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:

Histone deacetylation primes self-propagation of heterochromatin domains to promote epigenetic inheritance

Abstract

Heterochromatin assembly, involving histone H3 lysine-9 methylation (H3K9me), is nucleated at specific genomic sites but can self-propagate across extended domains and, indeed, generations. Self-propagation requires Clr4/Suv39h methyltransferase recruitment by pre-existing H3K9 tri-methylation (H3K9me3) to perpetuate H3K9me deposition and is dramatically affected by chromatin context. However, the mechanism priming self-propagation of heterochromatin remains undefined. We show that robust chromatin association of fission yeast class II histone deacetylase Clr3 is necessary and sufficient to support heterochromatin propagation in different chromosomal contexts. Efficient targeting of Clr3, which suppresses histone turnover and maintains H3K9me3, enables self-propagation of an ectopic heterochromatin domain via the Clr4/Suv39h read–write mechanism requiring methylated histones. The deacetylase activity of Clr3 is necessary and, when inactivated, heterochromatin propagation can be recapitulated by removing two major histone acetyltransferases. Our results show that histone deacetylation, a conserved heterochromatin feature, preserves H3K9me3 that transmits epigenetic memory for stable propagation of silenced chromatin domains through multiple generations.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Fusion of Clr3 HDAC to a chromodomain supports efficient sequence-independent localization to heterochromatin.
Fig. 2: Clr3 localization sustains self-templated heterochromatin propagation.
Fig. 3: HDAC chromatin loading is required for self-propagation of extended heterochromatin domains.
Fig. 4: Loss of the HATs Gcn5 and Mst2 enables self-propagation of heterochromatin.
Fig. 5: Effective chromatin association of Clr3 is required for heterochromatin spreading and suppression of histone turnover to preserve epigenetic memory.
Fig. 6: Clr3 localization promotes stable heterochromatin propagation at centromeres.
Fig. 7: CAS-like elements promote epigenetic inheritance of heterochromatin at the ectopic site.
Fig. 8: Direct targeting of Clr3 restores H3K9me3 density and heterochromatin propagation in mutants showing elevated histone turnover.

Similar content being viewed by others

Data availability

ChIP–chip and ChIP–seq data have been deposited in the GEO under accession number GSE184466. ChIP–seq data for Drosophila HDAC4 and H3K27me were downloaded from the NCBI Sequence Read Archive (GEO records GSE20000 and GSE49490, respectively)62. Source data are provided with this paper.

References

  1. Nicetto, D. & Zaret, K. S. Role of H3K9me3 heterochromatin in cell identity establishment and maintenance. Curr. Opin. Genet. Dev. 55, 1–10 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Elgin, S. C. & Reuter, G. Position-effect variegation, heterochromatin formation, and gene silencing in Drosophila. Cold Spring Harb. Perspect. Biol. 5, a017780 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  4. Janssen, A., Colmenares, S. U. & Karpen, G. H. Heterochromatin: guardian of the genome. Annu. Rev. Cell Dev. Biol. 34, 265–288 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Hall, I. M. et al. Establishment and maintenance of a heterochromatin domain. Science 297, 2232–2237 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Nakayama, J., Klar, A. J. & Grewal, S. I. A chromodomain protein, Swi6, performs imprinting functions in fission yeast during mitosis and meiosis. Cell 101, 307–317 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Hathaway, N. A. et al. Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Audergon, P. N. et al. Epigenetics. Restricted epigenetic inheritance of H3K9 methylation. Science 348, 132–135 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ragunathan, K., Jih, G. & Moazed, D. Epigenetics. Epigenetic inheritance uncoupled from sequence-specific recruitment. Science 348, 1258699 (2015).

    Article  PubMed  Google Scholar 

  10. Zhang, K., Mosch, K., Fischle, W. & Grewal, S. I. Roles of the Clr4 methyltransferase complex in nucleation, spreading and maintenance of heterochromatin. Nat. Struct. Mol. Biol. 15, 381–388 (2008).

    Article  CAS  PubMed  Google Scholar 

  11. Lee, N. N. et al. Mtr4-like protein coordinates nuclear RNA processing for heterochromatin assembly and for telomere maintenance. Cell 155, 1061–1074 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tashiro, S., Asano, T., Kanoh, J. & Ishikawa, F. Transcription-induced chromatin association of RNA surveillance factors mediates facultative heterochromatin formation in fission yeast. Genes Cells 18, 327–339 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Volpe, T. A. et al. Regulation of heterochromatic silencing and histone H3 lysine-9 methylation by RNAi. Science 297, 1833–1837 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Zofall, M. et al. RNA elimination machinery targeting meiotic mRNAs promotes facultative heterochromatin formation. Science 335, 96–100 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Al-Sady, B., Madhani, H. D. & Narlikar, G. J. Division of labor between the chromodomains of HP1 and Suv39 methylase enables coordination of heterochromatin spread. Mol. Cell 51, 80–91 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Muller, M. M., Fierz, B., Bittova, L., Liszczak, G. & Muir, T. W. A two-state activation mechanism controls the histone methyltransferase Suv39h1. Nat. Chem. Biol. 12, 188–193 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Holla, S. et al. Positioning heterochromatin at the nuclear periphery suppresses histone turnover to promote epigenetic inheritance. Cell 180, 150–164 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Aygun, O., Mehta, S. & Grewal, S. I. HDAC-mediated suppression of histone turnover promotes epigenetic stability of heterochromatin. Nat. Struct. Mol. Biol. 20, 547–554 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Taneja, N. et al. SNF2 family protein Fft3 suppresses nucleosome turnover to promote epigenetic inheritance and proper replication. Mol. Cell 66, 50–62 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yamada, T., Fischle, W., Sugiyama, T., Allis, C. D. & Grewal, S. I. The nucleation and maintenance of heterochromatin by a histone deacetylase in fission yeast. Mol. Cell 20, 173–185 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Sugiyama, T. et al. SHREC, an effector complex for heterochromatic transcriptional silencing. Cell 128, 491–504 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Job, G. et al. SHREC silences heterochromatin via distinct remodeling and deacetylation modules. Mol. Cell 62, 207–221 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fischer, T. et al. Diverse roles of HP1 proteins in heterochromatin assembly and functions in fission yeast. Proc. Natl Acad. Sci. USA 106, 8998–9003 (2009).

  24. Sadaie, M. et al. Balance between distinct HP1 family proteins controls heterochromatin assembly in fission yeast. Mol. Cell. Biol. 28, 6973–6988 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Leopold, K., Stirpe, A. & Schalch, T. Transcriptional gene silencing requires dedicated interaction between HP1 protein Chp2 and chromatin remodeler Mit1. Genes Dev. 33, 565–577 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cam, H. P., Noma, K., Ebina, H., Levin, H. L. & Grewal, S. I. Host genome surveillance for retrotransposons by transposon-derived proteins. Nature 451, 431–436 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Greenstein, R. A. et al. Noncoding RNA-nucleated heterochromatin spreading is intrinsically labile and requires accessory elements for epigenetic stability. eLife 7, 32948 (2018).

    Article  Google Scholar 

  28. Brehm, A. et al. Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature 391, 597–601 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Raiymbek, G. et al. An H3K9 methylation-dependent protein interaction regulates the non-enzymatic functions of a putative histone demethylase. eLife 9, 53155 (2020).

    Article  Google Scholar 

  30. Schultz, D. C., Friedman, J. R. & Rauscher, F. J. 3rd Targeting histone deacetylase complexes via KRAB-zinc finger proteins: the PHD and bromodomains of KAP-1 form a cooperative unit that recruits a novel isoform of the Mi-2alpha subunit of NuRD. Genes Dev. 15, 428–443 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Petrie, V. J., Wuitschick, J. D., Givens, C. D., Kosinski, A. M. & Partridge, J. F. RNA interference (RNAi)-dependent and RNAi-independent association of the Chp1 chromodomain protein with distinct heterochromatic loci in fission yeast. Mol. Cell. Biol. 25, 2331–2346 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schalch, T. et al. High-affinity binding of Chp1 chromodomain to K9 methylated histone H3 is required to establish centromeric heterochromatin. Mol. Cell 34, 36–46 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cutter DiPiazza, A. R. et al. Spreading and epigenetic inheritance of heterochromatin require a critical density of histone H3 lysine 9 tri-methylation. Proc. Natl Acad. Sci. USA 118, e2100699118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Thon, G., Cohen, A. & Klar, A. J. Three additional linkage groups that repress transcription and meiotic recombination in the mating-type region of Schizosaccharomyces pombe. Genetics 138, 29–38 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Canzio, D. et al. Chromodomain-mediated oligomerization of HP1 suggests a nucleosome-bridging mechanism for heterochromatin assembly. Mol. Cell 41, 67–81 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Noma, K. et al. RITS acts in cis to promote RNA interference-mediated transcriptional and post-transcriptional silencing. Nat. Genet. 36, 1174–1180 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Obersriebnig, M. J., Pallesen, E. M., Sneppen, K., Trusina, A. & Thon, G. Nucleation and spreading of a heterochromatic domain in fission yeast. Nat. Commun. 7, 11518 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Grewal, S. I. & Klar, A. J. Chromosomal inheritance of epigenetic states in fission yeast during mitosis and meiosis. Cell 86, 95–101 (1996).

    Article  CAS  PubMed  Google Scholar 

  39. Bjerling, P. et al. Functional divergence between histone deacetylases in fission yeast by distinct cellular localization and in vivo specificity. Mol. Cell. Biol. 22, 2170–2181 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Helmlinger, D. et al. The S. pombe SAGA complex controls the switch from proliferation to sexual differentiation through the opposing roles of its subunits Gcn5 and Spt8. Genes Dev. 22, 3184–3195 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wang, Y. et al. Histone H3 lysine 14 acetylation is required for activation of a DNA damage checkpoint in fission yeast. J. Biol. Chem. 287, 4386–4393 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Svensson, J. P. et al. A nucleosome turnover map reveals that the stability of histone H4 Lys20 methylation depends on histone recycling in transcribed chromatin. Genome Res. 25, 872–883 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Murawska, M. et al. The histone chaperone FACT facilitates heterochromatin spreading by regulating histone turnover and H3K9 methylation states. Cell Rep. 37, 109944 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lejeune, E. et al. The chromatin-remodeling factor FACT contributes to centromeric heterochromatin independently of RNAi. Curr. Biol. 17, 1219–1224 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ayoub, N. et al. A novel jmjC domain protein modulates heterochromatization in fission yeast. Mol. Cell. Biol. 23, 4356–4370 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zofall, M. & Grewal, S. I. Swi6/HP1 recruits a JmjC domain protein to facilitate transcription of heterochromatic repeats. Mol. Cell 22, 681–692 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Shimada, A. et al. Phosphorylation of Swi6/HP1 regulates transcriptional gene silencing at heterochromatin. Genes Dev. 23, 18–23 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bao, K., Shan, C. M., Moresco, J., Yates, J. 3rd & Jia, S. Anti-silencing factor Epe1 associates with SAGA to regulate transcription within heterochromatin. Genes Dev. 33, 116–126 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kasten, M. et al. Tandem bromodomains in the chromatin remodeler RSC recognize acetylated histone H3 Lys14. EMBO J. 23, 1348–1359 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Morrison, E. A. et al. DNA binding drives the association of BRG1/hBRM bromodomains with nucleosomes. Nat. Commun. 8, 16080 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Regadas, I. et al. A unique histone 3 lysine 14 chromatin signature underlies tissue-specific gene regulation. Mol. Cell 81, 1766–1780 (2021).

    Article  CAS  PubMed  Google Scholar 

  52. Oya, E. et al. H3K14 ubiquitylation promotes H3K9 methylation for heterochromatin assembly. EMBO Rep. 20, e48111 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Stirpe, A. et al. SUV39 SET domains mediate crosstalk of heterochromatic histone marks. eLife 10, 62682 (2021).

    Article  Google Scholar 

  54. Ekwall, K., Olsson, T., Turner, B. M., Cranston, G. & Allshire, R. C. Transient inhibition of histone deacetylation alters the structural and functional imprint at fission yeast centromeres. Cell 91, 1021–1032 (1997).

    Article  CAS  PubMed  Google Scholar 

  55. Grewal, S. I., Bonaduce, M. J. & Klar, A. J. Histone deacetylase homologs regulate epigenetic inheritance of transcriptional silencing and chromosome segregation in fission yeast. Genetics 150, 563–576 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Barrales, R. R., Forn, M., Georgescu, P. R., Sarkadi, Z. & Braun, S. Control of heterochromatin localization and silencing by the nuclear membrane protein Lem2. Genes Dev. 30, 133–148 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Deal, R. B., Henikoff, J. G. & Henikoff, S. Genome-wide kinetics of nucleosome turnover determined by metabolic labeling of histones. Science 328, 1161–1164 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Chory, E. J. et al. Nucleosome turnover regulates histone methylation patterns over the genome. Mol. Cell 73, 61–72 (2019).

    Article  CAS  PubMed  Google Scholar 

  59. Dobosy, J. R. & Selker, E. U. Emerging connections between DNA methylation and histone acetylation. Cell. Mol. Life Sci. 58, 721–727 (2001).

    Article  CAS  PubMed  Google Scholar 

  60. Coleman, R. T. & Struhl, G. Causal role for inheritance of H3K27me3 in maintaining the OFF state of a Drosophila HOX gene. Science 356, eaai8236 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Laprell, F., Finkl, K. & Muller, J. Propagation of Polycomb-repressed chromatin requires sequence-specific recruitment to DNA. Science 356, 85–88 (2017).

    Article  CAS  PubMed  Google Scholar 

  62. Negre, N. et al. A cis-regulatory map of the Drosophila genome. Nature 471, 527–531 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Braunstein, M., Rose, A. B., Holmes, S. G., Allis, C. D. & Broach, J. R. Transcriptional silencing in yeast is associated with reduced nucleosome acetylation. Genes Dev. 7, 592–604 (1993).

    Article  CAS  PubMed  Google Scholar 

  64. Jeppesen, P. & Turner, B. M. The inactive X chromosome in female mammals is distinguished by a lack of histone H4 acetylation, a cytogenetic marker for gene expression. Cell 74, 281–289 (1993).

    Article  CAS  PubMed  Google Scholar 

  65. Walther, M. et al. Heterochromatin formation in Drosophila requires genome-wide histone deacetylation in cleavage chromatin before mid-blastula transition in early embryogenesis. Chromosoma 129, 83–98 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Tang, W. W. et al. A unique gene regulatory network resets the human germline epigenome for development. Cell 161, 1453–1467 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Rowe, H. M. et al. KAP1 controls endogenous retroviruses in embryonic stem cells. Nature 463, 237–240 (2010).

    Article  CAS  PubMed  Google Scholar 

  68. Cam, H. P. et al. Comprehensive analysis of heterochromatin- and RNAi-mediated epigenetic control of the fission yeast genome. Nat. Genet. 37, 809–819 (2005).

    Article  CAS  PubMed  Google Scholar 

  69. Chen, S., Zhou, Y., Chen, Y. & Gu, J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884–i890 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Laboratory of Biochemistry and Molecular Biology, in particular the Grewal laboratory for helpful discussions, A. Cutter Dipiazza for strain constructions, C. Jayakumar and J. Dhakshnamoorthy for technical help and J. Barrowman for editing the manuscript. This study used the Helix Systems and Biowulf Linux cluster at the National Institutes of Health and the PomBase genome database. This work was supported by the Intramural Research Program of the National Institutes of Health, National Cancer Institute.

Author information

Authors and Affiliations

Authors

Contributions

S.I.S.G. and M.Z. conceived the project. M.Z., R.S. and S.H. performed experiments. R.S. performed Clr3 ChIP–seq. S.H. helped in performing histone turnover experiments. M.Z. performed all other experiments. D.W. performed bioinformatic analyses. All authors contributed to data interpretation. S.I.S.G. and M.Z. wrote the manuscript.

Corresponding author

Correspondence to Shiv I. S. Grewal.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural and Molecular Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Carolina Perdigoto, in collaboration with the Nature Structural & Molecular Biology team. Peer reviewer reports are available.

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 Chromodomain-coupled chimeric Clr3 protein is functional.

(a) Western blot analysis of HA epitope-tagged Clr3 carrying one or two copies of the Chp1 chromodomain fused at its carboxy-terminus and expressed under the control of the native clr3 promoter. Coomassie staining is shown as a loading control. Representative of two independent analyses is shown. (b) ChIP analysis of Clr3-CDx2 localization at the silent mat region in WT and clr4∆ cells. Quantitative duplex PCR analysis was used to calculate fold enrichment values. The region analyzed by ChIP is marked by the black bar in ‘c’. (c) Serial dilution analysis of heterochromatic repression of the mat2P::ura4+ reporter. The indicated strains were spotted on non-selective (NS), uracil depleted (-Uracil) or counter-selective medium containing FOA (FOA). Colonies grown on PMG minimal medium were stained with iodine vapor to detect haploid meiosis. Note that strains expressing chromodomain-coupled Clr3 are proficient in heterochromatic silencing at the silent mat region. As a control, cells carrying a mutation in the catalytic site of Clr3 (clr3D232N) show defective silencing, which is indicated by growth on medium lacking uracil, loss of viability on FOA medium and dark iodine staining. (d) Serial dilution analyses to test if chromodomain-coupled Clr3 can bypass the Swi6 requirement for heterochromatic silencing. In addition to mat2P::ura4+ expression, haploid meiosis was analyzed by iodine staining. (e) ChIP analysis of Clr3 localization at the silent mat region. Results of quantitative duplex PCR analysis are shown. Mean fold enrichments (MFE) and SEM from 3 or 2 measurements are indicated in b and e.

Source data

Extended Data Fig. 2 Clr3-CD dosage determines the efficacy of self-templated heterochromatin assembly.

(a) Western blot analysis of Clr3-CDx1 expression driven by the native clr3 or adh11 promoter. Extracts from cells expressing untagged Clr3 or Clr3-HA without the chromodomain fusion are included for comparison. Coomassie staining is shown as a loading control. Representative of two independent analyses is shown. (b) ade6+ expression in tetR-clr4ON or tetR-clr4OFF cells carrying the indicated clr3 allele was assayed by serial dilution on low adenine medium (top). Colonies formed by tetR-clr4OFF cells carrying either WT clr3, clr3-CDx1 or adh11::clr3-CDx1 on low adenine medium are shown (bottom). The presence of red colonies indicates stable propagation of ectopic heterochromatin at the ade6+ reporter.

Source data

Extended Data Fig. 3 Enhanced chromatin association of Clr3 HDAC stabilizes heterochromatin at endogenous loci.

(a) Schematic showing the silent mat region with the RNAi-targeted cenH element replaced by the ura4+ reporter (K∆::ura4+). In the absence of RNAi targeted cenH, de novo heterochromatin assembly is compromised, and heterochromatin propagation requires the read-write mechanism involving Clr4Suv39h recruitment by the parental H3K9me3. Cells carrying the silenced K∆::ura4+ OFF state are passaged for 10 or 30 generations in non-selective media and then serial dilutions are plated onto non-selective, -Uracil and FOA media to assay propagation of the heterochromatic state. Cells expressing chromodomain-coupled Clr3, in particular Clr3-CDx2, propagated heterochromatin more stably as compared to WT Clr3. (b) ChIP analysis of the indicated Clr3 proteins at facultative heterochromatin islands. (c) ChIP analyses of H3K9me levels at facultative heterochromatin islands in cells expressing WT Clr3 or Clr3 fused to one or two chromodomains. Relative ChIP enrichment values at the indicated loci, as compared to control leu1, that are shown in ‘b’ and ‘c’ were determined using quantitative duplex PCR. Mean fold enrichments (MFE) and SEM of two experiments are indicated.

Source data

Extended Data Fig. 4 Sequence-independent epigenetic inheritance of an ectopic heterochromatin domain in cells lacking histone acetyltransferases.

(a) Schematic showing the strategy used to assay epigenetic inheritance of an ectopic heterochromatin domain in cells lacking Mst2 and Gcn5. The self-propagation of ectopic heterochromatin domains assembled by TetR-Clr4 (tetR-clr4ON) was investigated upon the release of tethered Clr4 (tetR-clr4OFF) in the indicated single and double mutant strains. Heterochromatin persistence was measured by plating cells on low adenine medium to assay tetO-ade6+ expression. Red colony coloration indicates ade6+ repression, while white color indicates expression. (b) Close-up of colonies formed by WT and mst2∆ gcn5∆ cells on low adenine medium. The maintenance of the heterochromatic state at tetO-ade6+ in tetR-clr4OFF cells lacking Mst2 and Gcn5 is indicated by red-colored colonies, as compared to white colonies formed by WT background cells.

Extended Data Fig. 5 H3K9me and Clr3-CDx2 distribution across the ectopic heterochromatin domain.

Results of ChIP-chip analyses of H3K9me3 and Clr3-CDx2 are plotted. A strain expressing untagged Clr3 is included as a control.

Extended Data Fig. 6 Chromodomain-coupled Clr3 affects heterochromatin domains at pericentromeric repeats and at telomeres.

(a) Clr3-Myc distribution along S. pombe chromosomes as determined by ChIP-seq analysis. Note prominent peaks of Clr3 mapping to the mat locus and rDNA, in addition to its localization at centromeres, telomeres and other loci including Tf2 retroelements. (b) Clr3-CDx2 expression suppresses histone turnover at pericentromeric regions. ChIP-chip analysis of H3K9me distribution and incorporation of pulse-expressed H3-T7 at centromere 1 in cells carrying either WT clr3 or clr3-CDx2 allele. Replication-independent turnover of H3 was measured in cells blocked at the G2/M boundary. The central core domain (cnt) of centromere that contains Cenp-A does not incorporate histone H3. Note that cells expressing Clr3-CDx2 show lower histone turnover as compared to WT. (c) ChIP-chip analysis of Clr3-CDx2 and H3K9me3 distribution at telomere 1R. Cells expressing Clr3-CDx2 show enhanced spreading of H3K9me3 compared to WT cells.

Extended Data Fig. 7 Effective chromatin association of Clr3 HDAC, or prevention of histone acetylation, restores heterochromatic silencing in mutants showing high histone turnover.

The enhanced recruitment of chromodomain-coupled Clr3, or loss of both Mst2 and Gcn5 HATs, mitigates defective heterochromatic silencing at the silent mat region in cells lacking Amo1, involved in the nuclear peripheral tethering of heterochromatin, or the FACT histone chaperone component Pob3, implicated in suppression of histone turnover. Serial dilutions on non-selective (NS) and FOA-containing medium (FOA) were used to assay expression of the mat2P::ura4+ reporter gene at the silent mat region.

Extended Data Fig. 8 Drosophila class II HDAC, HDAC4, preferentially localizes to H3K27me3-coated domains silenced by the Polycomb proteins.

Results of ChIP-seq analysis of HDAC4 and H3K27me3 performed by Negre et al62 (GEO accession number: GSE49490 and GSE20000 respectively) are plotted. Note that major HDAC4 peaks map to the polycomb response elements (PREs; indicated by red triangles) located in the center of individual H3K27me3 domains. HDAC4 distribution resembles Clr3 class II HDAC localization at the silent mat region in S. pombe (see Fig. 6a). Similar to PRE elements in Drosophila, CAS elements, including the CAS element bound by ATF-CREB family proteins Atf1/Pcr1 implicated in heterochromatin assembly20, show prominent Clr3 HDAC peaks.

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables 1–2

List of strains and primers used in this study.

Source data

Source Data Fig. 1

Unprocessed phosphorimages.

Source Data Fig. 1

Phosphorimage quantification.

Source Data Fig. 2

Unprocessed phosphorimages.

Source Data Fig. 2

Phosphorimage quantification.

Source Data Fig. 3

Unprocessed phosphorimages.

Source Data Fig. 3

Phosphorimage quantification.

Source Data Fig. 4

Unprocessed phosphorimages and western blot.

Source Data Fig. 4

Phosphorimage quantification.

Source Data Fig. 6

Unprocessed phosphorimages.

Source Data Fig. 6

Phosphorimage quantification.

Source Data Fig. 7

Unprocessed phosphorimages.

Source Data Fig. 7

Phosphorimage quantification.

Source Data Fig. 8

Unprocessed phosphorimages.

Source Data Fig. 8

Phosphorimage quantification.

Source Data Extended Data Fig. 1

Unprocessed phosphorimages and western blot.

Source Data Extended Data Fig. 1

Phosphorimage quantification.

Source Data Extended Data Fig. 2

Unprocessed western blot.

Source Data Extended Data Fig. 3

Unprocessed phosphorimages.

Source Data Extended Data Fig. 3

Phosphorimage quantification.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zofall, M., Sandhu, R., Holla, S. et al. Histone deacetylation primes self-propagation of heterochromatin domains to promote epigenetic inheritance. Nat Struct Mol Biol 29, 898–909 (2022). https://doi.org/10.1038/s41594-022-00830-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-022-00830-7

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