is more, epitope-tagged EBNA3C could be shown bound near the transcription start site (TSS). EBV induces no consistent changes in the steady-state expression of PRC2 components, but lentivirus delivery of shRNAs against PRC2 and PRC1 subunits disrupted EBV repression of BIM. The activation mark H3K4me3 is largely unaltered at this locus irrespective of H3K27me3 status, suggesting the establishment of a 'bivalent' chromatin domain. Consistent with the 'poised' nature of these domains, RNA polymerase II (Pol II) occupancy was not altered by EBV at the BIM TSS, but analysis of phospho-serine 5 on Pol II indicated that EBNA3A and EBNA3C together inhibit initiation of BIM transcripts. B cell lines carrying EBV encoding a conditional EBNA3C-oestrogen receptor-fusion revealed that this epigenetic repression of BIM was reversible, but took more than 3 weeks from when EBNA3C was inactivated.


Subject(s)
Antigens, Viral/metabolism , Apoptosis Regulatory Proteins/genetics , Herpesvirus 4, Human/physiology , Membrane Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Repressor Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Line , Epstein-Barr Virus Nuclear Antigens/metabolism , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Polycomb-Group Proteins , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , RNA Polymerase II/metabolism , Repressor Proteins/antagonists & inhibitors , Transcription, Genetic
6.
J Biol Chem ; 277(49): 47197-204, 2002 Dec 06.
Article in English | MEDLINE | ID: mdl-12372828

ABSTRACT

CtBP (carboxyl-terminal binding protein) has been shown to be a highly conserved co-repressor of transcription that is important in development, cell cycle regulation, and transformation. Viral proteins E1A and EBNA3C and all the various Drosophila and vertebrate transcription factors to which CtBP has been reported to bind contain a conserved "PXDLS" CtBP-interaction domain. Here we show that EBNA3A binds CtBP both in vitro and in vivo but that this interaction does not require a near consensus (98)PLDLR(102) motif in the NH(2) terminus of EBNA3A. However, further deletion and mutation analysis revealed that CtBP interacts with this viral protein through a cryptic, bipartite motif located in the COOH terminus of EBNA3A. The two components of this binding domain are similar to the canonical PXDLS motif but do not include the highly conserved, and normally critical, first proline residue. These nonconsensus sites, (857)ALDLS(861) and (886)VLDLS(890), synergize to produce very efficient binding to CtBP. Interaction with CtBP was shown to be important in the repression of transcription by EBNA3A and in the ability of EBNA3A to cooperate with activated Ras to immortalize and transform primary rat embryo fibroblasts. Similar bipartite sequences can be found in other viral and cellular proteins that can interact with CtBP, including the retinoblastoma-interacting protein-methyltransferase RIZ, the oncoprotein EVI1, and Marek's disease virus transforming protein Meq.


Subject(s)
Antigens, Viral/chemistry , DNA-Binding Proteins/chemistry , Epstein-Barr Virus Nuclear Antigens/chemistry , Phosphoproteins/chemistry , Alcohol Oxidoreductases , Amino Acid Motifs , Amino Acid Sequence , Animals , Antigens, Viral/metabolism , Binding Sites , Blotting, Western , Cell Line, Transformed , DNA-Binding Proteins/metabolism , Down-Regulation , Fibroblasts/metabolism , Gene Deletion , Genes, Reporter , Glutathione Transferase/metabolism , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Oncogene Proteins, Viral/metabolism , Phosphoproteins/metabolism , Plasmids/metabolism , Precipitin Tests , Proline/chemistry , Protein Binding , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid , Transcription, Genetic , Transfection , Tumor Cells, Cultured , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...