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:

The chromatin remodeler DDM1 prevents transposon mobility through deposition of histone variant H2A.W

This article has been updated

Abstract

Mobile transposable elements (TEs) not only participate in genome evolution but also threaten genome integrity. In healthy cells, TEs that encode all of the components that are necessary for their mobility are specifically silenced, yet the precise mechanism remains unknown. Here, we characterize the mechanism used by a conserved class of chromatin remodelers that prevent TE mobility. In the Arabidopsis chromatin remodeler DECREASE IN DNA METHYLATION 1 (DDM1), we identify two conserved binding domains for the histone variant H2A.W, which marks plant heterochromatin. DDM1 is necessary and sufficient for the deposition of H2A.W onto potentially mobile TEs, yet does not act on TE fragments or host protein-coding genes. DDM1-mediated H2A.W deposition changes the properties of chromatin, resulting in the silencing of TEs and, therefore, prevents their mobility. This distinct mechanism provides insights into the interplay between TEs and their host in the contexts of evolution and disease, and potentiates innovative strategies for targeted gene silencing.

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: DDM1 binds directly to H2A.W through two conserved regions.
Fig. 2: DDM1 is necessary for H2A.W deposition on TEs.
Fig. 3: DDM1 changes the chromatin state of TEs.
Fig. 4: DDM1 binding to H2A.W is required for TE silencing.
Fig. 5: DDM1 deposits H2A.W over TEs to mediate silencing.
Fig. 6: Expression of TEs in ddm1 and other mutants.
Fig. 7: DNA methylation of TEs in ddm1 and F1 Col-0 × ddm1 plants.
Fig. 8: DDM1 silences mobile TEs.

Similar content being viewed by others

Data availability

The deep-sequencing (ChIP–seq, RNA-seq and EM-seq) data that support the findings of this study have been deposited to the NCBI Gene Expression Omnibus (GEO) under the accession number GSE150436 and the SRA (BioProject ID: PRJNA689609). All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

Custom code used to process and analyse the deep-sequencing data, as described in the Methods, is available from the corresponding author on request.

Change history

  • 04 May 2022

    In the version of this article initially published, the Statistical Source Data files for Figs. 2d, 5b,c, and 6 had misplaced decimal points. The files have now been corrected in the HTML version of the article.

References

  1. Cosby, R. L., Chang, N. C. & Feschotte, C. Host-transposon interactions: conflict, cooperation, and cooption. Genes Dev. 33, 1098–1116 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bourque, G. et al. Ten things you should know about transposable elements. Genome Biol. 19, 199 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Wicker, T. et al. A unified classification system for eukaryotic transposable elements. Nat. Rev. Genet. 8, 973–982 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Gagnier, L., Belancio, V. P. & Mager, D. L. Mouse germ line mutations due to retrotransposon insertions. Mob. DNA 10, 15 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Kidwell, M. G. & Lisch, D. Transposable elements as sources of variation in animals and plants. Proc. Natl Acad. Sci USA 94, 7704–7711 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chuong, E. B., Elde, N. C. & Feschotte, C. Regulatory evolution of innate immunity through co-option of endogenous retroviruses. Science 351, 1083–1087 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jangam, D., Feschotte, C. & Betran, E. Transposable element domestication as an adaptation to evolutionary conflicts. Trends Genet. 33, 817–831 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Velanis, C. N. et al. The domesticated transposase ALP2 mediates formation of a novel Polycomb protein complex by direct interaction with MSI1, a core subunit of Polycomb Repressive Complex 2 (PRC2). PLoS Genet. 16, e1008681 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rishishwar, L. et al. Evidence for positive selection on recent human transposable element insertions. Gene 675, 69–79 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Boissinot, S., Davis, J., Entezam, A., Petrov, D. & Furano, A. V. Fitness cost of LINE-1 (L1) activity in humans. Proc. Natl Acad. Sci. USA 103, 9590–9594 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Payer, L. M. & Burns, K. H. Transposable elements in human genetic disease. Nat. Rev. Genet. 20, 760–772 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Czech, B. et al. piRNA-guided genome defense: from biogenesis to silencing. Annu. Rev. Genet. 52, 131–157 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Handler, D. et al. The genetic makeup of the Drosophila piRNA pathway. Mol. Cell 50, 762–777 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ozata, D. M., Gainetdinov, I., Zoch, A., O’Carroll, D. & Zamore, P. D. PIWI-interacting RNAs: small RNAs with big functions. Nat. Rev. Genet. 20, 89–108 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Derkacheva, M. et al. Arabidopsis MSI1 connects LHP1 to PRC2 complexes. EMBO J. 32, 2073–2085 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Grewal, S. I. & Jia, S. Heterochromatin revisited. Nat. Rev. Genet. 8, 35–46 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. 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 

  18. Nuthikattu, S. et al. The initiation of epigenetic silencing of active transposable elements is triggered by RDR6 and 21-22 nucleotide small interfering RNAs. Plant Physiol. 162, 116–131 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Oberlin, S., Sarazin, A., Chevalier, C., Voinnet, O. & Mari-Ordonez, A. A genome-wide transcriptome and translatome analysis of Arabidopsis transposons identifies a unique and conserved genome expression strategy for Ty1/Copia retroelements. Genome Res. 27, 1549–1562 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Senti, K. A. & Brennecke, J. The piRNA pathway: a fly’s perspective on the guardian of the genome. Trends Genet. 26, 499–509 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yu, R., Wang, X. & Moazed, D. Epigenetic inheritance mediated by coupling of RNAi and histone H3K9 methylation. Nature 558, 615–619 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kato, M., Miura, A., Bender, J., Jacobsen, S. E. & Kakutani, T. Role of CG and non-CG methylation in immobilization of transposons in Arabidopsis. Curr. Biol. 13, 421–426 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Mirouze, M. et al. Selective epigenetic control of retrotransposition in Arabidopsis. Nature 461, 427–430 (2009).

    Article  CAS  PubMed  Google Scholar 

  24. Stroud, H. et al. DNA methyltransferases are required to induce heterochromatic re-replication in Arabidopsis. PLoS Genet. 8, e1002808 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Corem, S. et al. Redistribution of CHH methylation and small interfering RNAs across the genome of tomato ddm1 mutants. Plant Cell 30, 1628–1644 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Higo, H. et al. DDM1 (decrease in DNA methylation) genes in rice (Oryza sativa). Mol. Genet. Genomics 287, 785–792 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Hirochika, H., Okamoto, H. & Kakutani, T. Silencing of retrotransposons in Arabidopsis and reactivation by the ddm1 mutation. Plant Cell 12, 357–369 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Mittelsten Scheid, O., Afsar, K. & Paszkowski, J. Release of epigenetic gene silencing by trans-acting mutations in Arabidopsis. Proc. Natl Acad. Sci. USA 95, 632–637 (1998).

    Article  CAS  PubMed  Google Scholar 

  30. Singer, T., Yordan, C. & Martienssen, R. A. Robertson’s mutator transposons in A. thaliana are regulated by the chromatin-remodeling gene Decrease in DNA Methylation (DDM1). Genes Dev. 15, 591–602 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tan, F. et al. DDM1 represses noncoding RNA expression and RNA-directed DNA methylation in heterochromatin. Plant Physiol. 177, 1187–1197 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gendrel, A. V., Lippman, Z., Yordan, C., Colot, V. & Martienssen, R. A. Dependence of heterochromatic histone H3 methylation patterns on the Arabidopsis gene DDM1. Science 297, 1871–1873 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Soppe, W. J. et al. DNA methylation controls histone H3 lysine 9 methylation and heterochromatin assembly in Arabidopsis. EMBO J. 21, 6549–6559 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lippman, Z., May, B., Yordan, C., Singer, T. & Martienssen, R. Distinct mechanisms determine transposon inheritance and methylation via small interfering RNA and histone modification. PLoS Biol. 1, E67 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Stroud, H. et al. Non-CG methylation patterns shape the epigenetic landscape in Arabidopsis. Nat. Struct. Mol. Biol. 21, 64–72 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. 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 

  37. Basenko, E. Y., Kamei, M., Ji, L., Schmitz, R. J. & Lewis, Z. A. The LSH/DDM1 homolog MUS-30 is required for genome stability, but not for DNA methylation in Neurospora crassa. PLoS Genet. 12, e1005790 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Dunican, D. S. et al. Lsh regulates LTR retrotransposon repression independently of Dnmt3b function. Genome Biol. 14, R146 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Dennis, K., Fan, T., Geiman, T., Yan, Q. & Muegge, K. Lsh, a member of the SNF2 family, is required for genome-wide methylation. Genes Dev. 15, 2940–2944 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhu, H. et al. Lsh is involved in de novo methylation of DNA. EMBO J. 25, 335–345 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ren, J., Finney, R., Ni, K., Cam, M. & Muegge, K. The chromatin remodeling protein Lsh alters nucleosome occupancy at putative enhancers and modulates binding of lineage specific transcription factors. Epigenetics 14, 277–293 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

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

  43. Lei, B. et al. A Synthetic approach to reconstruct the evolutionary and functional innovations of the plant histone variant H2A.W. Curr. Biol. 31, 182–191.e5 (2021).

  44. Yelagandula, R. et al. The histone variant H2A.W defines heterochromatin and promotes chromatin condensation in Arabidopsis. Cell 158, 98–109 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Osakabe, A. et al. Histone H2A variants confer specific properties to nucleosomes and impact on chromatin accessibility. Nucleic Acids Res. 46, 7675–7685 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mizuguchi, G. et al. ATP-driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex. Science 303, 343–348 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Papamichos-Chronakis, M., Watanabe, S., Rando, O. J. & Peterson, C. L. Global regulation of H2A.Z localization by the INO80 chromatin-remodeling enzyme is essential for genome integrity. Cell 144, 200–213 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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 

  49. Sengoku, T., Nureki, O., Nakamura, A., Kobayashi, S. & Yokoyama, S. Structural basis for RNA unwinding by the DEAD-box protein Drosophila Vasa. Cell 125, 287–300 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Talbert, P. B. et al. A unified phylogeny-based nomenclature for histone variants. Epigenetics Chromatin 5, 7 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zemach, A. et al. DDM1 binds Arabidopsis methyl-CpG binding domain proteins and affects their subnuclear localization. Plant Cell 17, 1549–1558 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Workman, J. L. Nucleosome displacement in transcription. Genes Dev. 20, 2009–2017 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Li, X. et al. Mechanistic insights into plant SUVH family H3K9 methyltransferases and their binding to context-biased non-CG DNA methylation. Proc. Natl Acad. Sci. USA 115, E8793–E8802 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Deleris, A. et al. Loss of the DNA methyltransferase MET1 Induces H3K9 hypermethylation at PcG target genes and redistribution of H3K27 trimethylation to transposons in Arabidopsis thaliana. PLoS Genet. 8, e1003062 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Lanciano, S. et al. Sequencing the extrachromosomal circular mobilome reveals retrotransposon activity in plants. PLoS Genet. 13, e1006630 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Slotkin, R. K. et al. Epigenetic reprogramming and small RNA silencing of transposable elements in pollen. Cell 136, 461–472 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Rougée, M. et al. Polycomb mutant partially suppresses DNA hypomethylation–associated phenotypes in Arabidopsis. Life Sci. Alliance 4, e202000848 (2020).

  58. Thijssen, P. E. et al. Mutations in CDCA7 and HELLS cause immunodeficiency-centromeric instability-facial anomalies syndrome. Nat. Commun. 6, 7870 (2015).

    Article  CAS  PubMed  Google Scholar 

  59. Ni, K. et al. LSH mediates gene repression through macroH2A deposition. Nat. Commun. 11, 5647 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Douet, J. et al. MacroH2A histone variants maintain nuclear organization and heterochromatin architecture. J. Cell Sci. 130, 1570–1582 (2017).

    CAS  PubMed  Google Scholar 

  61. Cohen, J. Statistical power analysis. Curr. Dir. Psychol. Sci. 1, 98–101 (1992).

    Article  Google Scholar 

  62. Lister, R. et al. Highly integrated single-base resolution maps of the epigenome in Arabidopsis. Cell 133, 523–536 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kawakatsu, T. et al. Epigenomic diversity in a global collection of Arabidopsis thaliana accessions. Cell 166, 492–505 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. 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 

  65. Arimura, Y., Tachiwana, H., Oda, T., Sato, M. & Kurumizaka, H. Structural analysis of the hexasome, lacking one histone H2A/H2B dimer from the conventional nucleosome. Biochemistry 51, 3302–3309 (2012).

    Article  CAS  PubMed  Google Scholar 

  66. Tanaka, Y. et al. Expression and purification of recombinant human histones. Methods 33, 3–11 (2004).

    Article  CAS  PubMed  Google Scholar 

  67. Tachiwana, H. et al. Structural basis of instability of the nucleosome containing a testis-specific histone variant, human H3T. Proc. Natl Acad. Sci. USA 107, 10454–10459 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Osakabe, A. et al. Vertebrate Spt2 is a novel nucleolar histone chaperone that assists in ribosomal DNA transcription. J. Cell Sci. 126, 1323–1332 (2013).

    CAS  PubMed  Google Scholar 

  69. Lorkovic, Z. J. et al. Compartmentalization of DNA damage response between heterochromatin and euchromatin is mediated by distinct H2A histone variants. Curr. Biol. 27, 1192–1199 (2017).

    Article  CAS  PubMed  Google Scholar 

  70. Ito, T., Takahashi, N., Shimura, Y. & Okada, K. A serine/threonine protein kinase gene isolated by an in vivo binding procedure using the Arabidopsis floral homeotic gene product, AGAMOUS. Plant Cell Physiol. 38, 248–258 (1997).

    Article  CAS  PubMed  Google Scholar 

  71. Wollmann, H. et al. Dynamic deposition of histone variant H3.3 accompanies developmental remodeling of the Arabidopsis transcriptome. PLoS Genet. 8, e1002658 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Yelagandula, R., Osakabe, A., Axelsson, E., Berger, F. & Kawashima, T. Genome-wide profiling of histone modifications and histone variants in Arabidopsis thaliana and Marchantia polymorpha. Methods Mol. Biol. 1610, 93–106 (2017).

    Article  CAS  PubMed  Google Scholar 

  73. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Chen, K. et al. DANPOS: dynamic analysis of nucleosome position and occupancy by sequencing. Genome Res. 23, 341–351 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    Article  CAS  PubMed  Google Scholar 

  78. Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res 4, 1521 (2015).

    Article  PubMed  CAS  Google Scholar 

  79. 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  PubMed  PubMed Central  CAS  Google Scholar 

  80. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  81. Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer, 2016).

  82. Kawashima, T. et al. Dynamic F-actin movement is essential for fertilization in Arabidopsis thaliana. eLife 3, e04501 (2014).

Download references

Acknowledgements

F.B. acknowledges support from the staff at the next-generation sequencing and PlantS facilities at the Vienna BioCenter Core Facilities (VBCF), the BioOptics facility and Molecular Biology Services from the Institute for Molecular Pathology (IMP), and the Molecular Biology Services at the GMI. We thank all members of the Berger laboratory for their technical help and H. Wang for sharing material; E. Sasaki, Z. Harvey, M. Borg, A. Marí-Ordóñez, P. Refsing Andersen and J. M. Watson for discussions and reading of the manuscript. This research was supported by the Japan Society for the Promotion of Science (JSPS) Overseas Research Fellowships (to A.O.), the Austrian Science Fund (FWF): M2539-B21 (to A.O.), P26887, P28320, P32054, P30802 and P33380 (to F.B.), and DK1238 (to B.J. and S.A.M.), the Austrian Academy of Sciences (to F.B., Z.J.L., E.A., S.A., R.P. and R.Y.), MEXT/JSPS 15H05963 and 19H00995 (to T.K.) and JST CREST JPMJCR15O1 (to T.K.).

Author information

Authors and Affiliations

Authors

Contributions

A.O., B.J. and E.A. performed the genome-wide analysis. A.O., B.J. and R.P. performed DNA methylation analysis in ddm1 mutants. A.O., E.A. and S.A. performed RT–qPCR analysis of TEs in F1 DDM1/ddm1 plants and transgenic lines. A.O. and A.L.K. performed biochemical experiments. S.A.M. performed the phylogenic analysis. R.Y. contributed the initial set of ChIP–seq data. Z.J.L. contributed the anti-H1 antibodies and T.K. contributed the design of experiments based on F1 plants. F.B. and A.O. conceived, designed and supervised the project. The manuscript was drafted by F.B., A.O. and B.J. with comments from all of the coauthors.

Corresponding author

Correspondence to Frédéric Berger.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Cell Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 Validation of recombinant DDM1 protein and H2A.X or H2A.W pulldown assay with DDM1 fragments. Related to Fig. 1.

a, SDS-PAGE analysis of purified recombinant DDM1 and DDM1 E334N. Proteins were separated by 10% SDS-PAGE and visualized by Coomassie Brilliant Blue staining. Asterisk indicates the degraded products that could not be removed after purification. (Source data shows one of three representative gels obtained). b, Schematic representation of the ATPase assay. Malachite green solution was used to detect free phosphate liberated from ATP by enzyme-catalyzed hydrolysis. c, Graphical representation of the results of ATPase assays with buffer (green), wild type DDM1 (blue), or DDM1 E334N (red), respectively. DNA concentration is expressed as moles of nucleotides. d, The crystal structure of Drosophila RNA helicase protein VASA49 together with the ATP analog (AMPPNP) (PDB ID: 2DB3). The glutamic acid residue at position 400, which coordinates a catalytic water molecule for stabilizing the interaction with ATP, is indicated. e, Sequence alignment between the Glu400 between Drosophila VASA and Arabidopsis DDM1 centered on GLU400, indicated by the blue arrow. Glu400 in VASA corresponds to Glu334 in Arabidopsis. f, h Schematic representation of the DDM1 fragments used in the pulldown assay. g, i SDS-PAGE analysis of the pulldown assay. His6-tagged DDM1 and its truncations (panel g) or GST-tagged DDM1 fragments (panel i) were incubated with histone dimers containing either H2A.X or H2A.W. After pulldown, samples were analyzed by 15% SDS-PAGE and visualized by Coomassie Brilliant Blue. Data in a, g and i represent three independent experiments. Source data are provided in Source Data Extended Data Fig. 1.

Source data

Extended Data Fig. 2 DNA methylation in ddm1. Related to Fig. 2.

Violin plots for the proportion of DNA methylation in CG, CHG, and CHH contexts on protein coding genes (left, n = 27,655), TE fragments not including TEs (middle, n = 25,695), and TEs (right, n = 3,903) in wild type Col-0 (grey) and ddm1 (red).

Extended Data Fig. 3 Genomic distribution of H1, H3K9me2 and H2A.W in ddm1. Related to Figs. 2 and 3.

a, Aggregate profile plots (top) and heatmaps (bottom) of H2A.W, H2A.X, and H1 in Col-0 (grey) and ddm1 (red) over heterochromatic regions, defined by H3K9me2 enrichment. Antibodies against H1 used in this study recognizes both H1.1 and H1.2. b, IGV genome browser snapshot of H1 (black), H2A.W (green), and H3K9me2 (blue) in wild type Col-0 and ddm1. Coloured and grey shading indicate an enriched or depleted signal, respectively. Annotated protein coding genes, TE fragments, and TEs are shown as boxes colored black, pink, or light blue, respectively. c, Aggregate profile plots (top) and heatmaps (bottom) showing distribution of H3K9me2 and H2A.W over TE fragments not including TEs in wild type and ddm1. d, Chromosomal distributions of TEs classified as group 1 (orange; n = 3,257) and group 2 (grey; n = 646) in Fig. 3a for each chromosome. Green boxes represent pericentric heterochromatin. e, f IGV genome browser snapshot of H2A.W (green), H3K9me2 (blue), H3K36me3 (orange), and H3K27me3 (purple) over TEs in wild type Col-0 and ddm1. Coloured and grey shading indicate an enriched or depleted signal, respectively.

Extended Data Fig. 4 RT-qPCR analyses of TEs in complementation lines expressing DDM1 mutant forms. Related to Fig. 4.

a, RT-PCR analyses of transgenic lines. cDNA (+RT) and DNaseI-treated RNA (-RT) were prepared from each plant and used as template for RT-PCR analyses. ACT2 was used as a control for RT-PCR. PCR products were separated by agarose gel and visualized by SYBR Green I staining. b, Bar plots showing the results of RT-qPCR as fold change of enrichment over ACT2 at TEs analyzed in Extended Data Fig. 6c,d from wild type Col-0 (grey), ddm1 (red), ddm1 expressing DDM1-3xFLAG (purple), ddm1 expressing DDM1 ∆N-3xFLAG (green), ddm1 expressing DDM1 ∆C-3xFLAG (orange), or ddm1 expressing DDM1 ∆NC-3xFLAG (brown), respectively. Presented data are means of n = 3 independent biological replicates (each biological replicate has 2 technical repeats); error bars represent SD within biological replicates. Source data are provided in Source Data Extended Data Fig. 4.

Source data

Extended Data Fig. 5 Genomic distribution of H2A.W in complementation lines. Related to Fig. 4.

a, b, d, e IGV genome browser snapshot of the distribution for H2A.W and H3K9me2 over TEs (panels a and b), protein coding gene (panel d) and TE fragment (panel e) in wild type, ddm1, ddm1 expressing DDM1 FL, DDM1 ∆N, DDM1 ∆C, and DDM1 ∆NC. c, Aggregate profile plots (top) and heatmaps (bottom) showing distribution of H2A.W over TEs in wild type, ddm1, ddm1 expressing DDM1 FL, DDM1 ∆N, DDM1 ∆C, and DDM1 ∆NC.

Extended Data Fig. 6 ChIP-qPCR analyses of H3K9me2 or H2A.W over TEs and Expression of TEs in F1 DDM1/ddm1. Related to Fig. 5.

a, b, Bar plots showing the results of ChIP-qPCR as fold change of H3K9me2 (panel a) or H2A.W (panel b) enrichment over H3 at TEs analyzed in Extended Data Fig. 6c,d from wild type Col-0 (grey), F1 DDM1/ddm1 (blue), or ddm1 (red), respectively. Presented data are means of n = 3 independent biological replicates (each biological replicate has 3 technical repeats); error bars represent SD within biological replicates. The numbers below each TE show the expression level of TEs (log2FoldChange(F1/ddm1)) compared to ddm1 mutant plants. c-e, Bar plots showing relative expression levels of TEs classified as inactivated (blue) or neutral (pink) measured by RT-qPCR from wild type Col-0 (grey), F1 DDM1/ddm1 (blue), or ddm1 (red), respectively. The expression levels of each TE are presented as a fold change over ACT2. Presented data are means of n = 3 independent biological replicates (each biological replicate has 2 technical repeats); error bars represent SD within biological replicates. The dashed lines represent 50% of the ddm1 fold change over ACT2. Source Data are provided in Source Data Extended Data Fig. 6.

Source data

Extended Data Fig. 7 Transcriptome of TEs in ddm1 and other mutants. Related to Fig. 6.

Heatmap of the z-transformed expression levels of differentially expressed TEs (n = 1,878) in mutants from wild type Col-0, suvh4;5;6, met1, and ddm1.

Extended Data Fig. 8 DNA methylation of TEs in ddm1 and F1 cross Col-0 x ddm1. Related to Fig. 7.

Heatmaps showing weighted DNA methylation levels for TEs sorted in categories ‘Inactivated’, ‘neutral’ and ‘other’ in wild type Col-0, ddm1, artificial F1 cross [(Col-0 + ddm1)/2] and F1 cross Col-0 X ddm1 for a CG, b CHG, and c CHH contexts.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2

Reporting Summary

Peer Review Information

Supplementary Tables

Supplementary Table 1: Sequence list for the DDM1 orthologues across species, which were used for alignment in this study (Fig. 1a and Supplementary Information). Supplementary Table 2: Sequences for primers used in the current study

Source data

Source Data Fig. 1

Western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Western blots.

Source Data Extended Data Fig. 4

Original gels.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Osakabe, A., Jamge, B., Axelsson, E. et al. The chromatin remodeler DDM1 prevents transposon mobility through deposition of histone variant H2A.W. Nat Cell Biol 23, 391–400 (2021). https://doi.org/10.1038/s41556-021-00658-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-021-00658-1

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