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:

R loops regulate promoter-proximal chromatin architecture and cellular differentiation

Abstract

Numerous chromatin-remodeling factors are regulated by interactions with RNA, although the contexts and functions of RNA binding are poorly understood. Here we show that R loops, RNA-DNA hybrids consisting of nascent transcripts hybridized to template DNA, modulate the binding of two key chromatin-regulatory complexes, Tip60–p400 and polycomb repressive complex 2 (PRC2) in mouse embryonic stem cells (ESCs). Like PRC2, the Tip60–p400 histone acetyltransferase complex binds to nascent transcripts; however, transcription promotes chromatin binding of Tip60–p400 but not PRC2. Interestingly, we observed higher Tip60–p400 and lower PRC2 levels at genes marked by promoter-proximal R loops. Furthermore, disruption of R loops broadly decreased Tip60–p400 occupancy and increased PRC2 occupancy genome wide. In agreement with these alterations, ESCs partially depleted of R loops exhibited impaired differentiation. These results show that R loops act both positively and negatively in modulating the recruitment of key pluripotency regulators.

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

Access options

Buy this article

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

Figure 1: Tip60–p400 binds nascent transcripts.
Figure 2: Transcription promotes promoter-proximal association of Tip60–p400.
Figure 3: Promoter-proximal R loops colocalize with Tip60–p400.
Figure 4: R loops promote elevated Tip60 and p400 binding to many target genes.
Figure 5: R loops inhibit chromatin binding by PRC2.
Figure 6: Disruption of R loops impairs ESC differentiation.
Figure 7: Model of R-loop function.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

NCBI Reference Sequence

References

  1. Bonasio, R. & Shiekhattar, R. Regulation of transcription by long noncoding RNAs. Annu. Rev. Genet. 48, 433–455 (2014).

    Article  CAS  Google Scholar 

  2. Rinn, J.L. & Chang, H.Y. Genome regulation by long noncoding RNAs. Annu. Rev. Biochem. 81, 145–166 (2012).

    Article  CAS  Google Scholar 

  3. Flynn, R.A. & Chang, H.Y. Long noncoding RNAs in cell-fate programming and reprogramming. Cell Stem Cell 14, 752–761 (2014).

    Article  CAS  Google Scholar 

  4. Guttman, M. et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 477, 295–300 (2011).

    Article  CAS  Google Scholar 

  5. Rinn, J.L. et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129, 1311–1323 (2007).

    Article  CAS  Google Scholar 

  6. Zhao, J., Sun, B.K., Erwin, J.A., Song, J.-J. & Lee, J.T. Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science 322, 750–756 (2008).

    Article  CAS  Google Scholar 

  7. Pandey, R.R. et al. Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol. Cell 32, 232–246 (2008).

    Article  CAS  Google Scholar 

  8. Aloia, L., Di Stefano, B. & Di Croce, L. Polycomb complexes in stem cells and embryonic development. Development 140, 2525–2534 (2013).

    Article  CAS  Google Scholar 

  9. Cifuentes-Rojas, C., Hernandez, A.J., Sarma, K. & Lee, J.T. Regulatory interactions between RNA and polycomb repressive complex 2. Mol. Cell 55, 171–185 (2014).

    Article  CAS  Google Scholar 

  10. Kaneko, S., Son, J., Shen, S.S., Reinberg, D. & Bonasio, R. PRC2 binds active promoters and contacts nascent RNAs in embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1258–1264 (2013).

    Article  CAS  Google Scholar 

  11. Davidovich, C., Zheng, L., Goodrich, K.J. & Cech, T.R. Promiscuous RNA binding by Polycomb repressive complex 2. Nat. Struct. Mol. Biol. 20, 1250–1257 (2013).

    Article  CAS  Google Scholar 

  12. Kaneko, S., Son, J., Bonasio, R., Shen, S.S. & Reinberg, D. Nascent RNA interaction keeps PRC2 activity poised and in check. Genes Dev. 28, 1983–1988 (2014).

    Article  CAS  Google Scholar 

  13. Riising, E.M. et al. Gene silencing triggers polycomb repressive complex 2 recruitment to CpG islands genome wide. Mol. Cell 55, 347–360 (2014).

    Article  CAS  Google Scholar 

  14. Squatrito, M., Gorrini, C. & Amati, B. Tip60 in DNA damage response and growth control: many tricks in one HAT. Trends Cell Biol. 16, 433–442 (2006).

    Article  CAS  Google Scholar 

  15. Fazzio, T.G., Huff, J.T. & Panning, B. An RNAi screen of chromatin proteins identifies Tip60-p400 as a regulator of embryonic stem cell identity. Cell 134, 162–174 (2008).

    Article  CAS  Google Scholar 

  16. Chen, P.B. et al. Hdac6 regulates Tip60-p400 function in stem cells. eLife 2, e01557 (2013).

    Article  Google Scholar 

  17. Ginno, P.A., Lott, P.L., Christensen, H.C., Korf, I. & Chédin, F. R-loop formation is a distinctive characteristic of unmethylated human CpG island promoters. Mol. Cell 45, 814–825 (2012).

    Article  CAS  Google Scholar 

  18. Ginno, P.A., Lim, Y.W., Lott, P.L., Korf, I. & Chédin, F. GC skew at the 5' and 3′ ends of human genes links R-loop formation to epigenetic regulation and transcription termination. Genome Res. 23, 1590–1600 (2013).

    Article  CAS  Google Scholar 

  19. Davidovich, C. et al. Toward a consensus on the binding specificity and promiscuity of PRC2 for RNA. Mol. Cell 57, 552–558 (2015).

    Article  CAS  Google Scholar 

  20. Sun, Q., Csorba, T., Skourti-Stathaki, K., Proudfoot, N.J. & Dean, C. R-loop stabilization represses antisense transcription at the Arabidopsis FLC locus. Science 340, 619–621 (2013).

    Article  CAS  Google Scholar 

  21. Skourti-Stathaki, K., Kamieniarz-Gdula, K. & Proudfoot, N.J. R-loops induce repressive chromatin marks over mammalian gene terminators. Nature 516, 436–439 (2014).

    Article  CAS  Google Scholar 

  22. Castel, S.E. et al. Dicer promotes transcription termination at sites of replication stress to maintain genome stability. Cell 159, 572–583 (2014).

    Article  CAS  Google Scholar 

  23. Britton, S. et al. DNA damage triggers SAF-A and RNA biogenesis factors exclusion from chromatin coupled to R-loops removal. Nucleic Acids Res. 42, 9047–9062 (2014).

    Article  CAS  Google Scholar 

  24. Groh, M., Lufino, M.M.P., Wade-Martins, R. & Gromak, N. R-loops associated with triplet repeat expansions promote gene silencing in Friedreich ataxia and fragile X syndrome. PLoS Genet. 10, e1004318 (2014).

    Article  Google Scholar 

  25. Helmrich, A., Ballarino, M. & Tora, L. Collisions between replication and transcription complexes cause common fragile site instability at the longest human genes. Mol. Cell 44, 966–977 (2011).

    Article  CAS  Google Scholar 

  26. Huertas, P. & Aguilera, A. Cotranscriptionally formed DNA:RNA hybrids mediate transcription elongation impairment and transcription-associated recombination. Mol. Cell 12, 711–721 (2003).

    Article  CAS  Google Scholar 

  27. Li, X. & Manley, J.L. Inactivation of the SR protein splicing factor ASF/SF2 results in genomic instability. Cell 122, 365–378 (2005).

    Article  CAS  Google Scholar 

  28. Castellano-Pozo, M. et al. R loops are linked to histone H3 S10 phosphorylation and chromatin condensation. Mol. Cell 52, 583–590 (2013).

    Article  CAS  Google Scholar 

  29. Boque-Sastre, R. et al. Head-to-head antisense transcription and R-loop formation promotes transcriptional activation. Proc. Natl. Acad. Sci. USA 112, 5785–5790 (2015).

    Article  CAS  Google Scholar 

  30. Costantino, L. & Koshland, D. The Yin and Yang of R-loop biology. Curr. Opin. Cell Biol. 34, 39–45 (2015).

    Article  CAS  Google Scholar 

  31. Aguilera, A. & García-Muse, T. R loops: from transcription byproducts to threats to genome stability. Mol. Cell 46, 115–124 (2012).

    Article  CAS  Google Scholar 

  32. Shen, Y.J. et al. Genome-derived cytosolic DNA mediates type I interferon-dependent rejection of B cell lymphoma cells. Cell Reports 11, 460–473 (2015).

    Article  CAS  Google Scholar 

  33. Wang, H. et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).

    Article  CAS  Google Scholar 

  34. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  Google Scholar 

  35. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  Google Scholar 

  36. Kanhere, A. et al. Short RNAs are transcribed from repressed polycomb target genes and interact with polycomb repressive complex-2. Mol. Cell 38, 675–688 (2010).

    Article  CAS  Google Scholar 

  37. Lynch, M.D. et al. An interspecies analysis reveals a key role for unmethylated CpG dinucleotides in vertebrate Polycomb complex recruitment. EMBO J. 31, 317–329 (2012).

    Article  CAS  Google Scholar 

  38. Brinkman, A.B. et al. Sequential ChIP-bisulfite sequencing enables direct genome-scale investigation of chromatin and DNA methylation cross-talk. Genome Res. 22, 1128–1138 (2012).

    Article  CAS  Google Scholar 

  39. Mendenhall, E.M. et al. GC-rich sequence elements recruit PRC2 in mammalian ES cells. PLoS Genet. 6, e1001244 (2010).

    Article  Google Scholar 

  40. Skourti-Stathaki, K., Proudfoot, N.J. & Gromak, N. Human senataxin resolves RNA/DNA hybrids formed at transcriptional pause sites to promote Xrn2-dependent termination. Mol. Cell 42, 794–805 (2011).

    Article  CAS  Google Scholar 

  41. Wahba, L., Gore, S.K. & Koshland, D. The homologous recombination machinery modulates the formation of RNA-DNA hybrids and associated chromosome instability. eLife 2, e00505 (2013).

    Article  Google Scholar 

  42. Skourti-Stathaki, K. & Proudfoot, N.J. A double-edged sword: R loops as threats to genome integrity and powerful regulators of gene expression. Genes Dev. 28, 1384–1396 (2014).

    Article  CAS  Google Scholar 

  43. Cavalli, G. & Paro, R. The Drosophila Fab-7 chromosomal element conveys epigenetic inheritance during mitosis and meiosis. Cell 93, 505–518 (1998).

    Article  CAS  Google Scholar 

  44. Klymenko, T. & Müller, J. The histone methyltransferases Trithorax and Ash1 prevent transcriptional silencing by Polycomb group proteins. EMBO Rep. 5, 373–377 (2004).

    Article  CAS  Google Scholar 

  45. Schmitt, S., Prestel, M. & Paro, R. Intergenic transcription through a polycomb group response element counteracts silencing. Genes Dev. 19, 697–708 (2005).

    Article  CAS  Google Scholar 

  46. Shi, X. et al. ING2 PHD domain links histone H3 lysine 4 methylation to active gene repression. Nature 442, 96–99 (2006).

    Article  CAS  Google Scholar 

  47. Hooper, M., Hardy, K., Handyside, A., Hunter, S. & Monk, M. HPRT-deficient (Lesch-Nyhan) mouse embryos derived from germline colonization by cultured cells. Nature 326, 292–295 (1987).

    Article  CAS  Google Scholar 

  48. Hainer, S.J. et al. Suppression of pervasive noncoding transcription in embryonic stem cells by esBAF. Genes Dev. 29, 362–378 (2015).

    Article  CAS  Google Scholar 

  49. 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  Google Scholar 

  50. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  Google Scholar 

  51. 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  Google Scholar 

  52. Pervouchine, D.D. et al. Enhanced transcriptome maps from multiple mouse tissues reveal evolutionary constraint in gene expression. Nat. Commun. 6, 5903 (2015).

