Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

BRD4 assists elongation of both coding and enhancer RNAs by interacting with acetylated histones

Abstract

Small-molecule BET inhibitors interfere with the epigenetic interactions between acetylated histones and the bromodomains of the BET family proteins, including BRD4, and they potently inhibit growth of malignant cells by targeting cancer-promoting genes. BRD4 interacts with the pause-release factor P-TEFb and has been proposed to release RNA polymerase II (Pol II) from promoter-proximal pausing. We show that BRD4 occupies widespread genomic regions in mouse cells and directly stimulates elongation of both protein-coding transcripts and noncoding enhancer RNAs (eRNAs), in a manner dependent on bromodomain function. BRD4 interacts with elongating Pol II complexes and assists Pol II in progression through hyperacetylated nucleosomes by interacting with acetylated histones via bromodomains. On active enhancers, the BET inhibitor JQ1 antagonizes BRD4-associated eRNA synthesis. Thus, BRD4 is involved in multiple steps of the transcription hierarchy, primarily by facilitating transcript elongation both at enhancers and on gene bodies independently of P-TEFb.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: BET inhibitor JQ1 inhibits both initiation and elongation of protein-coding RNA.
Figure 2: BET inhibitor JQ1 antagonizes BRD4 and inhibits eRNA synthesis.
Figure 3: BRD4-dependent gene expression correlates with BRD4 enrichment in intergenic regions and on gene bodies but not at TSSs.
Figure 4: BD-mediated interaction with acetylated histones is required for BRD4-dependent expression of Myc and Klf4 but not for the recruitment of BRD4 and P-TEFb at TSSs.
Figure 5: BRD4 possesses a histone-chaperone activity and assists elongating Pol II in transcription through hyperacetylated nucleosomes in vitro.
Figure 6: BRD4 facilitates transcript elongation in a BD-dependent manner in vivo.
Figure 7: BRD4 physically interacts with Pol II elongation complexes in vivo.
Figure 8: A model depicting the interdependence between the passage of Pol II elongation complexes through nucleosomes and acetyl-histone binding of BRD4 at enhancers and within protein-coding genes.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Wang, Z. et al. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat. Genet. 40, 897–903 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. Shen, Y. et al. A map of the cis-regulatory sequences in the mouse genome. Nature 488, 116–120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wang, Z. et al. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 138, 1019–1031 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wu, S.Y. & Chiang, C.M. The double bromodomain-containing chromatin adaptor Brd4 and transcriptional regulation. J. Biol. Chem. 282, 13141–13145 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Filippakopoulos, P. et al. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell 149, 214–231 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kanno, T. et al. Selective recognition of acetylated histones by bromodomain proteins visualized in living cells. Mol. Cell 13, 33–43 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Jung, M. et al. Affinity map of bromodomain protein 4 (BRD4) interactions with the histone H4 tail and the small molecule inhibitor JQ1. J. Biol. Chem. 289, 9304–9319 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Shi, J. et al. Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell 25, 210–225 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Asangani, I.A. et al. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature 510, 278–282 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. LeRoy, G., Rickards, B. & Flint, S.J. The double bromodomain proteins Brd2 and Brd3 couple histone acetylation to transcription. Mol. Cell 30, 51–60 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mochizuki, K. et al. The bromodomain protein Brd4 stimulates G1 gene transcription and promotes progression to S phase. J. Biol. Chem. 283, 9040–9048 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Nicodeme, E. et al. Suppression of inflammation by a synthetic histone mimic. Nature 468, 1119–1123 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Delmore, J.E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524–528 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dawson, M.A. et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529–533 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lockwood, W.W., Zejnullahu, K., Bradner, J.E. & Varmus, H. Sensitivity of human lung adenocarcinoma cell lines to targeted inhibition of BET epigenetic signaling proteins. Proc. Natl. Acad. Sci. USA 109, 19408–19413 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chapuy, B. et al. Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma. Cancer Cell 24, 777–790 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ceribelli, M. et al. Blockade of oncogenic IκB kinase activity in diffuse large B-cell lymphoma by bromodomain and extraterminal domain protein inhibitors. Proc. Natl. Acad. Sci. USA 111, 11365–11370 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bandukwala, H.S. et al. Selective inhibition of CD4+ T-cell cytokine production and autoimmunity by BET protein and c-Myc inhibitors. Proc. Natl. Acad. Sci. USA 109, 14532–14537 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Matzuk, M.M. et al. Small-molecule inhibition of BRDT for male contraception. Cell 150, 673–684 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Anand, P. et al. BET bromodomains mediate transcriptional pause release in heart failure. Cell 154, 569–582 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bhatt, D.M. et al. Transcript dynamics of proinflammatory genes revealed by sequence analysis of subcellular RNA fractions. Cell 150, 279–290 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hargreaves, D.C., Horng, T. & Medzhitov, R. Control of inducible gene expression by signal-dependent transcriptional elongation. Cell 138, 129–145 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rahl, P.B. et al. c-Myc regulates transcriptional pause release. Cell 141, 432–445 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Buratowski, S. Progression through the RNA polymerase II CTD cycle. Mol. Cell 36, 541–546 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Adelman, K. & Lis, J.T. Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans. Nat. Rev. Genet. 13, 720–731 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Peterlin, B.M. & Price, D.H. Controlling the elongation phase of transcription with P-TEFb. Mol. Cell 23, 297–305 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Lin, C. et al. AFF4, a component of the ELL/P-TEFb elongation complex and a shared subunit of MLL chimeras, can link transcription elongation to leukemia. Mol. Cell 37, 429–437 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. He, N. et al. HIV-1 Tat and host AFF4 recruit two transcription elongation factors into a bifunctional complex for coordinated activation of HIV-1 transcription. Mol. Cell 38, 428–438 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Donner, A.J., Ebmeier, C.C., Taatjes, D.J. & Espinosa, J.M. CDK8 is a positive regulator of transcriptional elongation within the serum response network. Nat. Struct. Mol. Biol. 17, 194–201 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Eberhardy, S.R. & Farnham, P.J. Myc recruits P-TEFb to mediate the final step in the transcriptional activation of the cad promoter. J. Biol. Chem. 277, 40156–40162 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Barboric, M., Nissen, R.M., Kanazawa, S., Jabrane-Ferrat, N. & Peterlin, B.M. NF-κB binds P-TEFb to stimulate transcriptional elongation by RNA polymerase II. Mol. Cell 8, 327–337 (2001).

    Article  CAS  PubMed  Google Scholar 

  35. Jang, M.K. et al. The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Mol. Cell 19, 523–534 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Yang, Z. et al. Recruitment of P-TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol. Cell 19, 535–545 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Bisgrove, D.A., Mahmoudi, T., Henklein, P. & Verdin, E. Conserved P-TEFb-interacting domain of BRD4 inhibits HIV transcription. Proc. Natl. Acad. Sci. USA 104, 13690–13695 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Petesch, S.J. & Lis, J.T. Overcoming the nucleosome barrier during transcript elongation. Trends Genet. 28, 285–294 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Belotserkovskaya, R. et al. FACT facilitates transcription-dependent nucleosome alteration. Science 301, 1090–1093 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Reinberg, D. & Sims, R.J. III. de FACTo nucleosome dynamics. J. Biol. Chem. 281, 23297–23301 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Vahedi, G. et al. STATs shape the active enhancer landscape of T cell populations. Cell 151, 981–993 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Whyte, W.A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kieffer-Kwon, K.R. et al. Interactome maps of mouse gene regulatory domains reveal basic principles of transcriptional regulation. Cell 155, 1507–1520 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Andersson, R. et al. An atlas of active enhancers across human cell types and tissues. Nature 507, 455–461 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li, W. et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498, 516–520 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lam, M.T. et al. Rev-Erbs repress macrophage gene expression by inhibiting enhancer-directed transcription. Nature 498, 511–515 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kim, T.K. et al. Widespread transcription at neuronal activity-regulated enhancers. Nature 465, 182–187 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Mousavi, K. et al. eRNAs promote transcription by establishing chromatin accessibility at defined genomic loci. Mol. Cell 51, 606–617 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Patel, M.C. et al. BRD4 coordinates recruitment of pause release factor P-TEFb and the pausing complex NELF/DSIF to regulate transcription elongation of interferon-stimulated genes. Mol. Cell. Biol. 33, 2497–2507 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Valouev, A. et al. Determinants of nucleosome organization in primary human cells. Nature 474, 516–520 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ramirez-Carrozzi, V.R. et al. A unifying model for the selective regulation of inducible transcription by CpG islands and nucleosome remodeling. Cell 138, 114–128 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang, W. et al. Bromodomain-containing protein 4 (BRD4) regulates RNA polymerase II serine 2 phosphorylation in human CD4+ T cells. J. Biol. Chem. 287, 43137–43155 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lovén, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Jiang, Y.W. et al. Mammalian mediator of transcriptional regulation and its possible role as an end-point of signal transduction pathways. Proc. Natl. Acad. Sci. USA 95, 8538–8543 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Liu, W. et al. Brd4 and JMJD6-associated anti-pause enhancers in regulation of transcriptional pause release. Cell 155, 1581–1595 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Devaiah, B.N. et al. BRD4 is an atypical kinase that phosphorylates serine2 of the RNA polymerase II carboxy-terminal domain. Proc. Natl. Acad. Sci. USA 109, 6927–6932 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ottinger, M. et al. Kaposi's sarcoma-associated herpesvirus LANA-1 interacts with the short variant of BRD4 and releases cells from a BRD4- and BRD2/RING3-induced G1 cell cycle arrest. J. Virol. 80, 10772–10786 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Floyd, S.R. et al. The bromodomain protein Brd4 insulates chromatin from DNA damage signalling. Nature 498, 246–250 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. LeRoy, G. et al. Proteogenomic characterization and mapping of nucleosomes decoded by Brd and HP1 proteins. Genome Biol. 13, R68 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Nagarajan, S. et al. Bromodomain protein BRD4 is required for estrogen receptor-dependent enhancer activation and gene transcription. Cell Reports 8, 460–469 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. Dey, A. et al. A bromodomain protein, MCAP, associates with mitotic chromosomes and affects G2-to-M transition. Mol. Cell. Biol. 20, 6537–6549 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Stein, A. & Mitchell, M. Generation of different nucleosome spacing periodicities in vitro: possible origin of cell type specificity. J. Mol. Biol. 203, 1029–1043 (1988).

    Article  CAS  PubMed  Google Scholar 

  63. LeRoy, G. et al. A quantitative atlas of histone modification signatures from human cancer cells. Epigenetics Chromatin 6, 20 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. LeRoy, G., Orphanides, G., Lane, W.S. & Reinberg, D. Requirement of RSF and FACT for transcription of chromatin templates in vitro. Science 282, 1900–1904 (1998).

    Article  CAS  PubMed  Google Scholar 

  65. LeRoy, G., Drapkin, R., Weis, L. & Reinberg, D. Immunoaffinity purification of the human multisubunit transcription factor IIH. J. Biol. Chem. 273, 7134–7140 (1998).

    Article  CAS  PubMed  Google Scholar 

  66. Maldonado, E., Drapkin, R. & Reinberg, D. Purification of human RNA polymerase II and general transcription factors. Methods Enzymol. 274, 72–100 (1996).

    Article  CAS  PubMed  Google Scholar 

  67. Orphanides, G., LeRoy, G., Chang, C.H., Luse, D.S. & Reinberg, D. FACT, a factor that facilitates transcript elongation through nucleosomes. Cell 92, 105–116 (1998).

    Article  CAS  PubMed  Google Scholar 

  68. Schnitzler, G.R. Isolation of histones and nucleosome cores from mammalian cells. Curr. Protoc. Mol. Biol. 50, 21.5 (2001).

    Google Scholar 

  69. Wei, L. et al. Discrete roles of STAT4 and STAT6 transcription factors in tuning epigenetic modifications and transcription during T helper cell differentiation. Immunity 32, 840–851 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Zang, C. et al. A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 25, 1952–1958 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Core, L.J., Waterfall, J.J. & Lis, J.T. Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science 322, 1845–1848 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Quinlan, A.R. & Hall, I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Larschan, E. et al. X chromosome dosage compensation via enhanced transcriptional elongation in Drosophila. Nature 471, 115–118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank L.J. Core and J.T. Lis (Cornell University) for the global run-on sequencing (GRO-seq) protocol, V. Sartorelli and D. Clark for critical discussion and reading of the manuscript and J. Kadonaga (University of California, San Diego) for ND423. This work was supported by the Intramural Research Programs of the US National Institutes of Health NICHD, NIAMS and NIAID.

Author information

Authors and Affiliations

Authors

Contributions

T.K. conceived the study, designed experiments, performed all experiments except supercoiling assays and in vitro transcription analysis, interpreted all data and wrote and handled the manuscript as the corresponding author. Y.K. performed RNA-seq, GRO-seq and ChIP-seq, interpreted data and wrote the manuscript. G.L., E.C. and B.A.G. conceived, designed and performed in vitro transcription analysis and plasmid supercoiling assays. H.-W.S., S.R.B. and G.V. performed genome-wide data processing. T.D.H. oversaw the JQ1 experiment. D.R., U.S., J.J.O. and K.O. discussed the results and edited the manuscript.

Corresponding authors

Correspondence to Tomohiko Kanno, John J O'Shea or Keiko Ozato.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 The effects of BET inhibitor JQ1 on serum-induced nascent transcripts of protein-coding genes and flanking intergenic regions.

a, Metagene profiles of 66 genes larger than 2 kb, serum induction of which was inhibited by JQ1. (1) 0.5% + vehicle: cells were serum-starved, preincubated with vehicle, and left unstimulated. (2) 0.5% + JQ1: cells were serum-starved, preincubated with JQ1, and left unstimulated. (3) Serum + vehicle: cells were serum-starved, preincubated with vehicle, and then serum-stimulated. (4) 0.5% + JQ1: cells were serum-starved, preincubated with JQ1, and then serum-stimulated. For each gene, the sum of chromatin RNA-seq read-counts from 200 nt upstream of TSS to TES in cells treated with 0.5%+vehicle was calculated, and used to normalize read-counts in cells under all four experimental conditions. b, Genome browser views of the 3' enhancer regions of the Trib1 gene, characterized by chromatin RNA-seq, and the chromatin association of Pol II, BRD4, P-TEFb (CDK9), H3K27Ac, and H4Ac. For RNA-seq, sense- and antisense-strand signals are shown in upward and downward directions, respectively. Two regions exhibiting a prominent accumulation of ChIP signals as well as RNA-seq signals after serum stimulation are magnified in the lower part. JQ1 treatment mostly inhibited elongation of the non-coding transcripts without affecting acetylation of H3K27 and H4. Progression of Pol II and BRD4 were reduced by JQ1, but that of CDK9 was less affected. c, Genome browser views along the Dot1l gene and the upstream flanking region.

Supplementary Figure 2 Scatter plots and graphical displays of rank correlation.

Supplementary to Fig. 3 panels b, d, e, h, j and k. For each panel, scatter plots of measured values, and correlation displays of ranks are shown. For the displays of rank correlation, moving averages were calculated according to the order of BRD4 or CDK9. The subset sizes were 50 for panels b, d, and j, 75 for panel k, 100 for panel e, and 200 for panel h. The median points of the ranks of the y axis are represented by the dotted lines. Spearman’s ρ values and p-values are also shown. Note: these displays do not contain the lowest ranks on the x-axis (BRD4 or CDK9), because there were too many data points belonging to these ranks for the calculations.

Supplementary Figure 3 Live-cell fluorescence resonance energy transfer (FRET) analysis of interactions between YFP-histones and CFP-BRD proteins.

a, Diagram of CFP-BRD proteins. b, Three-dimensional (3D) surface plots of FRET signals from HEK293T cells transfected with YFP-H4 alone (i) or CFP-BRD4short alone (ii). In flow cytometric measurements, spillover signals (e.g., false FRET signals due to spillover of CFP signals into the FRET detector channel) were digitally compensated so that YFP- or CFP-protein alone exhibited only marginal FRET signals. c, 3D surface plots displaying FRET signals covering wide ranges in CFP/YFP protein expression. HEK293T cells were transfected with YFP-H4 and CFP-BRD proteins as indicated. The same spillover-compensation matrix set in b was applied to all plots. d, Cells were transfected with expression plasmids for CFP-BRD proteins and YFP-histones or an empty vector in combinations as indicated. Data were collected using a fixed window set for similar levels of YFP-proteins, and FRET signals (means ± SEM) were plotted against CFP signals. For cells transfected with CFP-BRD plasmids and an empty vector, all data were collected.

Supplementary Figure 4 BRD4 knockdown and reconstitution system.

a, Diagram of YFP-BRD4, YFP-BRD4-mBD, and their short forms. These BRD4 constructs contain nucleotide substitutions around the target site of BRD4 shRNA (marked by an asterisk) so that they are resistant to BRD4 shRNA while keeping the amino-acid coding unchanged. b, Transient transcription reporter assays using HIV-LTR-Luc and minP-Luc (minimal promoter Luc) luciferase reporters. HIV-LTR-Luc is sensitive to P-TEFb activity, while minP-Luc is not. Of note, low levels of P-TEFb-independent luciferase activities were observed both with BRD4 and BRD4short on minP-Luc, which were comparable with those observed with BRD4short on HIV-LTR-Luc. c, Work flow to establish cells stably expressing YFP-BRD4 or YFP-BRD4-mBD (or their short forms) in place of endogenous BRD4. NIH3T3 cells were sequentially infected with shRNA retrovirus and YFP-BRD4 expressing retrovirus as shown. The transduced cells were selected with G418 and puromycin. d, Left panel: immunoblot with anti-BRD4 antibody. The positions of YFP-tagged BRD4 proteins and endogenous BRD4 are shown. Right panel: immunoblot with anti-BRD4 antibody for endogenous BRD4, and with anti-YFP (GFP) antibody for YFP-BRD4short proteins. e, Microarray gene expression analysis of the recovery of BRD4-dependent genes after BRD4 shRNA-mediated knockdown. Paired differences in the recovery ratios (RRs) of 410 BRD4 target genes are binned and plotted as in Fig.3g. In the right, BRD4 target genes are classified by gene ontology. In parenthesis shown are the numbers of genes in each group and P values of paired t test on the RR compared between the YFP-BRD4 and YFP-BRD4-mBD reconstituted cells, all of which are statistically significant. Bars represent means ± s.e.m. of the RRs.

Supplementary Figure 5 ChIP assays to detect binding of BRD4, CDK9 and Pol II over the Myc gene loci.

a, Effects of BRD4 knockdown by BRD4 shRNA on BRD4 protein and Myc transcript levels in NIH3T3 cells. b, Genome browser views of ChIP-seq signals for BRD4, CDK9 and Pol II (pan) along the Myc gene in NIH3T3 cells treated as indicated on the left. c, BRD4 knockdown cells were reconstituted with YFP-BRD4, YFP-BRD4-mBD or YFP alone. ChIP-qPCR assays using anti-YFP (GFP) antibody demonstrate binding of YFP-BRD4 or YFP-BRD4-mBD on the Myc gene locus. Signals relative to input DNA (y-axis) are plotted against positions of PCR primers (x-axis: covering the same region as the Myc gene locus shown at the bottom). In the gene locus diagram, dotted lines represent the upstream and downstream regions flanking the Myc gene. Background signals were assessed with cells expressing YFP alone. (d, e) ChIP-qPCR assays demonstrating binding of BRD4 (d) and CDK9 (e) on the Myc gene locus (shown at the bottom) in cells treated with JQ1 (1 μM, 2 h) or vehicle (DMSO). Background signals were assessed using normal rabbit IgG.

Supplementary Figure 6 In vitro transcription assays on chromatin templates.

a, Diagram showing protocol for the in vitro transcription assays on chromatin templates. In vitro transcription elongation assays were performed on a hypo-acetylated or hyper-acetylated chromatin pG5MLP template, using recombinant FACT or BRD4; together with general transcription factors (GTFs) and RNA polymerase II. b, Relative quantification of histone H3 modifications by quantitative mass spectrometry in hypo-acetylated and hyper-acetylated nucleosomes. Results of two independent measurements are shown.

Supplementary Figure 7 BRD4short facilitates progression of Pol II and transcript elongation on the Myc gene by binding to acetylated histones through bromodomains in vivo.

a, Genome browser views of nascent transcript progression across the Myc gene locus, measured by chromatin RNA-seq analysis. b, BrU-labeled nuclear run-on RNA signals (measured by qRT-PCR) aligned along the Myc gene locus. Nuclei were isolated from BRD4 knockdown cells reconstituted with YFP-BRD4short, YFP-BRD4short-mBD or YFP alone, and subjected to a nuclear run-on reaction in the presence of 1% sarkosyl, and BrUTP, ATP, CTP, and GTP. The run-on RNAs labeled with BrU were fragmented, immunoprecipitated with anti-BrdU antibody, and analyzed by qRT-PCR using Myc locus specific primers. PCR efficiency was normalized with genomic DNA, and relative values of the run-on transcript signals are plotted against genomic positions corresponding to the PCR primers. (c, d) ChIP-qPCR assays for Ser2 phosphorylated Pol II (c) or K9/K14 acetylated H3 (d) along the Myc gene locus.

Supplementary Figure 8 BRD4 is associated with Pol II elongation complexes in vivo.

a, Validation of native RIP (RNA-Immunoprecipitation). Native nuclear complexes containing YFP-BRD4short and RNA were released by DNaseI digestion, and immunoprecipitated with anti-YFP antibody (for BRD4short) or control IgG, followed by extraction of co-precipitated RNA. Quantitative PCR using Klf4 specific primers was performed with or without reverse transcription (RT). b, Quantitative RT-PCR analysis of the RunOn-RIP signals along the Myc gene locus. Background signals were assessed using normal rabbit IgG. c, Genome browser view of RunOn-RIP-seq reads around the Myc gene locus, representing the actively elongating front of transcripts that was associated with YFP-BRD4short.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 4535 kb)

Supplementary Data Set 1

Uncropped images of blots (PDF 1865 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kanno, T., Kanno, Y., LeRoy, G. et al. BRD4 assists elongation of both coding and enhancer RNAs by interacting with acetylated histones. Nat Struct Mol Biol 21, 1047–1057 (2014). https://doi.org/10.1038/nsmb.2912

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2912

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer