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:

DNA G-quadruplex structures mold the DNA methylome

Abstract

Control of DNA methylation level is critical for gene regulation, and the factors that govern hypomethylation at CpG islands (CGIs) are still being uncovered. Here, we provide evidence that G-quadruplex (G4) DNA secondary structures are genomic features that influence methylation at CGIs. We show that the presence of G4 structure is tightly associated with CGI hypomethylation in the human genome. Surprisingly, we find that these G4 sites are enriched for DNA methyltransferase 1 (DNMT1) occupancy, which is consistent with our biophysical observations that DNMT1 exhibits higher binding affinity for G4s as compared to duplex, hemi-methylated, or single-stranded DNA. The biochemical assays also show that the G4 structure itself, rather than sequence, inhibits DNMT1 enzymatic activity. Based on these data, we propose that G4 formation sequesters DNMT1 thereby protecting certain CGIs from methylation and inhibiting local methylation.

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

Fig. 1: G4 formation is associated with hypomethylation at CGIs.
Fig. 2: DNMT1 is recruited to BG4 peaks associated with low methylation.
Fig. 3: DNMT1 selectively binds and is inhibited by G4 structures.
Fig. 4: Recruitment of DNMT1 by the G4 structures shapes the methylome in G-rich regions.

Similar content being viewed by others

Data availability

K562 datasets for DHS (ENCSR000EPC), DNMT1 ChIP-seq (ENCSR987PBI), and whole-genome bisulfite sequencing (ENCSR765JPC) were downloaded from ENCODE. G4-ChIP-seq datasets for K562 and WGBS datasets for entinostat-treated and untreated HaCaT cells are available at the GEO repository under accession GSE107690. G4-ChIP-seq data in entinostat-treated and untreated HaCaT cells were taken from GSE76688. Source data for Figs. 1d,e,h and 3 are available online.

References

  1. Smith, Z. D. & Meissner, A. DNA methylation: roles in mammalian development. Nat. Rev. Genet. 14, 204–220 (2013).

    Article  CAS  Google Scholar 

  2. Feinberg, A. P. & Tycko, B. The history of cancer epigenetics. Nat. Rev. Cancer 4, 143–153 (2004).

    Article  CAS  Google Scholar 

  3. Li, E., Bestor, T. H. & Jaenisch, R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69, 915–926 (1992).

    Article  CAS  Google Scholar 

  4. Okano, M., Bell, D. W., Haber, D. A. & Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247–257 (1999).

    Article  CAS  Google Scholar 

  5. Liao, J. et al. Targeted disruption of DNMT1, DNMT3A and DNMT3B in human embryonic stem cells. Nat. Genet. 47, 469–478 (2015).

    Article  CAS  Google Scholar 

  6. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 16, 6–21 (2002).

    Article  CAS  Google Scholar 

  7. Lister, R. et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462, 315–322 (2009).

    Article  CAS  Google Scholar 

  8. Illingworth, R. S. & Bird, A. P. CpG islands – ‘A rough guide’. FEBS Lett. 583, 1713–1720 (2009).

    Article  CAS  Google Scholar 

  9. Deaton, A. & Bird, A. CpG islands and the regulation of transcription. Genes Dev. 25, 1010–1022 (2011).

    Article  CAS  Google Scholar 

  10. Reik, W., Dean, W. & Walter, J. Epigenetic reprogramming in mammalian development. Science 293, 1089–1093 (2001).

    Article  CAS  Google Scholar 

  11. Li, E. Chromatin modification and epigenetic reprogramming in mammalian development. Nat. Rev. Genet. 3, 662–673 (2002).

    Article  CAS  Google Scholar 

  12. Reik, W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 447, 425–432 (2007).

    Article  CAS  Google Scholar 

  13. Long, H. K., King, H. W., Patient, R. K., Odom, D. T. & Klose, R. J. Protection of CpG islands from DNA methylation is DNA-encoded and evolutionarily conserved. Nucleic Acids Res. 44, 6693–6706 (2016).

    Article  CAS  Google Scholar 

  14. Lienert, F. et al. Identification of genetic elements that autonomously determine DNA methylation states. Nat. Genet. 43, 1091–1097 (2011).

    Article  CAS  Google Scholar 

  15. Wachter, E. et al. Synthetic CpG islands reveal DNA sequence determinants of chromatin structure. eLife 3, 1–16 (2014).

    Article  Google Scholar 

  16. Krebs, A. R., Dessus-Babus, S., Burger, L. & Schübeler, D. High-throughput engineering of a mammalian genome reveals building principles of methylation states at CG rich regions. eLife 3, e04094 (2014).