    Article  CAS  Google Scholar 

  53. Li, B. & Dewey, C.N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    Article  CAS  Google Scholar 

  54. Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

We thank W. Hardy and M. Green (University of Massachusetts Medical School) for the pCAGGS-ires-Hygro expression vector and S. Hainer, L. Ee and K. McCannell for input on the manuscript. This work was supported by US National Institutes of Health (NIH) grant R01HD072122 and American Cancer Society grant RSG-14-220-01 (both to T.G.F.) and NIH grant R01HD080224 (to O.J.R.). T.G.F. was supported as a Pew Scholar in the Biological Sciences and is supported as a Leukemia and Lymphoma Society Scholar. Deep sequencing was performed at the University of Massachusetts Medical School Core facility on a HiSeq2000 supported by NIH grant 1S10RR027052-01.

Author information

Authors and Affiliations

Authors

Contributions

P.B.C. performed all experiments and bioinformatics analyses, except as otherwise indicated. H.V.C. prepared libraries for RIP-seq, D.A. generated several of the ESC lines, and T.G.F. performed Suz12 ChIP-seq and analyzed the data. P.B.C., O.J.R. and T.G.F. designed the experiments. P.B.C. and T.G.F. wrote the paper with input from all authors.

Corresponding author

Correspondence to Thomas G Fazzio.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characteristics of Tip60–p400–bound transcripts.

a-b, Scatter plots of biological replicate RIP-seq data (Log2 RPKM) for p400 RIPs (a) and Ruvbl1 RIPs (b). Correlation coefficients (R) are noted. c, Higher enrichment of 5’ reads (within the 1st exon and 1st intron) among transcripts from RIP-seq experiments relative to RNA-seq. Shown are cumulative distribution plots indicating a significant increase in the proportion of reads in the first exon and first intron within the p400 RIP-seq libraries relative to RNA-seq. Similar findings were observed for Ruvbl1 RIP-seq. P-value indicating significant enrichment of 5’ proximal reads in RIP-seq data were calculated using a two-sample K-S test. d, Overlap between high confidence Tip60–p400 bound RNAs and genes. The significance of overlap was calculated using a hypergeometric test. e, Tip60–p400-bound transcripts occupy a broad range of expression. Scatter plot of biological replicate RNA-seq data (Log2 RPKM) overlaid with Tip60–p400-bound transcripts (red). f, Gene Ontology (GO) terms significantly overrepresented among Tip60–p400-bound transcripts, plotted as –Log10 (p-value).

Supplementary Figure 2 Acute transcription inhibition inhibits Tip60–p400 binding to chromatin.

a, Western blots of indicated proteins showing unchanged levels of Tip60–p400 subunits p400, Hdac6, Dmap1, Tip60, and Ruvbl1 upon DRB or Triptolide treatment. Actin serves as a loading control. b, Transcript levels of rapidly-degraded transcripts (Nanog, Myc) and slowly-degraded transcripts (Oct4, Gapdh), measured by RT-qPCR. Levels in cells treated with indicated transcription inhibitors are expressed relative to control cells. Error bars, s.d. (n = 3 technical replicates from one of two independent cell cultures for each condition.) c, ChIP-qPCR of the p400 subunit of Tip60–p400 complex with or without transcription inhibitor treatment. IgG serves as a negative control for ChIP. Enrichment levels are expressed for each locus as a fraction of input. Error bars, s.d. (n = 3 technical replicates from one of two independent cell cultures for each condition.) P values were calculated using two tailed students t-tests (*P < 0.05; **P < 0.01).

Supplementary Figure 3 Comparison of DRIP-RNA-seq method to standard DRIP-seq.

a, Published DRIP-seq data from human embryonal carcinoma cells (Ginno, P. et al., Molecular Cell 45:814–825, 2012) were aggregated over TSSs. Control: DRIP from unperturbed sample. RNaseH pre-treatment: samples were digested with RNaseH prior to DRIP, as a negative control for immunoprecipitation with the S9.6 antibody. b, Comparison of DRIP-seq and DRIP-RNA-seq protocol. c, Comparison of sense and antisense reads of DRIP data aggregated near promoters as in (a). d, Dot blots indicating changes in bulk R-loop abundance upon Rnaseh1 overexpression or treatment of cells with DRB or Triptolide. Treatment of purified genomic DNA with RNaseH in vitro (last lane) serves as a control. Significance of differences in (a) and (c) calculated using two-sample K-S tests, as above.

Supplementary Figure 4 Reduction in Tip60–p400 binding to target genes upon Rnaseh1 overexpression.

a-b, Browser tracks of Tip60 or p400 ChIP-seq data at two Tip60–p400 target genes, Rps9 (a) and Rpl8 (b) in control cells or Rnaseh1 overexpressing cells. Gene expression is shown as RNA-seq tracks for each gene. c, Two genes are shown with high (Cdc6) or low (Wipf2) expression levels to illustrate that some lowly expressed genes with R-loops exhibit Rnaseh1-sensitive Tip60–p400 binding, and some highly expressed genes without detectable R-loops are not highly bound by Tip60–p400.

Supplementary Figure 5 ChIP-qPCR validation of changes in factor binding upon Rnaseh1 overexpression.

a-b, ChIP-qPCR measurements of Tip60 (a) or Suz12 (b) association with genes that harbor minimal, modest, or high levels of R-loops, and the effect of R-loop destruction by Rnaseh1 overexpression are shown. Error bars, s.d. (n = 3 technical replicates from one of two independent cell cultures for each line.) P values comparing Rnaseh1 overexpression to controls were calculated using two tailed students t-tests (*P <0.05; **P < 0.01).

Supplementary Figure 6 Effects of Rnaseh1 overexpression on ESC proliferation and self-renewal.

a, Brightfield images or alkaline phosphatase staining of control or Rnaseh1 overexpressing ESCs. b, DNA content of control or Rnaseh1 overexpressing ESCs (X-axis) measured by propidium iodide staining and FACS. One representative curve of three biological replicates is shown for each cell type. c, Quantification of S-phase population for three biological replicate (independent cultures) control and Rnaseh1 overexpressing ESCs. N.S., differences in S-phase population are not statistically significant. d, RT-qPCR quantification of 18S and 28S rRNA levels in control or Rnaseh1 overexpressing ESCs. Data were normalized relative to Gapdh, and rRNA levels in control cells were set to 100%. Shown are technical replicates of one representative biological replicate of two. Error bars = standard deviations. e, Growth curve of three biological replicate (independent cultures) control or Rnaseh1 overexpressing ESCs compared to Ep400 knockdown (KD) ESCs. f, ChIP-qPCR measurements of TSS-proximal and gene body RNA Pol II association with genes that lose Tip60–p400 binding in RNaseH-oex cells show no reduction in RNA Pol II association upon Rnaseh1 overexpression. Error bars, s.d. (n = 3 technical replicates from one of two independent cell cultures for each line.) P values were calculated using two tailed students t-tests (*P < 0.05; **P < 0.01).

Supplementary Figure 7 Enhanced Suz12 binding and H3K27me3 localization upon Rnaseh1 overexpression.

a, Heatmaps, as in Fig. 5, showing promoter-proximal H3K27me3 localization for all genes in control or Rnaseh1 overexpressing ESCs. Heatmaps are sorted by H3K27me3 localization in control cells. b-c, Genes normally targeted by PRC2 show increased Suz12 enrichment in Rnaseh1 overexpressing cells. Browser tracks are depicted as in Fig. 5. Gene expression is shown as RNA-seq tracks for each gene. d, Browser tracks of R-loops and Tip60 and Suz12 association at a locus that switches from Tip60 binding to Suz12 binding upon disruption of R-loops, depicted as in Fig. 5. Gene expression is illustrated with RNA-seq tracks.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 4185 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, P., Chen, H., Acharya, D. et al. R loops regulate promoter-proximal chromatin architecture and cellular differentiation. Nat Struct Mol Biol 22, 999–1007 (2015). https://doi.org/10.1038/nsmb.3122

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3122

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing