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:

Covalent modifications of histones during development and disease pathogenesis

Abstract

Covalent modifications of histones are central to the regulation of chromatin dynamics, and, therefore, many biological processes involving chromatin, such as replication, repair, transcription and genome stability, are regulated by chromatin and its modifications. In this review, we discuss the biochemical, molecular and genetic properties of the enzymatic machinery involved in four different types of histone modification: acetylation, ubiquitination, phosphorylation and methylation. We also discuss how perturbation of the activity of this enzymatic machinery can cause developmental defects and disease.

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

Figure 1: Post-translational modifications of human nucleosomal histones.
Figure 2: Schematic representation of the molecular machinery required for proper histone H3 methylation on lysines 4 and 79.

Similar content being viewed by others

References

  1. Kornberg, R.D. Chromatin structure: a repeating unit of histones and DNA. Science 184, 868–871 (1974).

    Article  CAS  PubMed  Google Scholar 

  2. Kornberg, R.D. & Lorch, Y. Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome. Cell 98, 285–294 (1999).

    CAS  PubMed  Google Scholar 

  3. Woodcock, C.L. Chromatin architecture. Curr. Opin. Struct. Biol. 16, 213–220 (2006).

    CAS  PubMed  Google Scholar 

  4. Woodcock, C.L., Skoultchi, A.I. & Fan, Y. Role of linker histone in chromatin structure and function: H1 stoichiometry and nucleosome repeat length. Chromosome Res. 14, 17–25 (2006).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  6. Cosgrove, M.S. & Wolberger, C. How does the histone code work? Biochem. Cell Biol. 83, 468–476 (2005).

    CAS  PubMed  Google Scholar 

  7. Ehrenhofer-Murray, A.E. Chromatin dynamics at DNA replication, transcription and repair. Eur. J. Biochem. 271, 2335–2349 (2004).

    CAS  PubMed  Google Scholar 

  8. Groth, A., Rocha, W., Verreault, A. & Almouzni, G. Chromatin challenges during DNA replication and repair. Cell 128, 721–733 (2007).

    CAS  PubMed  Google Scholar 

  9. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Kusch, T. & Workman, J.L. Histone variants and complexes involved in their exchange. Subcell. Biochem. 41, 91–109 (2007).

    PubMed  Google Scholar 

  11. Li, B., Carey, M. & Workman, J.L. The role of chromatin during transcription. Cell 128, 707–719 (2007).

    CAS  PubMed  Google Scholar 

  12. Rice, J.C. et al. Histone methyltransferases direct different degrees of methylation to define distinct chromatin domains. Mol. Cell 12, 1591–1598 (2003).

    CAS  PubMed  Google Scholar 

  13. Shilatifard, A. Chromatin modifications by methylation and ubiquitination: implications in the regulation of gene expression. Annu. Rev. Biochem. 75, 243–269 (2006).

    CAS  PubMed  Google Scholar 

  14. Biron, V.L., McManus, K.J., Hu, N., Hendzel, M.J. & Underhill, D.A. Distinct dynamics and distribution of histone methyl-lysine derivatives in mouse development. Dev. Biol. 276, 337–351 (2004).

    CAS  PubMed  Google Scholar 

  15. Dodge, J.E., Kang, Y.K., Beppu, H., Lei, H. & Li, E. Histone H3–K9 methyltransferase ESET is essential for early development. Mol. Cell. Biol. 24, 2478–2486 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Lin, W. & Dent, S.Y. Functions of histone-modifying enzymes in development. Curr. Opin. Genet. Dev. 16, 137–142 (2006).

    CAS  PubMed  Google Scholar 

  17. Margueron, R., Trojer, P. & Reinberg, D. The key to development: interpreting the histone code? Curr. Opin. Genet. Dev. 15, 163–176 (2005).

    CAS  PubMed  Google Scholar 

  18. Torres-Padilla, M.E., Parfitt, D.E., Kouzarides, T. & Zernicka-Goetz, M. Histone arginine methylation regulates pluripotency in the early mouse embryo. Nature 445, 214–218 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Shahbazian, M.D. & Grunstein, M. Functions of site-specific histone acetylation and deacetylation. Annu. Rev. Biochem. 76, 75–100 (2007).

    CAS  PubMed  Google Scholar 

  20. Denslow, S.A. & Wade, P.A. The human Mi-2/NuRD complex and gene regulation. Oncogene 26, 5433–5438 (2007).

    CAS  PubMed  Google Scholar 

  21. Keogh, M.C. et al. Cotranscriptional set2 methylation of histone H3 lysine 36 recruits a repressive Rpd3 complex. