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.

  • Review Article
  • Published:

Old cogs, new tricks: the evolution of gene expression in a chromatin context

Abstract

Sophisticated gene-regulatory mechanisms probably evolved in prokaryotes billions of years before the emergence of modern eukaryotes, which inherited the same basic enzymatic machineries. However, the epigenomic landscapes of eukaryotes are dominated by nucleosomes, which have acquired roles in genome packaging, mitotic condensation and silencing parasitic genomic elements. Although the molecular mechanisms by which nucleosomes are displaced and modified have been described, just how transcription factors, histone variants and modifications and chromatin regulators act on nucleosomes to regulate transcription is the subject of considerable ongoing study. We explore the extent to which these transcriptional regulatory components function in the context of the evolutionarily ancient role of chromatin as a barrier to processes acting on DNA and how chromatin proteins have diversified to carry out evolutionarily recent functions that accompanied the emergence of differentiation and development in multicellular eukaryotes.

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: Histones and nucleosomes.
Fig. 2: Supercoiling in prokaryotes and eukaryotes.
Fig. 3: PRC1 and PRC2 in animals and plants.

Similar content being viewed by others

References

  1. Struhl, K. Fundamentally different logic of gene regulation in eukaryotes and prokaryotes. Cell 98, 1–4 (1999).

    CAS  PubMed  Google Scholar 

  2. 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).

    CAS  PubMed  Google Scholar 

  3. Talbert, P. B. & Henikoff, S. Histone variants — ancient wrap artists of the epigenome. Nat. Rev. Mol. Cell Biol. 11, 264–275 (2010).

    CAS  PubMed  Google Scholar 

  4. Schopf, J. W. Fossil evidence of Archaean life. Phil. Trans. R. Soc. B 361, 869–885 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Nutman, A. P., Bennett, V. C., Friend, C. R., Van Kranendonk, M. J. & Chivas, A. R. Rapid emergence of life shown by discovery of 3,700-million-year-old microbial structures. Nature 537, 535–538 (2016).

    CAS  PubMed  Google Scholar 

  6. Dodd, M. S. et al. Evidence for early life in Earth’s oldest hydrothermal vent precipitates. Nature 543, 60–64 (2017).

    CAS  PubMed  Google Scholar 

  7. Waldbauer, J. R., Sherman, L. S., Sumner, D. Y. & Summon, R. E. Late Archean molecular fossils from the Transvaal Supergroup record the antiquity of microbial diversity and aerobiosis. Precambrian Res. 169, 28–47 (2009).

    CAS  Google Scholar 

  8. Bengtson, S., Sallstedt, T., Belivanova, V. & Whitehouse, M. Three-dimensional preservation of cellular and subcellular structures suggests 1.6 billion-year-old crown-group red algae. PLOS Biol. 15, e2000735 (2017).

    PubMed  PubMed Central  Google Scholar 

  9. Butterfield, N. J. Bangiomorpha pubescens n. gen., n. sp.:implications for the evolution of sex, multicellularity, and the Mesoproterozoic/Neoproterozoic radiatin of eukaryotes. Paleobiology 26, 386–404 (2000).

    Google Scholar 

  10. Brown, M. W. et al. Phylogenomics places orphan protistan lineages in a novel eukaryotic super-group. Genome Biol. Evol. 10, 427–433 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Koumandou, V. L. et al. Molecular paleontology and complexity in the last eukaryotic common ancestor. Crit. Rev. Biochem. Mol. Biol. 48, 373–396 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Iyer, L. M., Anantharaman, V., Wolf, M. Y. & Aravind, L. Comparative genomics of transcription factors and chromatin proteins in parasitic protists and other eukaryotes. Int. J. Parasitol. 38, 1–31 (2008).

    CAS  PubMed  Google Scholar 

  13. Nasir, A., Kim, K. M., Da Cunha, V. & Caetano-Anolles, G. Arguments reinforcing the three-domain view of diversified cellular life. Archaea 2016, 1851865 (2016). The sources of eukaryotic protein superfold families are analysed and arguments are presented for three cellular domains, in contrast to Zaremba-Niedzwiedzka et al. (2017).

    PubMed  PubMed Central  Google Scholar 

  14. Tanny, J. C. Chromatin modification by the RNA Polymerase II elongation complex. Transcription 5, e988093 (2014).

    PubMed  Google Scholar 

  15. Zhang, T., Cooper, S. & Brockdorff, N. The interplay of histone modifications - writers that read. EMBO Rep. 16, 1467–1481 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Allshire, R. C. & Madhani, H. D. Ten principles of heterochromatin formation and function. Nat. Rev. Mol. Cell Biol. 19, 229–244 (2018).

    CAS  PubMed  Google Scholar 

  17. Sherafatian, M. & Mowla, S. J. The origins and evolutionary history of human non-coding RNA regulatory networks. J. Bioinform. Comput. Biol. 15, 1750005 (2017).

    CAS  PubMed  Google Scholar 

  18. Ishihama, A. Building a complete image of genome regulation in the model organism Escherichia coli. J. Gen. Appl. Microbiol. 63, 311–324 (2018).

    PubMed  Google Scholar 

  19. Blombach, F. & Grohmann, D. Same same but different: the evolution of TBP in archaea and their eukaryotic offspring. Transcription 8, 162–168 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Vannini, A. & Cramer, P. Conservation between the RNA polymerase I, II, and III transcription initiation machineries. Mol. Cell 45, 439–446 (2012).

    CAS  PubMed  Google Scholar 

  21. Jun, S. H., Reichlen, M. J., Tajiri, M. & Murakami, K. S. Archaeal RNA polymerase and transcription regulation. Crit. Rev. Biochem. Mol. Biol. 46, 27–40 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zentner, G. E. & Henikoff, S. Mot1 redistributes TBP from TATA-containing to TATA-less promoters. Mol. Cell. Biol. 33, 4996–5004 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Guglielmini, J., Woo, A., Krupovic, M., Forterre, P. & Gaia, M. Diversification of giant and large eukaryotic dsDNA viruses predated the origin of modern eukaryotes. Preprint at bioRxiv https://doi.org/10.1101/455816 (2018).

    Article  Google Scholar 

  24. Alva, V. & Lupas, A. N. Histones predate the split between bacteria and archaea. Bioinformatics https://doi.org/10.1093/bioinformatics/bty1000 (2018).

    Article  Google Scholar 

  25. Henneman, B., van Emmerik, C., van Ingen, H. & Dame, R. T. Structure and function of archaeal histones. PLOS Genet. 14, e1007582 (2018).

    PubMed  PubMed Central  Google Scholar 

  26. Mattiroli, F. et al. Structure of histone-based chromatin in Archaea. Science 357, 609–612 (2017). This paper shows that archaeal histones can form extended polymers that wrap DNA and affect gene regulation.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Xie, Y. & Reeve, J. N. Transcription by an archaeal RNA polymerase is slowed but not blocked by an archaeal nucleosome. J. Bacteriol. 186, 3492–3498 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Wilkinson, S. P., Ouhammouch, M. & Geiduschek, E. P. Transcriptional activation in the context of repression mediated by archaeal histones. Proc. Natl Acad. Sci. USA 107, 6777–6781 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Ammar, R. et al. Chromatin is an ancient innovation conserved between Archaea and Eukarya. eLife 1, e00078 (2012).

    PubMed  PubMed Central  Google Scholar 

  30. Malik, H. S. & Henikoff, S. Phylogenomics of the nucleosome. Nat. Struct. Biol. 10, 882–891 (2003).

    CAS  PubMed  Google Scholar 

  31. Erives, A. J. Phylogenetic analysis of the core histone doublet and DNA topo II genes of Marseilleviridae: evidence of proto-eukaryotic provenance. Epigenetics Chromatin 10, 55 (2017). This paper analyses the phylogeny of histones and topoisomerase II in giant viruses and proposes that they reflect a proto-eukaryotic stage of histone evolution.

    PubMed  PubMed Central  Google Scholar 

  32. Marinov, G. K. & Lynch, M. Diversity and divergence of dinoflagellate histone proteins. G3 (Bethesda) 6, 397–422 (2015).

    Google Scholar 

  33. Gornik, S. G. et al. Loss of nucleosomal DNA condensation coincides with appearance of a novel nuclear protein in dinoflagellates. Curr. Biol. 22, 2303–2312 (2012).

    CAS  PubMed  Google Scholar 

  34. Irwin, N. A. T. et al. Viral proteins as a potential driver of histone depletion in dinoflagellates. Nat. Commun. 9, 1535 (2018).

    PubMed  PubMed Central  Google Scholar 

  35. Dalmasso, M. C., Sullivan, W. J. Jr & Angel, S. O. Canonical and variant histones of protozoan parasites. Front. Biosci. (Landmark Ed) 16, 2086–2105 (2011).

    CAS  Google Scholar 

  36. Kasinsky, H. E., Lewis, J. D., Dacks, J. B. & Ausio, J. Origin of H1 linker histones. FASEB J. 15, 34–42 (2001).

    CAS  PubMed  Google Scholar 

  37. Shintomi, K. et al. Mitotic chromosome assembly despite nucleosome depletion in Xenopus egg extracts. Science 356, 1284–1287 (2017). The authors show that condensins and topoisomerase II can form chromosome axes without nucleosomes, but nucleosomes are needed for full condensation and to prevent fragility.

    CAS  PubMed  Google Scholar 

  38. Shintomi, K., Takahashi, T. S. & Hirano, T. Reconstitution of mitotic chromatids with a minimum set of purified factors. Nat. Cell Biol. 17, 1014–1023 (2015).

    CAS  PubMed  Google Scholar 

  39. Madhani, H. D. The frustrated gene: origins of eukaryotic gene expression. Cell 155, 744–749 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Gangadharan, S., Mularoni, L., Fain-Thornton, J., Wheelan, S. J. & Craig, N. L. DNA transposon Hermes inserts into DNA in nucleosome-free regions in vivo. Proc. Natl Acad. Sci. USA 107, 21966–21972 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Aravind, L., Burroughs, A. M., Zhang, D. & Iyer, L. M. Protein and DNA modifications: evolutionary imprints of bacterial biochemical diversification and geochemistry on the provenance of eukaryotic epigenetics. Cold Spring Harb. Perspect. Biol. 6, a016063 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Zemach, A. & Zilberman, D. Evolution of eukaryotic DNA methylation and the pursuit of safer sex. Curr. Biol. 20, R780–785 (2010).

    CAS  PubMed  Google Scholar 

  43. Blot, N., Mavathur, R., Geertz, M., Travers, A. & Muskhelishvili, G. Homeostatic regulation of supercoiling sensitivity coordinates transcription of the bacterial genome. EMBO Rep. 7, 710–715 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Muskhelishvili, G. & Travers, A. The regulatory role of DNA supercoiling in nucleoprotein complex assembly and genetic activity. Biophys. Rev. 8, 5–22 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Sobetzko, P. Transcription-coupled DNA supercoiling dictates the chromosomal arrangement of bacterial genes. Nucleic Acids Res. 44, 1514–1524 (2016).

    PubMed  PubMed Central  Google Scholar 

  46. Le, T. B., Imakaev, M. V., Mirny, L. A. & Laub, M. T. High-resolution mapping of the spatial organization of a bacterial chromosome. Science 342, 731–734 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Sperling, A. S., Jeong, K. S., Kitada, T. & Grunstein, M. Topoisomerase II binds nucleosome-free DNA and acts redundantly with topoisomerase I to enhance recruitment of RNA Pol II in budding yeast. Proc. Natl Acad. Sci. USA 108, 12693–12698 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Sivolob, A. & Prunell, A. Linker histone-dependent organization and dynamics of nucleosome entry/exit DNAs. J. Mol. Biol. 331, 1025–1040 (2003).

    CAS  PubMed  Google Scholar 

  49. Sheinin, M. Y., Li, M., Soltani, M., Luger, K. & Wang, M. D. Torque modulates nucleosome stability and facilitates H2A/H2B dimer loss. Nat. Commun. 4, 2579 (2013).

    PubMed  Google Scholar 

  50. Ramachandran, S., Ahmad, K. & Henikoff, S. Transcription and remodeling produce asymmetrically unwrapped nucleosomal intermediates. Mol. Cell 68, 1038–1053 (2017). Mapping of protected subnucleosomal DNA fragments in D. melanogaster cells demonstrates that positive torsion generated by RNAPII elongation disrupts DNA contacts with the +1 nucleosome and that subnucleosomal DNA fragments from cell-free DNA in human blood plasma can be used to infer transcriptional status.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Teves, S. S. & Henikoff, S. Transcription-generated torsional stress destabilizes nucleosomes. Nat. Struct. Mol. Biol. 21, 88–94 (2014).

    CAS  PubMed  Google Scholar 

  52. Gaykalova, D. A. et al. Structural analysis of nucleosomal barrier to transcription. Proc. Natl Acad. Sci. USA 112, E5787–E5795 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Baranello, L. et al. RNA polymerase II regulates topoisomerase 1 activity to favor efficient transcription. Cell 165, 357–371 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Kouzine, F. et al. Permanganate/S1 nuclease footprinting reveals non-B DNA structures with regulatory potential across a mammalian genome. Cell Syst. 4, 344–356 (2017). The non-B-form DNA is globally mapped in mammalian cells by potassium permanganate footprinting and found to be enriched in the promoters of developmentally regulated genes.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Kouzine, F., Levens, D. & Baranello, L. DNA topology and transcription. Nucleus 5, 195–202 (2014).

    PubMed  PubMed Central  Google Scholar 

  56. Tsochatzidou, M., Malliarou, M., Papanikolaou, N., Roca, J. & Nikolaou, C. Genome urbanization: clusters of topologically co-regulated genes delineate functional compartments in the genome of Saccharomyces cerevisiae. Nucleic Acids Res. 45, 5818–5828 (2017). This paper describes topologically co-regulated gene clusters that are concurrently either upregulated or downregulated in response to topological stress and shows that these clusters are organized nonrandomly in the budding yeast genome.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Naughton, C. et al. Transcription forms and remodels supercoiling domains unfolding large-scale chromatin structures. Nat. Struct. Mol. Biol. 20, 387–395 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Forterre, P. & Gadelle, D. Phylogenomics of DNA topoisomerases: their origin and putative roles in the emergence of modern organisms. Nucleic Acids Res. 37, 679–692 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Iyer, L. M., Balaji, S., Koonin, E. V. & Aravind, L. Evolutionary genomics of nucleo-cytoplasmic large DNA viruses. Virus Res. 117, 156–184 (2006).

    CAS  PubMed  Google Scholar 

  60. Forterre, P. & Gaia, M. Giant viruses and the origin of modern eukaryotes. Curr. Opin. Microbiol. 31, 44–49 (2016).

    PubMed  Google Scholar 

  61. Earnshaw, W. C., Halligan, B., Cooke, C. A., Heck, M. M. & Liu, L. F. Topoisomerase II is a structural component of mitotic chromosome scaffolds. J. Cell Biol. 100, 1706–1715 (1985).

    CAS  PubMed  Google Scholar 

  62. Baxter, J. et al. Positive supercoiling of mitotic DNA drives decatenation by topoisomerase II in eukaryotes. Science 331, 1328–1332 (2011).

    CAS  PubMed  Google Scholar 

  63. Uuskula-Reimand, L. et al. Topoisomerase II beta interacts with cohesin and CTCF at topological domain borders. Genome Biol. 17, 182 (2016).

    PubMed  PubMed Central  Google Scholar 

  64. Baxter, J. & Aragon, L. A model for chromosome condensation based on the interplay between condensin and topoisomerase II. Trends Genet. 28, 110–117 (2012).

    CAS  PubMed  Google Scholar 

  65. Perez-Rueda, E. & Janga, S. C. Identification and genomic analysis of transcription factors in archaeal genomes exemplifies their functional architecture and evolutionary origin. Mol. Biol. Evol. 27, 1449–1459 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Aravind, L., Anantharaman, V., Balaji, S., Babu, M. M. & Iyer, L. M. The many faces of the helix-turn-helix domain: transcription regulation and beyond. FEMS Microbiol. Rev. 29, 231–262 (2005).

    CAS  PubMed  Google Scholar 

  67. Clapier, C. R., Iwasa, J., Cairns, B. R. & Peterson, C. L. Mechanisms of action and regulation of ATP-ependent chromatin-remodelling complexes. Nat. Rev. Mol. Cell Biol. 18, 407–422 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Flaus, A., Martin, D. M., Barton, G. J. & Owen-Hughes, T. Identification of multiple distinct Snf2 subfamilies with conserved structural motifs. Nucleic Acids Res. 34, 2887–2905 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. de Souza, R. F., Iyer, L. M. & Aravind, L. Diversity and evolution of chromatin proteins encoded by DNA viruses. Biochim. Biophys. Acta 1799, 302–318 (2010).

    PubMed  Google Scholar 

  70. Dann, G. P. et al. ISWI chromatin remodellers sense nucleosome modifications to determine substrate preference. Nature 548, 607–611 (2017). The authors demonstrate that remodelling complexes with the same ATPase but different accessory subunits respond differently to the same histone modification and that all remodellers require the acidic patch of H2A or H2A.Z for efficient remodelling. They propose that modifications around the acidic patch can modulate remodelling efficiency.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Lessard, J. et al. An essential switch in subunit composition of a chromatin remodeling complex during neural development. Neuron 55, 201–215 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Lee, C. K., Shibata, Y., Rao, B., Strahl, B. D. & Lieb, J. D. Evidence for nucleosome depletion at active regulatory regions genome-wide. Nat. Genet. 36, 900–905 (2004).

    CAS  PubMed  Google Scholar 

  73. Yuan, G. C. et al. Genome-scale identification of nucleosome positions in S. cerevisiae. Science 309, 626–630 (2005).

    CAS  PubMed  Google Scholar 

  74. Struhl, K. & Segal, E. Determinants of nucleosome positioning. Nat. Struct. Mol. Biol. 20, 267–273 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Chereji, R. V. & Clark, D. J. Major determinants of nucleosome positioning. Biophys. J. 114, 2279–2289 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Chereji, R. V., Ramachandran, S., Bryson, T. D. & Henikoff, S. Precise genome-wide mapping of single nucleosomes and linkers in vivo. Genome Biol. 19, 19 (2018).

    PubMed  PubMed Central  Google Scholar 

  77. Song, L. et al. A transcription factor hierarchy defines an environmental stress response network. Science 354, aag1550 (2016).

    PubMed  PubMed Central  Google Scholar 

  78. Uyehara, C. M. et al. Hormone-dependent control of developmental timing through regulation of chromatin accessibility. Genes Dev. 31, 862–875 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Joseph, S. R. et al. Competition between histone and transcription factor binding regulates the onset of transcription in zebrafish embryos. eLife 6, e23326 (2017). In zebrafish embryos, the concentration of TFs and of histones determines the timing of zygotic transcription without altering nucleosome density, supporting the idea of competition between nucleosomes and TFs in transcription activation.

    PubMed  PubMed Central  Google Scholar 

  80. Ramachandran, S. & Henikoff, S. Transcriptional regulators compete with nucleosomes post-replication. Cell 165, 580–592 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Yan, C., Chen, H. & Bai, L. Systematic study of nucleosome-displacing factors in budding yeast. Mol. Cell 71, 294–305 (2018). The authors systematically evaluate the nucleosome-displacing properties of 104 TFs.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Kubik, S. et al. Sequence-directed action of RSC remodeler and general regulatory factors modulates +1 nucleosome position to facilitate transcription. Mol. Cell 71, 89–102 (2018).

    CAS  PubMed  Google Scholar 

  83. Sartorelli, V. & Puri, P. L. Shaping gene expression by landscaping chromatin architecture: lessons from a master. Mol. Cell 71, 375–388 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Iwafuchi-Doi, M. et al. The pioneer transcription factor FoxA maintains an accessible nucleosome configuration at enhancers for tissue-specific gene activation. Mol. Cell 62, 79–91 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Johnson, T. A. et al. Conventional and pioneer modes of glucocorticoid receptor interaction with enhancer chromatin in vivo. Nucleic Acids Res. 46, 203–214 (2018).

    CAS  PubMed  Google Scholar 

  86. Schulz, K. N. et al. Zelda is differentially required for chromatin accessibility, transcription factor binding, and gene expression in the early Drosophila embryo. Genome Res. 25, 1715–1726 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Talbert, P. B. & Henikoff, S. Histone variants on the move: substrates for chromatin dynamics. Nat. Rev. Mol. Cell Biol. 18, 115–126 (2017).

    CAS  PubMed  Google Scholar 

  88. Verdin, E. & Ott, M. 50 years of protein acetylation: from gene regulation to epigenetics, metabolism and beyond. Nat. Rev. Mol. Cell Biol. 16, 258–264 (2015).

    CAS  PubMed  Google Scholar 

  89. Protacio, R. U., Li, G., Lowary, P. T. & Widom, J. Effects of histone tail domains on the rate of transcriptional elongation through a nucleosome. Mol. Cell. Biol. 20, 8866–8878 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Narita, T., Weinert, B. T. & Choudhary, C. Functions and mechanisms of non-histone protein acetylation. Nat. Rev. Mol. Cell Biol. https://doi.org/10.1038/s41580-018-0081-3 (2018).

    Article  Google Scholar 

  91. Klein, B. J. et al. Recognition of histone H3K14 acylation by MORF. Structure 25, 650–654 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Sabari, B. R., Zhang, D., Allis, C. D. & Zhao, Y. Metabolic regulation of gene expression through histone acylations. Nat. Rev. Mol. Cell Biol. 18, 90–101 (2017).

    CAS  PubMed  Google Scholar 

  93. Ghosh, S., Padmanabhan, B., Anand, C. & Nagaraja, V. Lysine acetylation of the Mycobacterium tuberculosis HU protein modulates its DNA binding and genome organization. Mol. Microbiol. 100, 577–588 (2016).

    CAS  PubMed  Google Scholar 

  94. Soares, D. J., Marc, F. & Reeve, J. N. Conserved eukaryotic histone-fold residues substituted into an archaeal histone increase DNA affinity but reduce complex flexibility. J. Bacteriol. 185, 3453–3457 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Bell, S. D., Botting, C. H., Wardleworth, B. N., Jackson, S. P. & White, M. F. The interaction of Alba, a conserved archaeal chromatin protein, with Sir2 and its regulation by acetylation. Science 296, 148–151 (2002).

    CAS  PubMed  Google Scholar 

  96. Wang, Z. et al. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 138, 1019–1031 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Crump, N. T. et al. Dynamic acetylation of all lysine-4 trimethylated histone H3 is evolutionarily conserved and mediated by p300/CBP. Proc. Natl Acad. Sci. USA 108, 7814–7819 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Durant, M. & Pugh, B. F. Genome-wide relationships between TAF1 and histone acetyltransferases in Saccharomyces cerevisiae. Mol. Cell. Biol. 26, 2791–2802 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Brown, C. E. et al. Recruitment of HAT complexes by direct activator interactions with the ATM-related Tra1 subunit. Science 292, 2333–2337 (2001).

    CAS  PubMed  Google Scholar 

  100. Hilton, I. B. et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat. Biotechnol. 33, 510–517 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Boija, A. et al. CBP regulates recruitment and release of promoter-proximal RNA polymerase II. Mol. Cell 68, 491–503 (2017). In D. melanogaster promoters, the authors find that Cbp maintains RNAPII at the pause site, acetylates the +1 nucleosome and is required to overcome the +1 nucleosome barrier to transcription.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Fujisawa, T. & Filippakopoulos, P. Functions of bromodomain-containing proteins and their roles in homeostasis and cancer. Nat. Rev. Mol. Cell Biol. 18, 246–262 (2017).

    CAS  PubMed  Google Scholar 

  103. Hassan, A. H. et al. Function and selectivity of bromodomains in anchoring chromatin-modifying complexes to promoter nucleosomes. Cell 111, 369–379 (2002).

    CAS  PubMed  Google Scholar 

  104. Ali, M. et al. Tandem PHD fingers of MORF/MOZ acetyltransferases display selectivity for acetylated histone H3 and are required for the association with chromatin. J. Mol. Biol. 424, 328–338 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Klein, B. J. et al. Yaf9 subunit of the NuA4 and SWR1 complexes targets histone H3K27ac through its YEATS domain. Nucleic Acids Res. 46, 421–430 (2018).