    Article  Google Scholar 

  17. Takahashi, Y. et al. Integration of CpG-free DNA induces de novo methylation of CpG islands in pluripotent stem cells. Science 356, 503–508 (2017).

    Article  CAS  Google Scholar 

  18. Quante, T. & Bird, A. Do short, frequent DNA sequence motifs mould the epigenome? Nat. Rev. Mol. Cell Biol. 17, 257–262 (2016).

    Article  CAS  Google Scholar 

  19. Domcke, S. et al. Competition between DNA methylation and transcription factors determines binding of NRF1. Nature 528, 575–579 (2015).

    Article  CAS  Google Scholar 

  20. Stadler, M. B. et al. DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature 480, 490–495 (2011).

    CAS  PubMed  Google Scholar 

  21. Thurman, R. E. et al. The accessible chromatin landscape of the human genome. Nature 489, 75–82 (2012).

    Article  CAS  Google Scholar 

  22. Ooi, S. K. T. et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448, 714–717 (2007).

    Article  CAS  Google Scholar 

  23. Du, J., Johnson, L. M., Jacobsen, S. E. & Patel, D. J. DNA methylation pathways and their crosstalk with histone methylation. Nat. Rev. Mol. Cell Biol. 16, 519–532 (2015).

    Article  CAS  Google Scholar 

  24. Clouaire, T. et al. Cfp1 integrates both CpG content and gene activity for accurate H3K4me3 deposition in embryonic stem cells. Genes Dev. 26, 1714–1728 (2012).

    Article  CAS  Google Scholar 

  25. Thomson, J. P. et al. CpG islands influence chromatin structure via the CpG-binding protein Cfp1. Nature 464, 1082–1086 (2010).

    Article  CAS  Google Scholar 

  26. Hänsel-Hertsch, R., Di Antonio, M. & Balasubramanian, S. DNA G-quadruplexes in the human genome: Detection, functions and therapeutic potential. Nat. Rev. Mol. Cell Biol. 18, 279–284 (2017).

    Article  Google Scholar 

  27. Chambers, V. S. et al. High-throughput sequencing of DNA G-quadruplex structures in the human genome. Nat. Biotechnol. 33, 1–7 (2015).

    Article  Google Scholar 

  28. Biffi, G., Tannahill, D., McCafferty, J. & Balasubramanian, S. Quantitative visualization of DNA G-quadruplex structures in human cells. Nat. Chem. 5, 182–186 (2013).

    Article  CAS  Google Scholar 

  29. Rhodes, D. & Lipps, H. J. G-quadruplexes and their regulatory roles in biology. Nucleic Acids Res. 43, 8627–8637 (2015).

    Article  CAS  Google Scholar 

  30. Hänsel-Hertsch, R., Spiegel, J., Marsico, G., Tannahill, D. & Balasubramanian, S. Genome-wide mapping of endogenous G-quadruplex DNA structures by chromatin immunoprecipitation and high-throughput sequencing. Nat. Protoc. 13, 551–564 (2018).

    Article  Google Scholar 

  31. Hänsel-Hertsch, R. et al. G-quadruplex structures mark human regulatory chromatin. Nat. Genet. 48, 1267–1272 (2016).

    Article  Google Scholar 

  32. De, S. & Michor, F. DNA secondary structures and epigenetic determinants of cancer genome evolution. Nat. Struct. Mol. Biol. 18, 950–955 (2011).

    Article  CAS  Google Scholar 

  33. Dunham, I. et al. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  CAS  Google Scholar 

  34. Gardiner-Garden, M. & Frommer, M. CpG Islands in vertebrate genomes. J. Mol. Biol. 196, 261–282 (1987).

    Article  CAS  Google Scholar 

  35. Jones, P. A. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 13, 484–492 (2012).

    Article  CAS  Google Scholar 

  36. Hackenberg, M. et al. CpGcluster: A distance-based algorithm for CpG-island detection. BMC Bioinformatics 7, 1–13 (2006).

    Article  Google Scholar 

  37. Chen, L. et al. R-ChIP using inactive RNase H reveals dynamic coupling of R-loops with transcriptional pausing at gene promoters. Mol. Cell 68, 745–757.e5 (2017).

    Article  CAS  Google Scholar 

  38. Fan, G. et al. DNA hypomethylation perturbs the function and survival of CNS neurons in postnatal animals. J. Neurosci. 21, 788–797 (2001).

    Article  CAS  Google Scholar 

  39. Jackson-Grusby, L. et al. Loss of genomic methylation causes p53-dependent apoptosis and epigenetic deregulation. Nat. Genet. 27, 31–39 (2001).

