EBNA2 Drives Formation of New Chromosome Binding Sites and Target Genes for B-Cell Master Regulatory Transcription Factors RBP-jκ and EBF1

Fang Lu, Horng-Shen Chen, Andrew V Kossenkov, Karen DeWispeleare, Kyoung-Jae Won, Paul M Lieberman, Fang Lu, Horng-Shen Chen, Andrew V Kossenkov, Karen DeWispeleare, Kyoung-Jae Won, Paul M Lieberman

Abstract

Epstein-Barr Virus (EBV) transforms resting B-lymphocytes into proliferating lymphoblasts to establish latent infections that can give rise to malignancies. We show here that EBV-encoded transcriptional regulator EBNA2 drives the cooperative and combinatorial genome-wide binding of two master regulators of B-cell fate, namely EBF1 and RBP-jκ. Previous studies suggest that these B-cell factors are statically bound to target gene promoters. In contrast, we found that EBNA2 induces the formation of new binding for both RBP-jκ and EBF1, many of which are in close physical proximity in the cellular and viral genome. These newly induced binding sites co-occupied by EBNA2-EBF1-RBP-jκ correlate strongly with transcriptional activation of linked genes that are important for B-lymphoblast function. Conditional expression or repression of EBNA2 leads to a rapid alteration in RBP-jκ and EBF1 binding. Biochemical and shRNA depletion studies provide evidence for cooperative assembly at co-occupied sites. These findings reveal that EBNA2 facilitate combinatorial interactions to induce new patterns of transcription factor occupancy and gene programming necessary to drive B-lymphoblast growth and survival.

Conflict of interest statement

I have read the journal's policy and the authors of this manuscript have the following competing interests: PML is a founder of a company with interest in EBV associated disease. This does not alter our adherence to all PLOS policies on sharing data and materials.

Figures

Fig 1. Differential binding of transcription factors…
Fig 1. Differential binding of transcription factors on EBV genomes in type I and type III latency.
(A) ChIP-Seq tracks mapped to EBV genome in LCL (red) or Mutu I (blue) for EBF1 or RBP-jκ as indicated. EBNA2 ChIP-Seq for LCL (black). Y-axis represent raw read counts. (B) ChIP-qPCR for EBF1 or RBP-jκ in LCL (red) or Mutu I (blue) at various EBV genome regulatory regions as indicated and Actin genomic region as negative control. (C) Same as in B, except for isogenic EBV positive BL cells Kem III (red) or Kem I (blue). Asterisk indicates p < 0.05. (D) Western blot of Mutu I, LCL, Kem I, and Kem III probed with antibody to EBF1, RBP-jκ, LMP1, and EBNA2, with cellular loading controls for GAPDH and H3K4me3, as indicated. (E) RT-qPCR for EBV type III gene expression in LCL or Mutu I (left panel) or Kem I and Kem III (right panel).
Fig 2. Differential binding of transcription factors…
Fig 2. Differential binding of transcription factors on cellular genome in type I and type III infected B-lymphocytes.
(A) Venn diagram showing EBF1 and RBP-jκ occupancy sites specific to LCL (red) and Mutu I (blue) or common to both cell types. (B) Scatter plot showing the distribution of EBF1 (left) or RBP-jκ (right) occupancies that are specific to either LCL (L, red) or Mutu I (M, green) or common to both cell types (grey). (C) Heatmaps showing co-occupancy of EBF1 and RBP-jκ from either LCL (L) or Mutu I (M) cells, centered on either EBF1 in LCL (left panel) or RBP-jκ in LCL (right panel). Binding sites were clustered based on their common occurrence in MutuI and LCL (a), LCL-specific (b), or Mutu I-specific (c). (D and F) ChIP-qPCR validation for various cellular gene ChIP-Seq peaks for EBF1 (left) or RBP-jκ (right) in either Mutu I (blue) or LCL (red) cells. (E) Same as in (D) except for Kem I and Kem III cell types. (G) Same as in (F) except for Kem I and Kem III cell types. Asterisk indicates p < 0.05.
Fig 3. Colocalization of EBNA2 with RBP-jκ…
Fig 3. Colocalization of EBNA2 with RBP-jκ and EBF1 co-occupied sites.
(A) Heatmap of EBNA2 ChIP-Seq peaks from LCL was compared with EBNA2 peaks from Mutu III, EBF1 and RBP-jκ from LCL or Mutu I, EBNA3C from LCL or Mutu III, or EBNALP from LCL. Peaks were sorted based on the EBNA2 levels in LCL and show -/+ 4kb window. (B) The fold changes (LCL versus Mutu I) were plotted for EBF1 (x-axis) and RBP-jκ (y-axis) for all EBF1 and RBP-jκ peaks. The peaks were colored based on the EBNA2 occupancy. The positive correlation (Pearson’s correlation coefficient = 0.8) between the fold changes suggests that EBF1 and RBP-jκ co-localize with each other. The strong occupancy of EBNA2 at the top-right panel of the plot shows that high occupancy EBNA2 correlates with sites of high occupancies for EBF1 and RBP-jκ. (C) Heatmap comparison of histone modification marks (H3K4me3, H3Km4me1, H3K27me3) and transcription factors BATF, JunD, and EBNA2 from LCL centered at EBF1 peaks enriched in LCL (cluster i) or Mutu I (cluster ii). (D) Box plot quantitation of peak number and intensity for ChIP-Seq clustered sets i and ii shown in panel C as indicated. (E and F) Same as in C and D, except centered at cell-specific RBP-jκ peaks.
Fig 4. Characterization of cellular genes highly…
Fig 4. Characterization of cellular genes highly activated by EBF1-RBP-jκ-EBNA2 co-occupancy in LCL.
(A) Heatmap of mRNA expression for genes with co-localized EBF1, RBP-jκ, and EBNA2 and at least 3-fold over expression in LCL relative to Mutu I. (B) Percentage of genes with co-localized EBF1, RBP-jκ, and EBNA2 sites at least 10kb from the TSS that are over-expressed in LCLs relative to Mutu I. (C) Predicted regulation network for LCL over-expressed genes co-occupied by EBNA2, EBF1, and RBP-jκ.
Fig 5. EBNA2-dependent transcription factor redistribution in…
Fig 5. EBNA2-dependent transcription factor redistribution in chromatin occupancy.
EREB2.5 cells were treated with (+) or without (-) estradiol (E2) for 24 or 48 hrs and then assayed by ChIP for binding to EBF1 (A and B), RBP-jκ (C and D), or EBNA2 (E and F) at cellular (A, C, E) or EBV genome sites (B, D, F). Actin genomic region (cellular) or Qp (EBV) was used as negative binding control for EBF1, RBP-jκ, ορ EBNA2 ChIP. (G) ChIP binding for CTCF, PU.1, or PAX5 in EREB2.5 cells treated (blue) or untreated (red) with E2 for 48 hrs. PPP1R1B or KCTD17 genomic region was negative binding control PU.1 or PAX5, respectively. Asterisk indicates p < 0.05. (H) Western blot showing protein levels for EBF1, RBP-jκ, EBNA2, Actin, and CTCF in EREB2.5 cells at 24 and 48 hrs after E2 withdrawal.
Fig 6. Cooperative binding and colocalization of…
Fig 6. Cooperative binding and colocalization of EBF1, RBP-jκ, and EBNA2 at DNA regulatory elements.
(A) DNA-affinity binding assays with nuclear extracts from EREB2.5 cells with (+) or without (-) estradiol (E2) using DNA regulatory elements from IL7, HES1 (left), or LMP1, Qp (middle), or TOM1 (right). Input (10%) is indicated, and bound proteins were assayed by Western blot for EBF1, RBP-jκ, EBNA2, and specificity controls for PARP1 and Actin. (B) ChIP-reChIP assays in LCLs with EBF1 as first ChIP, followed by a reChIP with either no antibody control or antibody to EBF1, RBP-jκ or EBNA2. ReChIP DNA was quantified by qPCR at LMP1, IL7, and BDH2 (an EBF1 only site) binding sites. (C) Same as in B, except first ChIP was with RBP-jκ followed by a ReChIP with no antibody control, or EBF1, RBP-jκ, or EBNA2 antibody and assayed at LMP1, IL7, or LZTFL1 (an RBP-jκ only site) binding sites. Asterisk indicates p < 0.05.
Fig 7. EBF1 and RBP-jκ function at…
Fig 7. EBF1 and RBP-jκ function at EBNA2 co-occupied sites.
(A) Western blots of LCL cells transduced with either control shRNA (shCtrl) or shEBF1 (left) or shCtrl or shRBP-jκ (right) lentivirus and probed for EBF1, RBP-jκ LMP1, EBNA2, EBNA3C, or Actin. (B) Cell viability determined by Resazurin assay for LCL cells 7 days after shRNA lentivirus transduction. (C) RT-qPCR (ΔΔCT) analysis of EBV or cellular gene transcription (as indicated) in LCLs transduced with either shEBF1 (red), shRBP-jκ (green), or shCtrl lentivirus (black). (D-M) ChIP-qPCR in LCLs with antibodies to either EBF1 (panels D, E, J, K), RBP-jκ (panels F, G, H, I), or EBNA2 (panels L and M). LCLs were transduced with either shCtrl (black), shEBF1 (red), or shRBP-jκ (green). Viral (panels D, F, H, J, L), or cellular (E, G, I, K, M) sites were analyzed as indicated. Asterisk indicates p < 0.05.
Fig 8. Model of EBNA2-induced transcription factor…
Fig 8. Model of EBNA2-induced transcription factor redistribution in chromosome occupancy.
EBNA2 is shown to facilitate the formation of new binding sites that are co-occupied by RBP-jκ and EBF1 relative to cells lacking EBNA2.

References

    1. Longnecker R, Kieff E, Cohen JI. Epstein-Barr Virus In: Knipe DM, Howley PM, editors. Fields Virology. I. 6th ed. Philadelphia: Lipincott; 2013. p. 1898–959.
    1. Young LS, Rickinson AB. Epstein-Barr virus: 40 years on. Nature reviews Cancer. 2004;4(10):757–68.
    1. Thorley-Lawson DA, Hawkins JB, Tracy SI, Shapiro M. The pathogenesis of Epstein-Barr virus persistent infection. Current opinion in virology. 2013;3(3):227–32. 10.1016/j.coviro.2013.04.005
    1. Raab-Traub N. Novel mechanisms of EBV-induced oncogenesis. Current opinion in virology. 2012;2(4):453–8. 10.1016/j.coviro.2012.07.001
    1. Thorley-Lawson DA. Epstein-Barr virus: exploiting the immune system. Nature reviews Immunology. 2001;1(1):75–82.
    1. Kang MS, Kieff E. Epstein-Barr virus latent genes. Experimental & molecular medicine. 2015;47:e131.
    1. Hsieh JJ, Henkel T, Salmon P, Robey E, Peterson MG, Hayward SD. Truncated mammalian Notch1 activates CBF1/RBPJk-repressed genes by a mechanism resembling that of Epstein-Barr virus EBNA2. Molecular and cellular biology. 1996;16(3):952–9.
    1. Strobl LJ, Hofelmayr H, Stein C, Marschall G, Brielmeier M, Laux G, et al. Both Epstein-Barr viral nuclear antigen 2 (EBNA2) and activated Notch1 transactivate genes by interacting with the cellular protein RBP-J kappa. Immunobiology. 1997;198(1–3):299–306.
    1. Hayward SD, Liu J, Fujimuro M. Notch and Wnt signaling: mimicry and manipulation by gamma herpesviruses. Science's STKE: signal transduction knowledge environment. 2006;2006(335):re4
    1. Hayward SD. Viral interactions with the Notch pathway. Seminars in cancer biology. 2004;14(5):387–96.
    1. Borggrefe T, Oswald F. The Notch signaling pathway: transcriptional regulation at Notch target genes. Cellular and molecular life sciences: CMLS. 2009;66(10):1631–46. 10.1007/s00018-009-8668-7
    1. Portal D, Zhou H, Zhao B, Kharchenko PV, Lowry E, Wong L, et al. Epstein-Barr virus nuclear antigen leader protein localizes to promoters and enhancers with cell transcription factors and EBNA2. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(46):18537–42. 10.1073/pnas.1317608110
    1. Boller S, Grosschedl R. The regulatory network of B-cell differentiation: a focused view of early B-cell factor 1 function. Immunological reviews. 2014;261(1):102–15. 10.1111/imr.12206
    1. Zhou H, Schmidt SC, Jiang S, Willox B, Bernhardt K, Liang J, et al. Epstein-Barr virus oncoprotein super-enhancers control B cell growth. Cell host & microbe. 2015;17(2):205–16.
    1. Iwafuchi-Doi M, Zaret KS. Pioneer transcription factors in cell reprogramming. Genes & development. 2014;28(24):2679–92.
    1. Rothenberg EV. Transcriptional control of early T and B cell developmental choices. Annual review of immunology. 2014;32:283–321. 10.1146/annurev-immunol-032712-100024
    1. Tempera I, Klichinsky M, Lieberman PM. EBV latency types adopt alternative chromatin conformations. PLoS pathogens. 2011;7(7):e1002180 10.1371/journal.ppat.1002180
    1. Zhao B, Zou J, Wang H, Johannsen E, Peng CW, Quackenbush J, et al. Epstein-Barr virus exploits intrinsic B-lymphocyte transcription programs to achieve immortal cell growth. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(36):14902–7. 10.1073/pnas.1108892108
    1. McClellan MJ, Wood CD, Ojeniyi O, Cooper TJ, Kanhere A, Arvey A, et al. Modulation of enhancer looping and differential gene targeting by Epstein-Barr virus transcription factors directs cellular reprogramming. PLoS pathogens. 2013;9(9):e1003636 10.1371/journal.ppat.1003636
    1. Jiang S, Willox B, Zhou H, Holthaus AM, Wang A, Shi TT, et al. Epstein-Barr virus nuclear antigen 3C binds to BATF/IRF4 or SPI1/IRF4 composite sites and recruits Sin3A to repress CDKN2A. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(1):421–6. 10.1073/pnas.1321704111
    1. Consortium EP, Bernstein BE, Birney E, Dunham I, Green ED, Gunter C, et al. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57–74. Epub 2012/09/08. 10.1038/nature11247
    1. Calo E, Wysocka J. Modification of enhancer chromatin: what, how, and why? Molecular cell. 2013;49(5):825–37. 10.1016/j.molcel.2013.01.038
    1. Kempkes B, Spitkovsky D, Jansen-Durr P, Ellwart JW, Kremmer E, Delecluse HJ, et al. B-cell proliferation and induction of early G1-regulating proteins by Epstein-Barr virus mutants conditional for EBNA2. The EMBO journal. 1995;14(1):88–96.
    1. Tempera I, Lieberman PM. Epigenetic regulation of EBV persistence and oncogenesis. Seminars in cancer biology. 2014;26:22–9. 10.1016/j.semcancer.2014.01.003
    1. Trivedi P, Spinsanti P, Cuomo L, Volpe M, Takada K, Frati L, et al. Differential regulation of Epstein-Barr virus (EBV) latent gene expression in Burkitt lymphoma cells infected with a recombinant EBV strain. Journal of virology. 2001;75(10):4929–35.
    1. Wang H, Zou J, Zhao B, Johannsen E, Ashworth T, Wong H, et al. Genome-wide analysis reveals conserved and divergent features of Notch1/RBPJ binding in human and murine T-lymphoblastic leukemia cells. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(36):14908–13. 10.1073/pnas.1109023108
    1. Schmidt SC, Jiang S, Zhou H, Willox B, Holthaus AM, Kharchenko PV, et al. Epstein-Barr virus nuclear antigen 3A partially coincides with EBNA3C genome-wide and is tethered to DNA through BATF complexes. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(2):554–9. 10.1073/pnas.1422580112
    1. Harth-Hertle ML, Scholz BA, Erhard F, Glaser LV, Dolken L, Zimmer R, et al. Inactivation of intergenic enhancers by EBNA3A initiates and maintains polycomb signatures across a chromatin domain encoding CXCL10 and CXCL9. PLoS pathogens. 2013;9(9):e1003638 10.1371/journal.ppat.1003638
    1. Tzellos S, Correia PB, Karstegl CE, Cancian L, Cano-Flanagan J, McClellan MJ, et al. A single amino acid in EBNA-2 determines superior B lymphoblastoid cell line growth maintenance by Epstein-Barr virus type 1 EBNA-2. Journal of virology. 2014;88(16):8743–53. 10.1128/JVI.01000-14
    1. Ohashi M, Holthaus AM, Calderwood MA, Lai CY, Krastins B, Sarracino D, et al. The EBNA3 Family of Epstein-Barr Virus Nuclear Proteins Associates with the USP46/USP12 Deubiquitination Complexes to Regulate Lymphoblastoid Cell Line Growth. PLoS pathogens. 2015;11(4):e1004822 10.1371/journal.ppat.1004822
    1. Miele L. Transcription factor RBPJ/CSL: a genome-wide look at transcriptional regulation. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(36):14715–6. 10.1073/pnas.1110570108
    1. Yashiro-Ohtani Y, Wang H, Zang C, Arnett KL, Bailis W, Ho Y, et al. Long-range enhancer activity determines Myc sensitivity to Notch inhibitors in T cell leukemia. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(46):E4946–53. 10.1073/pnas.1407079111
    1. Wang H, Zang C, Taing L, Arnett KL, Wong YJ, Pear WS, et al. NOTCH1-RBPJ complexes drive target gene expression through dynamic interactions with superenhancers. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(2):705–10. 10.1073/pnas.1315023111
    1. Castel D, Mourikis P, Bartels SJ, Brinkman AB, Tajbakhsh S, Stunnenberg HG. Dynamic binding of RBPJ is determined by Notch signaling status. Genes & development. 2013;27(9):1059–71.
    1. Rowe M, Raithatha S, Shannon-Lowe C. Counteracting effects of cellular Notch and Epstein-Barr virus EBNA2: implications for stromal effects on virus-host interactions. Journal of virology. 2014;88(20):12065–76. 10.1128/JVI.01431-14
    1. Treiber T, Mandel EM, Pott S, Gyory I, Firner S, Liu ET, et al. Early B cell factor 1 regulates B cell gene networks by activation, repression, and transcription- independent poising of chromatin. Immunity. 2010;32(5):714–25. 10.1016/j.immuni.2010.04.013
    1. Gyory I, Boller S, Nechanitzky R, Mandel E, Pott S, Liu E, et al. Transcription factor Ebf1 regulates differentiation stage-specific signaling, proliferation, and survival of B cells. Genes & development. 2012;26(7):668–82.
    1. Guilhamon P, Eskandarpour M, Halai D, Wilson GA, Feber A, Teschendorff AE, et al. Meta-analysis of IDH-mutant cancers identifies EBF1 as an interaction partner for TET2. Nature communications. 2013;4:2166 10.1038/ncomms3166
    1. Bossen C, Murre CS, Chang AN, Mansson R, Rodewald HR, Murre C. The chromatin remodeler Brg1 activates enhancer repertoires to establish B cell identity and modulate cell growth. Nature immunology. 2015.
    1. Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Molecular cell. 2010;38(4):576–89. 10.1016/j.molcel.2010.05.004
    1. Tanigaki K, Han H, Yamamoto N, Tashiro K, Ikegawa M, Kuroda K, et al. Notch-RBP-J signaling is involved in cell fate determination of marginal zone B cells. Nature immunology. 2002;3(5):443–50.
    1. Lee TI, Johnstone SE, Young RA. Chromatin immunoprecipitation and microarray-based analysis of protein location. Nature protocols. 2006;1(2):729–48.
    1. Chau CM, Lieberman PM. Dynamic chromatin boundaries delineate a latency control region of Epstein-Barr virus. Journal of virology. 2004;78(22):12308–19.
    1. Moss T, Leblanc B. DNA-protein interactions principles and protocols. New York: Humana,; 2009. Available from: Connect to full text.
    1. Lu F, Day L, Gao SJ, Lieberman PM. Acetylation of the latency-associated nuclear antigen regulates repression of Kaposi's sarcoma-associated herpesvirus lytic transcription. Journal of virology. 2006;80(11):5273–82.
    1. Atanasiu C, Lezina L, Lieberman PM. DNA affinity purification of Epstein-Barr virus OriP-binding proteins. Methods Mol Biol. 2005;292:267–76.
    1. Deng Z, Wang Z, Stong N, Plasschaert R, Moczan A, Chen HS, et al. A role for CTCF and cohesin in subtelomere chromatin organization, TERRA transcription, and telomere end protection. The EMBO journal. 2012;31(21):4165–78. 10.1038/emboj.2012.266
    1. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome biology. 2009;10(3):R25 Epub 2009/03/06. 10.1186/gb-2009-10-3-r25
    1. The ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57–74. . 10.1038/nature11247
    1. Storey JD, Dai JY, Leek JT. The optimal discovery procedure for large-scale significance testing, with applications to comparative microarray experiments. Biostatistics. 2007;8(2):414–32.

Source: PubMed

3
Abonnieren