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:

Structure of nucleosome-bound DNA methyltransferases DNMT3A and DNMT3B

Abstract

CpG methylation by de novo DNA methyltransferases (DNMTs) 3A and 3B is essential for mammalian development and differentiation and is frequently dysregulated in cancer1. These two DNMTs preferentially bind to nucleosomes, yet cannot methylate the DNA wrapped around the nucleosome core2, and they favour the methylation of linker DNA at positioned nucleosomes3,4. Here we present the cryo-electron microscopy structure of a ternary complex of catalytically competent DNMT3A2, the catalytically inactive accessory subunit DNMT3B3 and a nucleosome core particle flanked by linker DNA. The catalytic-like domain of the accessory DNMT3B3 binds to the acidic patch of the nucleosome core, which orients the binding of DNMT3A2 to the linker DNA. The steric constraints of this arrangement suggest that nucleosomal DNA must be moved relative to the nucleosome core for de novo methylation to occur.

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: Cryo-EM structure of the human DNMT3A2–DNMT3B3–NCP complex.
Fig. 2: Details of the acidic patch and DNA–catalytic domain interactions.
Fig. 3: The TRD mediates DNA binding but prevents NCP core binding.
Fig. 4: The DNMT3B3–acidic patch interaction is required for chromatin recruitment.

Similar content being viewed by others

Data availability

Density maps and structure coordinates have been deposited in the Electron Microscopy Data Bank with accession codes EMD-20281 and EMD-21689 and the Protein Data Bank with accession code 6PA7. The DNA methylation and MNase data have been deposited in the GEO database with accession code GSE152640. No restrictions are placed on data availability. Source data are provided with this paper.

References

  1. Jones, P. A. & Baylin, S. B. The fundamental role of epigenetic events in cancer. Nat. Rev. Genet. 3, 415–428 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Jones, P. A. & Liang, G. Rethinking how DNA methylation patterns are maintained. Nat. Rev. Genet. 10, 805–811 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Takeshima, H., Suetake, I. & Tajima, S. Mouse Dnmt3a preferentially methylates linker DNA and is inhibited by histone H1. J. Mol. Biol. 383, 810–821 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Kelly, T. K. et al. Genome-wide mapping of nucleosome positioning and DNA methylation within individual DNA molecules. Genome Res. 22, 2497–2506 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bourc’his, D., Xu, G. L., Lin, C. S., Bollman, B. & Bestor, T. H. Dnmt3L and the establishment of maternal genomic imprints. Science 294, 2536–2539 (2001).

    Article  ADS  PubMed  Google Scholar 

  6. Duymich, C. E., Charlet, J., Yang, X., Jones, P. A. & Liang, G. DNMT3B isoforms without catalytic activity stimulate gene body methylation as accessory proteins in somatic cells. Nat. Commun. 7, 11453 (2016).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  7. Jia, D., Jurkowska, R. Z., Zhang, X., Jeltsch, A. & Cheng, X. Structure of Dnmt3a bound to Dnmt3L suggests a model for de novo DNA methylation. Nature 449, 248–251 (2007).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Guo, X. et al. Structural insight into autoinhibition and histone H3-induced activation of DNMT3A. Nature 517, 640–644 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Zhang, Z. M. et al. Structural basis for DNMT3A-mediated de novo DNA methylation. Nature 554, 387–391 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bowman, R. L., Busque, L. & Levine, R. L. Clonal hematopoiesis and evolution to hematopoietic malignancies. Cell Stem Cell 22, 157–170 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Heyn, P. et al. Gain-of-function DNMT3A mutations cause microcephalic dwarfism and hypermethylation of Polycomb-regulated regions. Nat. Genet. 51, 96–105 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Sendžikaitė, G., Hanna, C. W., Stewart-Morgan, K. R., Ivanova, E. & Kelsey, G. A DNMT3A PWWP mutation leads to methylation of bivalent chromatin and growth retardation in mice. Nat. Commun. 10, 1884 (2019).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  14. Tatton-Brown, K. et al. Mutations in the DNA methyltransferase gene DNMT3A cause an overgrowth syndrome with intellectual disability. Nat. Genet. 46, 385–388 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lowary, P. T. & Widom, J. New DNA sequence rules for high affinity binding to histone octamer and sequence-directed nucleosome positioning. J. Mol. Biol. 276, 19–42 (1998).

    Article  CAS  PubMed  Google Scholar 

  16. Kareta, M. S., Botello, Z. M., Ennis, J. J., Chou, C. & Chédin, F. Reconstitution and mechanism of the stimulation of de novo methylation by human DNMT3L. J. Biol. Chem. 281, 25893–25902 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Rhee, I. et al. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature 416, 552–556 (2002).

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Egger, G. et al. Identification of DNMT1 (DNA methyltransferase 1) hypomorphs in somatic knockouts suggests an essential role for DNMT1 in cell survival. Proc. Natl Acad. Sci. USA 103, 14080–14085 (2006).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yang, X. et al. Gene body methylation can alter gene expression and is a therapeutic target in cancer. Cancer Cell 26, 577–590 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Rondelet, G., Dal Maso, T., Willems, L. & Wouters, J. Structural basis for recognition of histone H3K36me3 nucleosome by human de novo DNA methyltransferases 3A and 3B. J. Struct. Biol. 194, 357–367 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Dukatz, M. et al. H3K36me2/3 binding and DNA binding of the DNA methyltransferase DNMT3A PWWP domain both contribute to its chromatin interaction. J. Mol. Biol. 431, 5063–5074 (2019).

    Article  CAS  PubMed  Google Scholar 

  22. Weinberg, D. N. et al. The histone mark H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape. Nature 573, 281–286 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Qiu, C., Sawada, K., Zhang, X. & Cheng, X. The PWWP domain of mammalian DNA methyltransferase Dnmt3b defines a new family of DNA-binding folds. Nat. Struct. Biol. 9, 217–224 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen, T., Tsujimoto, N. & Li, E. The PWWP domain of Dnmt3a and Dnmt3b is required for directing DNA methylation to the major satellite repeats at pericentric heterochromatin. Mol. Cell. Biol. 24, 9048–9058 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ge, Y. Z. et al. Chromatin targeting of de novo DNA methyltransferases by the PWWP domain. J. Biol. Chem. 279, 25447–25454 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Zhang, Y. et al. Chromatin methylation activity of Dnmt3a and Dnmt3a/3L is guided by interaction of the ADD domain with the histone H3 tail. Nucleic Acids Res. 38, 4246–4253 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Klimasauskas, S., Kumar, S., Roberts, R. J. & Cheng, X. HhaI methyltransferase flips its target base out of the DNA helix. Cell 76, 357–369 (1994).

    Article  CAS  PubMed  Google Scholar 

  28. Song, J., Rechkoblit, O., Bestor, T. H. & Patel, D. J. Structure of DNMT1-DNA complex reveals a role for autoinhibition in maintenance DNA methylation. Science 331, 1036–1040 (2011).

    Article  ADS  CAS  PubMed  Google Scholar 

  29. Henikoff, J. G., Belsky, J. A., Krassovsky, K., MacAlpine, D. M. & Henikoff, S. Epigenome characterization at single base-pair resolution. Proc. Natl Acad. Sci. USA 108, 18318–18323 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chodavarapu, R. K. et al. Relationship between nucleosome positioning and DNA methylation. Nature 466, 388–392 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Barisic, D., Stadler, M. B., Iurlaro, M. & Schübeler, D. Mammalian ISWI and SWI/SNF selectively mediate binding of distinct transcription factors. Nature 569, 136–140 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lyons, D. B. & Zilberman, D. DDM1 and Lsh remodelers allow methylation of DNA wrapped in nucleosomes. eLife 6, e30674 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Goll, M. G. & Bestor, T. H. Eukaryotic cytosine methyltransferases. Annu. Rev. Biochem. 74, 481–514 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Yu, W. et al. Genome-wide DNA methylation patterns in LSH mutant reveals de-repression of repeat elements and redundant epigenetic silencing pathways. Genome Res. 24, 1613–1623 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Simon, M. D. & Shokat, K. M. A method to site-specifically incorporate methyl-lysine analogues into recombinant proteins. Methods Enzymol. 512, 57–69 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Simon, M. D. et al. The site-specific installation of methyl-lysine analogs into recombinant histones. Cell 128, 1003–1012 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bouazoune, K., Miranda, T. B., Jones, P. A. & Kingston, R. E. Analysis of individual remodeled nucleosomes reveals decreased histone-DNA contacts created by hSWI/SNF. Nucleic Acids Res. 37, 5279–5294 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dyer, P. N. et al. Reconstitution of nucleosome core particles from recombinant histones and DNA. Methods Enzymol. 375, 23–44 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Luger, K., Rechsteiner, T. J. & Richmond, T. J. Preparation of nucleosome core particle from recombinant histones. Methods Enzymol. 304, 3–19 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Ma, H. et al. A D53 repression motif induces oligomerization of TOPLESS corepressors and promotes assembly of a corepressor-nucleosome complex. Sci. Adv. 3, e1601217 (2017).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  41. Jeong, S. et al. Selective anchoring of DNA methyltransferases 3A and 3B to nucleosomes containing methylated DNA. Mol. Cell. Biol. 29, 5366–5376 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sharma, S., De Carvalho, D. D., Jeong, S., Jones, P. A. & Liang, G. Nucleosomes containing methylated DNA stabilize DNA methyltransferases 3A/3B and ensure faithful epigenetic inheritance. PLoS Genet. 7, e1001286 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhou, W., Triche, T. J. Jr, Laird, P. W. & Shen, H. SeSAMe: reducing artifactual detection of DNA methylation by Infinium BeadChips in genomic deletions. Nucleic Acids Res. 46, e123 (2018).

    PubMed  PubMed Central  Google Scholar 

  44. Kastner, B. et al. GraFix: sample preparation for single-particle electron cryomicroscopy. Nat. Methods 5, 53–55 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Stark, H. GraFix: stabilization of fragile macromolecular complexes for single particle cryo-EM. Methods Enzymol. 481, 109–126 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  47. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rohou, A. & Grigorieff, N. CTFFIND4: Fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 192, 216–221 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. Rosenthal, P. B. & Henderson, R. Optimal determination of particle orientation, absolute hand, and contrast loss in single-particle electron cryomicroscopy. J. Mol. Biol. 333, 721–745 (2003).

    Article  CAS  PubMed  Google Scholar 

  52. Kucukelbir, A., Sigworth, F. J. & Tagare, H. D. Quantifying the local resolution of cryo-EM density maps. Nat. Methods 11, 63–65 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  54. Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N. & Sternberg, M. J. The Phyre2 web portal for protein modeling, prediction and analysis. Nat. Protocols 10, 845–858 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D 60, 2126–2132 (2004).

    Article  PubMed  CAS  Google Scholar 

  56. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Cryo-EM data was collected at the David Van Andel Advanced Cryo-Electron Microscopy Suite at the Van Andel Institute (VAI). We thank X. Meng for cryo-EM data collection and EM technical support, the HPC team at VAI for computational support, D. Nadziejka for language editing of the manuscript, the VAI genomics core for performing the Illumina Infinium methylation EPIC array assay and J.-H. Min (Washington State University) and S. Baylin (John Hopkins University) for the indicated plasmids. This work was supported by the VAI (H.E.X., K.M., P.A.J.) and US National Cancer Institute (NCI; grants R35CA209859 to G.L., K.M. and P.A.J. and R50CA243878 to M.L.).

Author information

Authors and Affiliations

Authors

Contributions

H.E.X., K.M. and P.A.J. designed research. T.-H.X. expressed and purified proteins, prepared and screened samples and collected and processed cryo-EM data. T.-H.X. and M.L. performed biochemistry experiments. T.-H.X. and X.E.Z. built the atomic models and performed the refinements. T.-H.X., M.L., X.E.Z., G.L., G.Z., H.E.X., K.M. and P.A.J. analysed data. H.E.X., K.M. and P.A.J. wrote the paper with input from all authors.

Corresponding authors

Correspondence to H. Eric Xu, Karsten Melcher or Peter A. Jones.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Protein expression and nucleosome reconstitution.

a, Histone octamer expression and purification. Histones H3 and H4 and His6-Sumo-tagged H2A and H3 were coexpressed from a polycistronic expression vector. The octamer was purified by native Ni-affinity chromatography, and then cleavage of the His6-Sumo tags with ULP1 Sumo protease was followed by size exclusion chromatography (SEC). The SDS–PAGE gel shows analysis of histone octamer purification steps. b, SEC profile of histone octamer with indicated size standards. c, Native gel analysis of NCP reconstitution. NCPs reconstituted by double dialysis were separated by native gel electrophoresis (lane 1) together with a size standard (lane 2) and free nucleosomal DNA (lane 3). The gel was stained for both DNA (left) and protein (right). d, SEC profile of cleaved (red) and non-cleaved (blue) DNMT3 complex. e, SDS–PAGE analysis of GFP-tagged DNMT and untagged DNMT purification steps. The GFP tag allowed easy DNMT3B3 visualization in DNMT–NCP complexes. For gel source data, see Supplementary Fig. 3.

Extended Data Fig. 2 DNMT3A2, DNMT3B3 and NCP form a stable complex.

a, DNMT3 and NCP stably interact in an AlphaScreen luminescence proximity assay. Top panel, cartoon of AlphaScreen assay; bottom panel, AlphaScreen interaction data. The nucleosomal DNA of the NCP is biotinylated for immobilization on AlphaScreen streptavidin donor beads, and DNMT3B3 is His8-GFP-tagged for immobilization of Ni-chelating AlphaScreen acceptor beads. Donor beads contained a photosensitizer that, upon activation at 680 nm, converts ambient oxygen to singlet oxygen. If donor and acceptor beads are brought into close proximity by DNMT–NCP interaction, energy is transferred from singlet oxygen to thioxene derivatives in the acceptor beads, resulting in light emission at 520–620 nm. Data are mean ± s.e.m., n = 10. b, Increasing concentrations of GFP-tagged DNMT3A2–DNMT3B3 complex supershift the NCP band. M, DNA size marker. Panels 1–4 show native gel with indicated binding reactions in (1) the ethidium bromide channel to visualize the DNA of the NCP and (2) the fluorescence channel to visualize the GFP-tagged DNMT3A2–DNMT3B3 complex; (3) a merged image of the two channels (yellow bands indicates presence of both DNMT complex and NCP; and (4) gel stained with Coomassie blue. For gel source data, see Supplementary Fig. 4.

Source Data

Extended Data Fig. 3 DNMT3A–DNMT3B3 methylation activity and nucleosome interaction.

a, Free DNA methylation activity of acidic-patch-interacting and control mutant DNMT3B3 proteins. Histone-free (naked) 301-bp nucleosomal DNA was incubated with the indicated DNMT3A2–DNMT3B3 complexes containing wild-type or mutant DNMT3B3. Shown is the percentage methylation. Data are mean with individual values, n = 2. The total number of CpGs in the DNA is 24, of which a subset was preferentially methylated. b, Binding strength and structure resolution of DNMT3A2–DNMT3B3–NCP complexes with varying linker DNA lengths. Left, competition binding curves. AlphaScreen interaction between biotin-tagged NCPs and His6-tagged DNMT3A2–DNMT3B3 in the presence of increasing concentrations of untagged DNMT3A2–DNMT3B3. Data were generated from 3 independent experiments (n = 3) and normalized in each group with highest data point as 100%. Right, IC50 calculated in each experiment and represented in bar graphs. Data are mean ± s.e.m., n = 3, P values determined by non-parametric test with Dunn’s multiple comparisons test. ns, not significant. The table below compares binding strengths (IC50) and cryo-EM resolution of DNMT3A2–DNMT3B3 complexes with NCPs containing different length DNAs. The 200-kV data were collected on a Talos Arctica microscope with a Falcon 3 detector and the 300-kV data on a Titan Krios microscope with a K2 detector.

Source Data

Extended Data Fig. 4 Overall structure of DNMT bound to the nucleosome.

a, Left, local resolution map of DNMT–nucleosome core complex (global resolution of 2.94 Å); middle, cutaway view; right, corrected Fourier shell correlation (FSC) curves of the 3D reconstruction. b, Left, overview of DNMT–nucleosome complex with ADD domains; middle, cartoon representation. Right, overlay of distal ADD EM density with aligned ADD in the auto-inhibitory conformation from PDB 4U7P. The inhibitory ADD loop, residues 526–533, is highlighted in blue. c, Overall map fitting of the nucleosome, DNMT ADD-CD/CLD dimer and DNMT CD/CLD dimer with local resolution colour code. The resolutions were determined using the 0.143 FSC criterion. d, Density map of the CD–DNA interaction region generated in PyMOL and contoured at 8σ (left) and 5.5σ (right). The dashed line indicates the position of the TRD loop, which is only visible at the lower σ level and we have omitted in the final model. e, Density maps of the two SAH ligands generated in PyMOL and contoured at 8σ.

Extended Data Fig. 5 Cryo-EM 3D reconstruction and refinement.

a, Representative micrograph, with one particle highlighted in a green circle of 250 Å diameter. b, Representative 2D class averages. c, Workflow for cryo-electron microscopy data processing by cryoSPARC and RELION3.0. Boxed 3D classes were selected for further processing. The final global nominal resolution for the DNMT–NCP complex was 2.94 Å.

Extended Data Fig. 6 Focused refinement of DNMT and NCP.

About 559,000 good particles were selected from 3D reconstructions for further focused refinement. The final resolution for the DNMT3A2–DNMT3B3 complex including the ADD domains was 3.40 Å, for the DNMT3A2 CD–DNMT3B3 CLD complex was 3.24 Å and for the NCP was 2.78 Å.

Extended Data Fig. 7 Conformation and density asymmetry of linker DNA (CD-bound versus free) and CLD (NCP-bound versus free).

ab, Structure models overlaid with density maps (grey mesh) generated in PyMOL and contoured at 8σ (a) and 6σ (b). Note that at the 8σ contoured level, complete density is only visible for the CD-bound linker DNA and the histone-octamer-bound CLD. c, While the CD-bound linker DNA adopts a single conformation, the unbound linker DNA adopts four distinguishable conformations. Left, composite model; right, four different conformations shown as cryo-EM densities with their relative frequencies. d, Model of the DNMT3A2–DNMT3B3 complex with extended linker DNA and 3A2 ADD domain in its active conformation. The ADD has been modelled by structural alignment with PDB 4U7T. The structure model is fully compatible with the extended linker DNA. e, Model of the DNMT3A2–DNMT3B3 complex with extended linker DNA and 3A2 ADD domain in the autoinhibitory conformation. The ADD has been modelled by structural alignment with PDB 4U7P. This conformation is not compatible with the extended linker DNA.

Extended Data Fig. 8 MNase footprinting assay.

a, b, NCP301 alone (a) or in complex with DNMT3A2–DNMT3B3 (b) was subjected to MNase footprinting followed by DNA isolation and sequencing. Sequence reads shown by IGV viewer were aligned to the 301-bp reference to reveal the position of DNMT3A2–DNMT3B3 on individual nucleosomal DNA molecules (green, A; red, T; blue, C; brown, G). Magenta oval, position of the histone octamer (Widom 601 147-bp nucleosomal sequence); cyan bars, flexible linker region. Dashed lines demarcate the 10-bp linker regions of NCP167, and red dashed boxes represent the main DNMT protection region (protection of the DNMT–NCP complex relative to the DNMT-free NCP). The percentages of reads covering the DNMT protection regions are shown in red.

Extended Data Fig. 9 Histone H3 K36cMe3 did not improve the stability of ADD domains.

a, The H3K36cMe3 methyl-lysine analogue (MLA) peptide and H2A–H2B–H3K36cMe3 MLA–H4 histone octamer, but not the wild-type (WT) octamer, are recognized by anti-H3K36Me3 antibody. Four titrations (H3K36me3 peptide) or three titrations (H3Kc36me3 octamer) with different amounts of proteins were performed. b, AlphaScreen interaction assay between His-tagged DNMT3A2–DNMT3B3 complexes and biotinylated NCPs. The histone H3 K36cMe3 methyl analogue does not increase DNMT3A2–DNMT3B–NCP binding in the reconstituted system. Data are mean ± s.e.m., n = 3. c, Cryo-EM structure of the DNMT3A2–DNMT3B–H3K36cMe3 MLA NCP. The structure shows no additional density relative to that of the WT NCP complex. The ADD density is still poor and no reliable PWWP density is observed. d, Structural alignment of the DNMT–NCP WT (grey) and DNMT–NCP K36cMe3 (yellow) density maps shows their similar overall architecture. For gel source data, see Supplementary Fig. 5.

Source Data

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics

Supplementary information

Supplementary Information

This file contains: Supplementary Fig. 1: Sequence alignment; Supplementary Figs S2-S5: uncropped gels and size marker indications related to Fig. 4b and Extended Data Figs. 1a, 1c, 1e, 2b, and 9a and Supplementary Table 1: List of primers.

Reporting Summary

Video 1

Cryo-EM density map for human DNMT3A (CD) and DNMT3B3 (CLD) with NCP. Cryo-EM map and molecular model fitted in transparent density. The same color code is used as in Fig. 1c.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, TH., Liu, M., Zhou, X.E. et al. Structure of nucleosome-bound DNA methyltransferases DNMT3A and DNMT3B. Nature 586, 151–155 (2020). https://doi.org/10.1038/s41586-020-2747-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2747-1

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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