    Article  CAS  Google Scholar 

  40. Dai, J. et al. An intramolecular G-quadruplex structure with mixed parallel/antiparallel G-strands formed in the human BCL-2 promoter region in solution. J. Am. Chem. Soc. 128, 1096–1098 (2006).

    Article  CAS  Google Scholar 

  41. Kuryavyi, V., Phan, A. T. & Patel, D. J. Solution structures of all parallel-stranded monomeric and dimeric G-quadruplex scaffolds of the human c-kit2 promoter. Nucleic Acids Res. 38, 6757–6773 (2010).

    Article  CAS  Google Scholar 

  42. Ambrus, A., Chen, D., Dai, J., Jones, R. A. & Yang, D. Solution structure of the biologically relevant G-quadruplex element in the human c-MYC promoter. Implications for G-quadruplex stabilization. Biochemistry 44, 2048–2058 (2005).

    Article  CAS  Google Scholar 

  43. Biffi, G., Tannahill, D. & Balasubramanian, S. An intramolecular G-quadruplex structure is required for binding of telomeric repeat-containing RNA to the telomeric protein TRF2. J. Am. Chem. Soc. 134, 11974–11976 (2012).

    Article  CAS  Google Scholar 

  44. Giraldo, R. & Rhodes, D. The yeast telomere-binding protein RAP1 binds to and promotes the formation of DNA quadruplexes in telomeric DNA. EMBO J. 13, 2411–2420 (1994).

    Article  CAS  Google Scholar 

  45. Bacolla, A., Pradhan, S., Roberts, R. J. & Wells, R. D. Recombinant human DNA (cytosine-5) methyltransferase. J. Biol. Chem. 274, 33002–33010 (1999).

    Article  Google Scholar 

  46. Sanz, L. A. et al. Prevalent, dynamic, and conserved R-loop structures associate with specific epigenomic signatures in mammals. Mol. Cell 63, 167–178 (2016).

    Article  CAS  Google Scholar 

  47. 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 

  48. Wang, X. et al. Targeting of Polycomb Repressive Complex 2 to RNA by short repeats of consecutive guanines. Mol. Cell 65, 1056–1067.e5 (2017).

    Article  CAS  Google Scholar 

  49. Wang, X. et al. Molecular analysis of PRC2 recruitment to DNA in chromatin and its inhibition by RNA. Nat. Struct. Mol. Biol. (2017). https://doi.org/10.1038/nsmb.3487

    Article  CAS  Google Scholar 

  50. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10 (2011).

    Article  Google Scholar 

  51. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

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

    Article  Google Scholar 

  53. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  Google Scholar 

  54. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  CAS  Google Scholar 

  55. Peat, J. R. et al. Genome-wide bisulfite sequencing in zygotes identifies demethylation targets and maps the contribution of TET3 oxidation. Cell Rep. 9, 1990–2000 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work is supported by a core CRUK award (C14303/A17197). S.B. is a Senior Investigator of the Wellcome Trust (grant no. 099232/z/12/z). J.S. is a Marie Curie Fellow of the European Union (747297-QAPs-H2020-MSCA-IF-2016).

Author information

Authors and Affiliations

Authors

Contributions

The project was conceived by S.M. and S.B. S.M. designed and carried out all the experiments with discussions with D.B., J.S., R.H.H., D.T., and S.B. S.M. designed the analysis strategies with input from A.G., D.T., and S.B. J.S. performed the G4-ChIP-seq experiments. A.G. and S.M.C. carried out all the computational analysis with discussions with S.M., D.T., D.B., R.H.H., and G.M. M.D. carried out the circular dichroism spectroscopy and ultraviolet melting experiments. All authors interpreted the results. S.M., D.T., and S.B. wrote the paper with input from all of the authors.

Corresponding author

Correspondence to Shankar Balasubramanian.

Ethics declarations

Competing interests

S.B. is an advisor and shareholder of Cambridge Epigenetix Limited.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 BG4 binds unmethylated and methylated G4 structures with the same affinity.

ELISA assays testing binding of BG4 antibody to unmethylated and methylated G4 structures and control non-G4-forming oligonucleotides. CpG sites are highlighted in red. MG denotes methylated CpG. Shown are the mean and s.d. from three measurements.

Supplementary Figure 2 Methylation is depleted in BG4 regions.

a, Box-and-whisker plot showing the average methylation for BG4 peaks (n = 8,210), DHSs (n = 142,115) and CGIs (n = 22,673). Similar to Fig. 1e, apart from using the CGI set generated by CpGCluster. b, Box-and-whisker plot showing the methylation levels for BG4 peaks and CGIs at different CpG densities. Similar to Fig. 1h, apart from using the CGI set generated by CpGCluster. c, Box-and-whisker plot showing the average methylation levels for CGIs with or without a BG4 peak at different CpG densities. d, Box-and-whisker plot showing average methylation on CGIs with respect to BG4 peaks in the presence (+) or absence (–) of a DHS or promoter. The number of CGI regions in each category is presented on top of the plot. e, Box-and-whisker plot showing the average methylation for BG4 peaks (n = 17,101), ATAC (ATAC-seq peaks denoting open chromatin, n = 23,217) and CGI regions (n = 26,580) in untreated HaCaT cells. f, Box and scatterplot showing differential percentage methylation in entinostat-treated versus untreated cells for promoter CGIs in open chromatin regions containing sequences with potential to form G4s. i, BG4-negative CGIs without a G4 ChIP-seq peak but having potential to form a G4 structure (n = 1,504); ii, BG4-constant CGIs with a least one high-confidence G4 ChIP-seq peak that does not significantly change before and after treatment (n = 3,261); iii, BG4 increases where a BG4 peak significantly increases in size after treatment (n = 307). Each gray dot represents one CGI region. P values were calculated using a Mann–Whitney U-test.

Supplementary Figure 3 DNMT1 is enriched at BG4 peaks associated with low methylation.

Binding profile of DNMT1 in CGIs with low (less than 20%, n = 14,983), intermediate (between 20% and 80%, n = 4,864) and high (more than 80%, n = 2,826) methylation. Above each plot is a heat map showing the enrichment of BG4 peaks and DHSs across the respective regions. Similar to Fig. 2b, apart from using the CGI set generated by CpGCluster.

Supplementary Figure 4 Structure verification of oligonucleotides used in this study and inhibition of DNMT1 by G4 DNA.

ac, Circular dichroism spectra of BCL2 and BCL2-mut (a), KIT2 and BKIT2-mut (b), and MYC and MYC-mut (c). Sequences are listed below the graph. df, ultraviolet melting profiles of BCL2 (d), KIT2 (e), and MYC (f). Mutated oligonucleotides lose the capacity to form G4s and therefore have no absorbance at 295 nm. gi, Circular dichroism spectra of BCL2-0CG/2CG/3CG and BCL2-CCC (g), KIT2-0CG/2CG/CGCG and KIT2-CCC (h), and MYC-2CG/CTCA/4CG and MYC-CCC (i). Sequences are listed below the graph. Note that circular dichroism spectra of all G4-forming oligonucleotides show a positive peak at ~263 nm and a negative peak at ~240 nm, which is characteristic of a G4 structures. j, DNMT1 activity in the presence of BCL2 (G4 structure), BCL2-CCC (C-rich, non-G4 forming with 5 CpGs), BCL2-mut (wild-type BCL2 with mutations in G4 tetrad Gs, non-G4 forming), BCL2-2CG (G4 forming with 2 CpGs), BCL2-3CG (G4 forming with 3CpGs) and BCL2-0CG (G4 forming without CpGs). k, KIT2 (G4 structure), KIT2-CCC (C-rich, non-G4 forming with 4 CpGs), KIT2-mut (wild-type KIT2 with mutations in G4 tetrad Gs, non-G4 forming), KIT2-0CG (G4 forming without CpGs), KIT2-2CG (G4 forming with 2 CpGs), and KIT-CGCG (G4 forming with 2 adjacent CpGs). l, MYC (G4 structure), MYC-CCC (C-rich, non-G4 forming with no CpGs), MYC-mut (wild-type MYC with mutations in G4 tetrad Gs, non-G4 forming), MYC-2CG (G4 forming with 2 CpGs), MYC-CTCA (G4 forming without CpGs), and MYC-4CG (G4 forming with 4 CpGs). Sequences of the oligonucleotides used are given below the graphs. Shown are mean±s.d., n = 3 independent experiments. Statistical tests were done using two-way ANOVA.

Supplementary Figure 5 Depletion of DNA methylation is independent of R-loop formation.

Plot showing the average methylation profile centered around BG4 peak regions overlapping with R-loop regions (BG4 + R-loop, red and blue are replicates 1 and 2, respectively, n = 5,464) or BG4 peak regions without overlapping R-loop (BG4 – R-loop, pink and green are replicates 1 and 2, respectively, n = 3,111). The plot extends ±5 kb from the center of BG4 peaks.

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mao, SQ., Ghanbarian, A.T., Spiegel, J. et al. DNA G-quadruplex structures mold the DNA methylome. Nat Struct Mol Biol 25, 951–957 (2018). https://doi.org/10.1038/s41594-018-0131-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-018-0131-8

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