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.

  • Letter
  • Published:

Molecular basis of chromatin remodelling by DDM1 involved in plant DNA methylation

Abstract

Eukaryotic gene regulation occurs at the chromatin level, which requires changing the chromatin structure by a group of ATP-dependent DNA translocases—namely, the chromatin remodellers1. In plants, chromatin remodellers function in various biological processes and possess both conserved and plant-specific components2,3,4,5. DECREASE IN DNA METHYLATION 1 (DDM1) is a plant chromatin remodeller that plays a key role in the maintenance DNA methylation6,7,8,9,10,11. Here we determined the structures of Arabidopsis DDM1 in complex with nucleosome in ADP–BeFx-bound, ADP-bound and nucleotide-free conformations. We show that DDM1 specifically recognizes the H4 tail and nucleosomal DNA. The conformational differences between ADP–BeFx-bound, ADP-bound and nucleotide-free DDM1 suggest a chromatin remodelling cycle coupled to ATP binding, hydrolysis and ADP release. This, in turn, triggers conformational changes in the DDM1-bound nucleosomal DNA, which alters the nucleosome structure and promotes DNA sliding. Together, our data reveal the molecular basis of chromatin remodelling by DDM1.

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: Overall structure of the DDM1–NCP complex in the ADP-bound conformation.
Fig. 2: Interactions between DDM1 and NCP in the ADP-bound conformation.
Fig. 3: The conformational change of DDM1 in an ATPase cycle.
Fig. 4: DNA translocation by DDM1 during the chromatin remodelling cycle.

Similar content being viewed by others

Data availability

The structures have been deposited in the Protein Data Bank under accession codes 8WH8, 8WH9, 8WHA, 8WHB and 8WH5. The cryo-EM maps have been deposited in the Electron Microscopy Data Bank under accession codes EMD-37533, EMD-37535, EMD-37537, EMD-37538 and EMD-37529. Source data are provided with this paper.

References

  1. Clapier, C. R. & Cairns, B. R. The biology of chromatin remodeling complexes. Annu. Rev. Biochem. 78, 273–304 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Shang, J. Y. & He, X. J. Chromatin-remodeling complexes: conserved and plant-specific subunits in Arabidopsis. J. Integr. Plant Biol. 64, 499–515 (2022).

    Article  CAS  PubMed  Google Scholar 

  3. Bieluszewski, T., Prakash, S., Roule, T. & Wagner, D. The role and activity of SWI/SNF chromatin remodelers. Annu. Rev. Plant Biol. 74, 139–163 (2023).

    Article  CAS  PubMed  Google Scholar 

  4. Guo, J. et al. Comprehensive characterization of three classes of Arabidopsis SWI/SNF chromatin remodelling complexes. Nat. Plants 8, 1423–1439 (2022).

    Article  CAS  PubMed  Google Scholar 

  5. Fu, W. et al. Organization, genomic targeting, and assembly of three distinct SWI/SNF chromatin remodeling complexes in Arabidopsis. Plant Cell 35, 2464–2483 (2023).

    Article  PubMed  Google Scholar 

  6. Jeddeloh, J. A., Stokes, T. L. & Richards, E. J. Maintenance of genomic methylation requires a SWI2/SNF2-like protein. Nat. Genet. 22, 94–97 (1999).

    Article  CAS  PubMed  Google Scholar 

  7. Vongs, A., Kakutani, T., Martienssen, R. A. & Richards, E. J. Arabidopsis thaliana DNA methylation mutants. Science 260, 1926–1928 (1993).

    Article  CAS  PubMed  Google Scholar 

  8. Zhong, Z. et al. DNA methylation-linked chromatin accessibility affects genomic architecture in Arabidopsis. Proc. Natl Acad. Sci. USA 118, e2023347118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Choi, S. H. et al. Mutation in DDM1 inhibits the homology directed repair of double strand breaks. PLoS ONE 14, e0211878 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Miura, A. et al. Mobilization of transposons by a mutation abolishing full DNA methylation in Arabidopsis. Nature 411, 212–214 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Akinmusola, R. Y., Wilkins, C. A. & Doughty, J. DDM1-mediated TE silencing in plants. Plants (Basel) 12, 437 (2023).

    Article  CAS  PubMed  Google Scholar 

  12. Zhang, H., Lang, Z. & Zhu, J. K. Dynamics and function of DNA methylation in plants. Nat. Rev. Mol. Cell Biol. 19, 489–506 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Law, J. A. & Jacobsen, S. E. Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat. Rev. Genet. 11, 204–220 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. 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  PubMed  PubMed Central  Google Scholar 

  16. Kanno, T. et al. Involvement of putative SNF2 chromatin remodeling protein DRD1 in RNA-directed DNA methylation. Curr. Biol. 14, 801–805 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Kanno, T. et al. A SNF2-like protein facilitates dynamic control of DNA methylation. EMBO Rep. 6, 649–655 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zemach, A. et al. The Arabidopsis nucleosome remodeler DDM1 allows DNA methyltransferases to access H1-containing heterochromatin. Cell 153, 193–205 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Brzeski, J. & Jerzmanowski, A. Deficient in DNA methylation 1 (DDM1) defines a novel family of chromatin-remodeling factors. J. Biol. Chem. 278, 823–828 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Osakabe, A. et al. The chromatin remodeler DDM1 prevents transposon mobility through deposition of histone variant H2A.W. Nat. Cell Biol. 23, 391–400 (2021).

    Article  CAS  PubMed  Google Scholar 

  21. Jamge, B. et al. Histone variants shape chromatin states in Arabidopsis. eLife 12, RP87714 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zhou, J. et al. DDM1-mediated R-loop resolution and H2A.Z exclusion facilitates heterochromatin formation in Arabidopsis. Sci. Adv. 9, eadg2699 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, M. et al. Mechanism of DNA translocation underlying chromatin remodelling by Snf2. Nature 567, 409–413 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Liu, X., Li, M., Xia, X., Li, X. & Chen, Z. Mechanism of chromatin remodelling revealed by the Snf2-nucleosome structure. Nature 544, 440–445 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Chabre, M. Aluminofluoride and beryllofluoride complexes: new phosphate analogs in enzymology. Trends Biochem. Sci. 15, 6–10 (1990).

    Article  CAS  PubMed  Google Scholar 

  26. Luger, K., Mader, A. W., Richmond, R. K., Sargent, D. F. & Richmond, T. J. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 389, 251–260 (1997).

    Article  CAS  PubMed  Google Scholar 

  27. Lee, S. C. et al. Chromatin remodeling of histone H3 variants by DDM1 underlies epigenetic inheritance of DNA methylation. Cell 186, 4100–4116 e15 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Osakabe, A. et al. Molecular and structural basis of the heterochromatin-specific chromatin remodeling activity by Arabidopsis DDM1. Preprint at bioRxiv https://doi.org/10.1101/2023.07.10.548306 (2023).

  29. Paintsil, E. A. & Morrison, E. A. Preparation of recombinant histones and Widom 601 DNA for reconstitution of nucleosome core particles. Methods Mol. Biol. 2599, 163–175 (2023).

    Article  CAS  PubMed  Google Scholar 

  30. Hammonds, E. F. & Morrison, E. A. Nucleosome core particle reconstitution with recombinant histones and Widom 601 DNA. Methods Mol. Biol. 2599, 177–190 (2023).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  33. 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 

  34. 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 

  35. 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 

  36. Zivanov, J., Nakane, T. & Scheres, S. H. W. Estimation of high-order aberrations and anisotropic magnification from cryo-EM data sets in RELION-3.1. IUCrJ 7, 253–267 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Scheres, S. H. & Chen, S. Prevention of overfitting in cryo-EM structure determination. Nat. Methods 9, 853–854 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Baek, M. et al. Accurate prediction of protein structures and interactions using a three-track neural network. Science 373, 871–876 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  40. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D Struct. Biol. 74, 531–544 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. Kiianitsa, K., Solinger, J. A. & Heyer, W. D. NADH-coupled microplate photometric assay for kinetic studies of ATP-hydrolyzing enzymes with low and high specific activities. Anal. Biochem. 321, 266–271 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H. Huang for discussion, the staff at SUSTech Cryo-EM Center for help in data collection and SUSTech Core Research Facilities for help with the circular dichroism experiment. This work was supported by the National Natural Science Foundation of China (grant no. 32325008 to J.D.), the Shenzhen Science and Technology Program (grant nos RCJC20221008092720004, KQTD20190929173906742 and 20231120201445001 to J.D.) and the EMBO Postdoctoral Fellowship (ALTF 579-2022 to H.H.).

Author information

Authors and Affiliations

Authors

Contributions

Y.L. and Z.Z. performed the experiments. F.Z., Q.W., C.W., H.H., W.C. and K.Y. contributed to the data collection, structural determination, analysis and discussion. J.D. conceived the study and wrote the paper.

Corresponding authors

Correspondence to Kaige Yan or Jiamu Du.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Plants thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 ATPase activity assay and Circular Dichroism (CD) spectroscopy.

a. Principle of ATPase activity assay. b. The ATPase activity of DDM1, data are mean ± SD (n = 3 independent experiments), two-tailed Student’s t-test was applied, *** indicates P-value < 0.001. Green dots represent individual data points. The P values are: no NCP, 6.3 × 10−8; no DDM1, 1.1 × 10−7; no DDM1/NCP, 4.6 × 10−7; no DDM1/NCP/ATP, 1.8 × 10−8. c. The SDS-PAGE of purified AtDDM1-ΔN25 (ΔN) and full-length protein (FL) stained by Coomassie brilliant blue. The proteins were purified more than 3 times with similar result. d. The ATPase activity of DDM1 FL and ΔN. Data are mean ± SD (n = 3 independent experiments), two-tailed Student’s t-test was applied with P-value of 0.14 which is greater than 0.05, suggesting no statistical significance (ns) between the two datasets. Green dots represent individual data points. e. CD spectra of AtDDM1-ΔN25, full-length (FL), and the mutants at wavelength 200–240 nm. All the proteins possess similar CD patterns, suggesting a well-folded state with similar secondary structure components.

Source data

Extended Data Fig. 2 Cryo-EM structure analysis of the DDM1-NCP complex in ADP-bound state.

a. The SDS-PAGE of purified AtDDM1-ΔN25 stained with Coomassie brilliant blue. The protein was purified more than 3 times with similar result. b. Flow chart of cryo-EM data processing. c. Representative micrograph of cryo-EM sample. Scale bar: 50 nm. d. Representative 2D class averages. e. Local resolution map. f. The gold-standard FSC curves calculated between two halves of datasets. g. Angular distribution of particle projections of ADP-complex.

Source data

Extended Data Fig. 3 Cryo-EM maps for representative regions of DDM1-NCP complex in ADP bound form.

a-h. Electron density maps showed the fitting of ADP (a) and representative protein regions (b-h).

Extended Data Fig. 4 Comparison of the structures of the DDM1-NCP complex and ScSnf2-NCP complex.

a. Superimposition of the structures of the DDM1-NCP complex (color-coded) and ScSnf2-NCP complex (gray, PDB code: 5Z3O). b. Comparison of the structures of the H4 tail binding pockets in DDM1-NCP complex and the ScSnf2-NCP complex (gray, PDB code: 5X0Y). Acidic residues of structures surrounding the H4-binding pocket are shown as sticks and labelled, respectively.

Extended Data Fig. 5 Cryo-EM structure analysis of the DDM1-NCP complex in nucleotide-free state.

a. Flow chart of cryo-EM data processing. b. Representative micrograph of cryo-EM sample. Scale bar: 50 nm. c. Representative 2D class averages. d. Local resolution map. e. The gold-standard FSC curves calculated between two halves of datasets. f. Angular distribution of particle projections of the DDM1-nucleosome complex.

Extended Data Fig. 6 Cryo-EM structure analysis of the DDM1-NCP complex in ADP-BeFx-complexed states.

a. Flow chart of cryo-EM data processing. b. Representative micrograph of cryo-EM sample. Scale bar: 50 nm. c. Representative 2D class averages. d. The gold-standard FSC curves calculated between two halves of datasets.

Extended Data Fig. 7 Cryo-EM maps.

a-s. Electron density maps showed the fitting of representative regions of DDM1-NCP complex in nucleotide-free form (a-g), ADP-BeFx-bound conformation (h-o), and free Arabidopsis NCP (p-s).

Extended Data Fig. 8 Cryo-EM structure of DDM1-NCP complex in ADP-BeFx-bound state and free Arabidopsis NCP.

a. The 2DDM1-1NCP complex in ADP-BeFx-bound state with DDM1 bound to SHL2 and SHL-2. b-c. The superimpositions of the 1:1 complex (in silver) to the DDM1 at SHL2 (b) and SHL-2 (c) positions in the 2:1 complex showing very similar overall structures and NCP binding mode. d. Superimposition of the structures of the Arabidopsis NCP (in color) and the Xenopus laevis NCP (in silver, PDB code: 1AOI). The overall superimposition RMSD is only 0.56 Å.

Extended Data Fig. 9 Proposed mechanism for the directional translocation of DNA by DDM1 upon ATP binding and hydrolysis at the DDM1-NCP interface.

a. Superimposition of the structures of DDM1-NCP in nucleotide-free (color-coded) and ADP-BeFx-bound (gray) states aligned by the histone octamers. The boxed regions are enlarged for further analysis in panels b-c. b. DDM1 Lobe2-DNA interface in the purple boxed region in panel a. The directions of Lobe2 movement and DNA sliding upon ATP binding are highlighted by black and red arrows, respectively. c. Lobe1-DNA interface in the orange boxed region in panel a. d. Superimposition of the structures of DDM1-NCP in ADP-BeFx-bound (color-coded) and ADP-bound (gray) states aligned by the histone octamers. The boxed region is enlarged for further analysis in panel e. e. DDM1-DNA interface in the boxed region in panel d.

Extended Data Fig. 10 Structural comparison of DDM1-H3.1 NCP complex and DDM1-H3.3/H2A.W NCP complex (PDB code: 7UX9).

a. Superimpositions of the DDM1-NCP complexes in the nucleotide-free, ADP-BeFx-bound, and ADP-bound states, and the DDM1-H3.3/H2A.W NCP complex. The histone octamers are aligned. b. Comparison of the detailed interactions of DDM1 with histone H3.1 in ADP-bound state in this study (color-coded) and H3.3 in the H3.3/H2A.W NCP complex (gray) state. The boxed region is enlarged for further analysis in right panel, the interacting residues are shown as sticks and labelled, respectively. c. A view of residues C615 and C634 in the Lobe2 domain of DDM1 in our study. The boxed region is enlarged for further analysis in right panel, the residues are shown as sticks, cryo-EM density is shown as a gray volume.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Supplementary Video 1

Video showing the coupling of DNA movement with the domain motion of DDM1 in a chromatin remodelling cycle.

Source data

Source Data Fig. 2

Original data for Fig. 2g.

Source Data Extended Data Fig. 1

Original data for Extended Data Fig. 1b,d and unmodified gel for Extended Data Fig. 1c.

Source Data Extended Data Fig. 2

Unmodified gel for Extended Data Fig. 2a.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Zhang, Z., Hu, H. et al. Molecular basis of chromatin remodelling by DDM1 involved in plant DNA methylation. Nat. Plants 10, 374–380 (2024). https://doi.org/10.1038/s41477-024-01640-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41477-024-01640-z

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