    CAS  PubMed  Google Scholar 

  106. Wang, A. Y. et al. Asf1-like structure of the conserved Yaf9 YEATS domain and role in H2A. Z deposition and acetylation. Proc. Natl Acad. Sci. USA 106, 21573–21578 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Hsu, C. C. et al. Gas41 links histone acetylation to H2A. Z deposition and maintenance of embryonic stem cell identity. Cell Discov. 4, 28 (2018).

    PubMed  PubMed Central  Google Scholar 

  108. Alvarez-Venegas, R., Sadder, M., Tikhonov, A. & Avramova, Z. Origin of the bacterial SET domain genes: vertical or horizontal? Mol. Biol. Evol. 24, 482–497 (2007).

    CAS  PubMed  Google Scholar 

  109. Martin, B. J. et al. Histone H3K4 and H3K36 methylation independently recruit the NuA3 histone acetyltransferase in Saccharomyces cerevisiae. Genetics 205, 1113–1123 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Morgan, M. A. J. et al. A cryptic Tudor domain links BRWD2/PHIP to COMPASS-mediated histone H3K4 methylation. Genes Dev. 31, 2003–2014 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Hodl, M. & Basler, K. Transcription in the absence of histone H3.2 and H3K4 methylation. Curr. Biol. 22, 2253–2257 (2012).

    CAS  PubMed  Google Scholar 

  112. Song, M. J. et al. Epigenome mapping highlights chromatin-mediated gene regulation in the protozoan parasite Trichomonas vaginalis. Sci. Rep. 7, 45365 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Gupta, A. P. & Bozdech, Z. Epigenetic landscapes underlining global patterns of gene expression in the human malaria parasite. Plasmodium falciparum. Int. J. Parasitol. 47, 399–407 (2017).

    PubMed  Google Scholar 

  114. Hsu, D. W., Chubb, J. R., Muramoto, T., Pears, C. J. & Mahadevan, L. C. Dynamic acetylation of lysine-4-trimethylated histone H3 and H3 variant biology in a simple multicellular eukaryote. Nucleic Acids Res. 40, 7247–7256 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Vanacova, S., Liston, D. R., Tachezy, J. & Johnson, P. J. Molecular biology of the amitochondriate parasites, Giardia intestinalis, Entamoeba histolytica and Trichomonas vaginalis. Int. J. Parasitol. 33, 235–255 (2003).

    CAS  PubMed  Google Scholar 

  116. Carranza, P. G. et al. Specific histone modifications play critical roles in the control of encystation and antigenic variation in the early-branching eukaryote. Giardia lamblia. Int. J. Biochem. Cell Biol. 81, 32–43 (2016).

    CAS  PubMed  Google Scholar 

  117. Wedel, C., Forstner, K. U., Derr, R. & Siegel, T. N. GT-rich promoters can drive RNA pol II transcription and deposition of H2A. Z in African trypanosomes. EMBO J. 36, 2581–2594 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Siegel, T. N. et al. Four histone variants mark the boundaries of polycistronic transcription units in Trypanosoma brucei. Genes Dev. 23, 1063–1076 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Raisner, R. M. et al. Histone variant H2A. Z marks the 5′ ends of both active and inactive genes in euchromatin. Cell 123, 233–248 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Ramachandran, S., Zentner, G. E. & Henikoff, S. Asymmetric nucleosomes flank promoters in the budding yeast genome. Genome Res. 25, 381–390 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Ranjan, A. et al. Nucleosome-free region dominates histone acetylation in targeting SWR1 to promoters for H2A. Z replacement. Cell 154, 1232–1245 (2013).

    CAS  PubMed  Google Scholar 

  122. Watanabe, S., Radman-Livaja, M., Rando, O. J. & Peterson, C. L. A histone acetylation switch regulates H2A. Z deposition by the SWR-C remodeling enzyme. Science 340, 195–199 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Gomez-Zambrano, A. et al. Arabidopsis SWC4 binds DNA and recruits the SWR1 complex to modulate histone H2A.Z deposition at key regulatory genes. Mol. Plant 11, 815–832 (2018).

    CAS  PubMed  Google Scholar 

  124. Brahma, S. et al. INO80 exchanges H2A. Z for H2A by translocating on DNA proximal to histone dimers. Nat. Commun. 8, 15616 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Wang, F., Ranjan, A., Wei, D. & Wu, C. Comment on “A histone acetylation switch regulates H2A. Z deposition by the SWR-C remodeling enzyme”. Science 353, 358 (2016).

    CAS  PubMed  Google Scholar 

  127. Papamichos-Chronakis, M., Krebs, J. E. & Peterson, C. L. Interplay between Ino80 and Swr1 chromatin remodeling enzymes regulates cell cycle checkpoint adaptation in response to DNA damage. Genes Dev. 20, 2437–2449 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Adam, M., Robert, F., Larochelle, M. & Gaudreau, L. H2A. Z is required for global chromatin integrity and for recruitment of RNA polymerase II under specific conditions. Mol. Cell. Biol. 21, 6270–6279 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Tramantano, M. et al. Constitutive turnover of histone H2A.Z at yeast promoters requires the preinitiation complex. eLife 5, e14243 (2016).

    PubMed  PubMed Central  Google Scholar 

  130. Meneghini, M. D., Wu, M. & Madhani, H. D. Conserved histone variant H2A. Z protects euchromatin from the ectopic spread of silent heterochromatin. Cell 112, 725–736 (2003).

    CAS  PubMed  Google Scholar 

  131. Rudnizky, S. et al. H2A. Z controls the stability and mobility of nucleosomes to regulate expression of the LH genes. Nat. Commun. 7, 12958 (2016). Optical tweezers are used to show that H2A.Z nucleosomes have a lower breaking force than H2A nucleosomes and that positioning of H2A.Z nucleosomes relative to the TSS can lead to distinct regulation strategies.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  133. Ishibashi, T. et al. Acetylation of vertebrate H2A. Z and its effect on the structure of the nucleosome. Biochemistry 48, 5007–5017 (2009).

    CAS  PubMed  Google Scholar 

  134. Weber, C. M., Ramachandran, S. & Henikoff, S. Nucleosomes are context-specific, H2A. Z-modulated barriers to RNA polymerase. Mol. Cell 53, 819–830 (2014).

    CAS  PubMed  Google Scholar 

  135. Cortijo, S. et al. Transcriptional regulation of the ambient temperature response by H2A. Z nucleosomes and HSF1 transcription factors in Arabidopsis. Mol. Plant 10, 1258–1273 (2017).

    CAS  PubMed  Google Scholar 

  136. Hu, G. et al. H2A. Z facilitates access of active and repressive complexes to chromatin in embryonic stem cell self-renewal and differentiation. Cell Stem Cell 12, 180–192 (2013).

    CAS  PubMed  Google Scholar 

  137. Zilberman, D., Coleman-Derr, D., Ballinger, T. & Henikoff, S. Histone H2A. Z and DNA methylation are mutually antagonistic chromatin marks. Nature 456, 125–129 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Zhang, X., Bernatavichute, Y. V., Cokus, S., Pellegrini, M. & Jacobsen, S. E. Genome-wide analysis of mono-, di- and trimethylation of histone H3 lysine 4 in Arabidopsis thaliana. Genome Biol. 10, R62 (2009).

    PubMed  PubMed Central  Google Scholar 

  139. Wang, X. et al. Merge and separation of NuA4 and SWR1 complexes control cell fate plasticity in Candida albicans. Cell Discov. 4, 45 (2018).

    PubMed  PubMed Central  Google Scholar 

  140. Muller, J. et al. Histone methyltransferase activity of a Drosophila polycomb group repressor complex. Cell 111, 197–208 (2002).

    CAS  PubMed  Google Scholar 

  141. Kuzmichev, A., Nishioka, K., Erdjument-Bromage, H., Tempst, P. & Reinberg, D. Histone methyltransferase activity associated with a human multiprotein complex containing the enhancer of zeste protein. Genes Dev. 16, 2893–2905 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Shaver, S., Casas-Mollano, J. A., Cerny, R. L. & Cerutti, H. Origin of the polycomb repressive complex 2 and gene silencing by an E(z) homolog in the unicellular alga Chlamydomonas. Epigenetics 5, 301–312 (2010).

    CAS  PubMed  Google Scholar 

  143. Jamieson, K. et al. Telomere repeats induce domains of H3K27 methylation in Neurospora. eLife 7, e31216 (2018).

    PubMed  PubMed Central  Google Scholar 

  144. Grossniklaus, U. & Paro, R. Transcriptional silencing by polycomb-group proteins. Cold Spring Harb. Perspect. Biol. 6, a019331 (2014).

    PubMed  PubMed Central  Google Scholar 

  145. Wang, H. et al. Arabidopsis flower and embryo developmental genes are repressed in seedlings by different combinations of polycomb group proteins in association with distinct sets of cis-regulatory elements. PLOS Genet. 12, e1005771 (2016). The authors show that the H3K27me3-binding chromodomain protein LHP1 in A. thaliana is associated with the PRC2 histone methyltransferase CLF and aids spreading of methylation to repress floral development, while PRC1 components BMI1 and RING1 associate with histone methyltransferases CLF and SWR and suppress embryo-specific genes.

    PubMed  PubMed Central  Google Scholar 

  146. Li, H. et al. Polycomb-like proteins link the PRC2 complex to CpG islands. Nature 549, 287–291 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Kuzmichev, A., Jenuwein, T., Tempst, P. & Reinberg, D. Different EZH2-containing complexes target methylation of histone H1 or nucleosomal histone H3. Mol. Cell 14, 183–193 (2004).

    CAS  PubMed  Google Scholar 

  148. Poepsel, S., Kasinath, V. & Nogales, E. Cryo-EM structures of PRC2 simultaneously engaged with two functionally distinct nucleosomes. Nat. Struct. Mol. Biol. 25, 154–162 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Pengelly, A. R., Kalb, R., Finkl, K. & Muller, J. Transcriptional repression by PRC1 in the absence of H2A monoubiquitylation. Genes Dev. 29, 1487–1492 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Kundu, S. et al. Polycomb repressive complex 1 generates discrete compacted domains that change during differentiation. Mol. Cell 65, 432–446 (2017). Super-resolution microscopy of mammalian cells identifies PRC1 compacted chromatin domains that depend on Polyhomeotic but not on PRC1-dependent ubiquitylation and that are lost as PRC1 binding is lost during differentiation.

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Boettiger, A. N. et al. Super-resolution imaging reveals distinct chromatin folding for different epigenetic states. Nature 529, 418–422 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Robinson, A. K. et al. The growth-suppressive function of the polycomb group protein polyhomeotic is mediated by polymerization of its sterile alpha motif (SAM) domain. J. Biol. Chem. 287, 8702–8713 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Dumesic, P. A. et al. Product binding enforces the genomic specificity of a yeast polycomb repressive complex. Cell 160, 204–218 (2015).

    CAS  PubMed  Google Scholar 

  154. Berke, L. & Snel, B. The plant polycomb repressive complex 1 (PRC1) existed in the ancestor of seed plants and has a complex duplication history. BMC Evol. Biol. 15, 44 (2015).

    PubMed  PubMed Central  Google Scholar 

  155. Li, Z., Fu, X., Wang, Y., Liu, R. & He, Y. Polycomb-mediated gene silencing by the BAH-EMF1 complex in plants. Nat. Genet. 50, 1254–1261 (2018).

    CAS  PubMed  Google Scholar 

  156. Peng, L. et al. Structural analysis of the arabidopsis AL2-PAL and PRC1 complex provides mechanistic insight into active-to-repressive chromatin state switch. J. Mol. Biol. 430, 4245–4259 (2018).

    CAS  PubMed  Google Scholar 

  157. Baranello, L. et al. DNA break mapping reveals topoisomerase II activity genome-wide. Int. J. Mol. Sci. 15, 13111–13122 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Gray, M. W. & Doolittle, W. F. Has the endosymbiont hypothesis been proven? Microbiol. Rev. 46, 1–42 (1982).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Yang, D., Oyaizu, Y., Oyaizu, H., Olsen, G. J. & Woese, C. R. Mitochondrial origins. Proc. Natl Acad. Sci. USA 82, 4443–4447 (1985).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Woese, C. R. & Fox, G. E. Phylogenetic structure of the prokaryotic domain: the primary kingdoms. Proc. Natl Acad. Sci. USA 74, 5088–5090 (1977).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Woese, C. R., Magrum, L. J. & Fox, G. E. Archaebacteria. J. Mol. Evol. 11, 245–251 (1978).

    CAS  PubMed  Google Scholar 

  162. Harish, A. What is an archaeon and are the Archaea really unique? PeerJ 6, e5770 (2018).

    PubMed  PubMed Central  Google Scholar 

  163. Zaremba-Niedzwiedzka, K. et al. Asgard archaea illuminate the origin of eukaryotic cellular complexity. Nature 541, 353–358 (2017). New species of archaea are identified with proteins homologous to eukaryotic endomembrane system components, and a two-domain model is supported in which eukaryotes are rooted within archaea, in contrast to Nasir et al. (2008).

    CAS  PubMed  Google Scholar 

  164. Da Cunha, V., Gaia, M., Nasir, A. & Forterre, P. Asgard archaea do not close the debate about the universal tree of life topology. PLOS Genet. 14, e1007215 (2018).

    PubMed  PubMed Central  Google Scholar 

  165. Moreira, D. & Lopez-Garcia, P. Symbiosis between methanogenic archaea and delta-proteobacteria as the origin of eukaryotes: the syntrophic hypothesis. J. Mol. Evol. 47, 517–530 (1998).

    CAS  PubMed  Google Scholar 

  166. Pittis, A. A. & Gabaldon, T. Late acquisition of mitochondria by a host with chimaeric prokaryotic ancestry. Nature 531, 101–104 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Filee, J. Multiple occurrences of giant virus core genes acquired by eukaryotic genomes: the visible part of the iceberg? Virology 466–467, 53–59 (2014).

    PubMed  Google Scholar 

  168. Huff, J. T. & Zilberman, D. Dnmt1-independent CG methylation contributes to nucleosome positioning in diverse eukaryotes. Cell 156, 1286–1297 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Ambrosi, C., Manzo, M. & Baubec, T. Dynamics and context-dependent roles of DNA methylation. J. Mol. Biol. 429, 1459–1475 (2017).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  171. 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).

    CAS  PubMed  Google Scholar 

  172. Satyaki, P. R. & Gehring, M. DNA methylation and imprinting in plants: machinery and mechanisms. Crit. Rev. Biochem. Mol. Biol. 52, 163–175 (2017).

    CAS  PubMed  Google Scholar 

  173. Williams, B. P. & Gehring, M. Stable transgenerational epigenetic inheritance requires a DNA methylation-sensing circuit. Nat. Commun. 8, 2124 (2017).

    PubMed  PubMed Central  Google Scholar 

  174. 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).

    CAS  PubMed  Google Scholar 

  175. Otani, J. et al. Structural basis for recognition of H3K4 methylation status by the DNA methyltransferase 3A ATRX-DNMT3-DNMT3L domain. EMBO Rep. 10, 1235–1241 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Neri, F. et al. Intragenic DNA methylation prevents spurious transcription initiation. Nature 543, 72–77 (2017).

    CAS  PubMed  Google Scholar 

  177. Coleman-Derr, D. & Zilberman, D. Deposition of histone variant H2A. Z within gene bodies regulates responsive genes. PLOS Genet. 8, e1002988 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Murphy, P. J., Wu, S. F., James, C. R., Wike, C. L. & Cairns, B. R. Placeholder nucleosomes underlie germline-to-embryo DNA methylation reprogramming. Cell 172, 993–1006 (2018).

    CAS  PubMed  Google Scholar 

  179. Havas, K. et al. Generation of superhelical torsion by ATP-dependent chromatin remodeling activities. Cell 103, 1133–1142 (2000).

    CAS  PubMed  Google Scholar 

  180. Saha, A., Wittmeyer, J. & Cairns, B. R. Chromatin remodeling by RSC involves ATP-dependent DNA translocation. Genes Dev. 16, 2120–2134 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Gamarra, N., Johnson, S. L., Trnka, M. J., Burlingame, A. L. & Narlikar, G. J. The nucleosomal acidic patch relieves auto-inhibition by the ISWI remodeler SNF2h. eLife 7, e35322 (2018).

    PubMed  PubMed Central  Google Scholar 

  182. Yan, L., Wang, L., Tian, Y., Xia, X. & Chen, Z. Structure and regulation of the chromatin remodeller ISWI. Nature 540, 466–469 (2016).

    CAS  PubMed  Google Scholar 

  183. Sinha, K. K., Gross, J. D. & Narlikar, G. J. Distortion of histone octamer core promotes nucleosome mobilization by a chromatin remodeler. Science 355, eaaa3761 (2017). Deformation of the histone octamer is shown to be required for remodelling activity of the SNF2h remodeller but not INO80, and it reduces nucleosome eviction by RSC. The authors propose that altered nucleosome conformations may be relevant in other processes such as eviction by pioneer factors.

    PubMed  PubMed Central  Google Scholar 

  184. Chaban, Y. et al. Structure of a RSC-nucleosome complex and insights into chromatin remodeling. Nat. Struct. Mol. Biol. 15, 1272–1277 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  186. 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).

    CAS  PubMed  Google Scholar 

  187. Rivera-Casas, C., Gonzalez-Romero, R., Cheema, M. S., Ausio, J. & Eirin-Lopez, J. M. The characterization of macroH2A beyond vertebrates supports an ancestral origin and conserved role for histone variants in chromatin. Epigenetics 11, 415–425 (2016).

    PubMed  PubMed Central  Google Scholar 

  188. Chakravarthy, S., Patel, A. & Bowman, G. D. The basic linker of macroH2A stabilizes DNA at the entry/exit site of the nucleosome. Nucleic Acids Res. 40, 8285–8295 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Muthurajan, U. M., McBryant, S. J., Lu, X., Hansen, J. C. & Luger, K. The linker region of macroH2A promotes self-association of nucleosomal arrays. J. Biol. Chem. 286, 23852–23864 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Bowerman, S. & Wereszczynski, J. Effects of macroH2A and H2A.Z on nucleosome dynamics as elucidated by molecular dynamics simulations. Biophys. J. 110, 327–337 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Doyen, C. M. et al. Mechanism of polymerase II transcription repression by the histone variant macroH2A. Mol. Cell. Biol. 26, 1156–1164 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Chang, E. Y. et al. MacroH2A allows ATP-dependent chromatin remodeling by SWI/SNF and ACF complexes but specifically reduces recruitment of SWI/SNF. Biochemistry 47, 13726–13732 (2008).

    CAS  PubMed  Google Scholar 

  193. Chadwick, B. P., Valley, C. M. & Willard, H. F. Histone variant macroH2A contains two distinct macrochromatin domains capable of directing macroH2A to the inactive X chromosome. Nucleic Acids Res. 29, 2699–2705 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Lavigne, M. D. et al. Composite macroH2A/NRF-1 nucleosomes suppress noise and generate robustness in gene expression. Cell Rep. 11, 1090–1101 (2015).

    CAS  PubMed  Google Scholar 

  195. Pliatska, M., Kapasa, M., Kokkalis, A., Polyzos, A. & Thanos, D. The histone variant macroH2A blocks cellular reprogramming by inhibiting mesenchymal-to-epithelial transition. Mol. Cell. Biol. 38, e00669–17 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Molaro, A., Young, J. M. & Malik, H. S. Evolutionary origins and diversification of testis-specific short histone H2A variants in mammals. Genome Res. 28, 460–473 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Soboleva, T. A. et al. A new link between transcriptional initiation and pre-mRNA splicing: the RNA binding histone variant H2A. B. PLOS Genet. 13, e1006633 (2017).

    PubMed  PubMed Central  Google Scholar 

  198. Barral, S. et al. Histone variant H2A. L.2 guides transition protein-dependent protamine assembly in male germ cells. Mol. Cell 66, 89–101 (2017).

    CAS  PubMed  Google Scholar 

  199. Contrepois, K. et al. Histone variant H2A. J accumulates in senescent cells and promotes inflammatory gene expression. Nat. Commun. 8, 14995 (2017).

    PubMed  PubMed Central  Google Scholar 

  200. Ponger, L. & Li, W. H. Evolutionary diversification of DNA methyltransferases in eukaryotic genomes. Mol. Biol. Evol. 22, 1119–1128 (2005).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank S. Brahma, K. Ahmad and the reviewers for helpful suggestions on the manuscript.

Reviewer information

Nature Reviews Genetics thanks K. Struhl and the other, anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

P.B.T. and M.P.M. researched content for the article. All authors contributed to the discussion of content, writing the article and reviewing and editing the manuscript before submission.

Corresponding author

Correspondence to Steven Henikoff.

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.

Glossary

Histone fold domain

(HFD). A protein dimerization domain of three helices separated by two loops that is characteristic of archaeal and core eukaryotic histones, TATA-binding protein-associated factors and some other proteins.

Four-helix bundles

Structures formed by two helices of each of two histones that enable dimers to assemble into more complex structures.

Metamonads

Anaerobic cells typically with two pairs of basal bodies with one posterior and three anterior flagella. Metamonads include the diplomonad Giardia intestinalis and the parabasalid Trichomonas vaginalis, among others, and may represent one of the earliest branches of the eukaryotic phylogenetic tree.

Kinetoplastids

Flagellates that have a dense mass of DNA called a kinetoplast, which contains many copies of the mitochondrial genome. Kinetoplastids include Bodonids and trypanosomes and are thought to represent an early branch of the eukaryotic phylogenetic tree.

SET domain

A protein domain generally associated with protein lysine methyltransferase activity.

Last eukaryotic common ancestor

(LECA). The most recent cell that was ancestral to all extant eukaryotes, inferred to be a complex nucleated cell with an endomembrane system, a cytoskeleton, a mitochondrion, and linear chromosomes that were packaged in nucleosomes and underwent mitosis and meiosis.

First eukaryotic common ancestor

(FECA). A hypothetical first cell that was ancestral to all eukaryotes and distinct from bacteria and archaea, variously conceived as an urkaryote or a chimeric organism composed of host and endosymbiont.

Urkaryote

A hypothetical cell ancestral to the eukaryotic cytoplasm with distinct features of the translational apparatus that distinguish it from archaea and bacteria and that arose independently from the last universal cellular ancestor.

Endosymbiont

A cell or organism living inside another cell or organism.

Proto-eukaryote

A cell in any stage of eukaryotic evolution between the first eukaryotic common ancestor to the last eukaryotic common ancestor.

General transcription factors

The factors that together with RNA polymerase II make up the pre-initiation complex for transcription.

Nucleocytoplasmic large DNA viruses

(NCLDVs). A diverse monophyletic group of fairly large, complex DNA viruses, including giant viruses such as the Marseilleviridae as well as other more modestly sized viruses.

CenH3

Histone H3 variant specific to centromeric nucleosomes, examples of which include mammalian CENP-A and budding yeast Cse4.

Eukaryogenesis

The process of evolving from the first eukaryotic common ancestor to the last eukaryotic common ancestor.

Hypernucleosomes

Archaeal histone polymers of variable length that wrap DNA.

Monophyletic group

A group of genes, proteins or organisms that includes all the descendants of a single common ancestor and excludes others.

Capsid

A proteinaceous structure that encloses a viral genome for protection and dispersion.

Winged helix domain

A protein domain with combined specific and nonspecific DNA-binding affinity characterized by a helix–turn–helix motif flanked by β-sheets on one or both sides.

Superhelicity

The degree of torsion or supercoiling in a DNA molecule.

Plectonemes

Writhed loops resulting from supercoiling.

Enhancer RNAs

(eRNAs). Non-coding RNAs transcribed from enhancers.

Topologically associating domains

(TADs). Regions of the genome that interact with themselves in 3D nuclear space more often than regions outside the TAD.

+1 Nucleosome

A highly conserved nucleosome positioned downstream of the nucleosome-depleted region, which acts as a barrier to transcription elongation by RNA polymerase.

Pioneer factors

Transcription factors that can bind to nucleosome-occluded DNA and promote accessibility to transcription, often at an early step in a developmental pathway.

CpG islands

Regions of more than 200 bp near many mammalian promoters that are enriched for CG dinucleotides.

Gonadotropes

Cells of the pituitary gland that secrete luteinizing hormone and follicle-stimulating hormone.

Homotypic nucleosomes

Nucleosomes in which both members of any particular histone family are the same histone variant.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Talbert, P.B., Meers, M.P. & Henikoff, S. Old cogs, new tricks: the evolution of gene expression in a chromatin context. Nat Rev Genet 20, 283–297 (2019). https://doi.org/10.1038/s41576-019-0105-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-019-0105-7

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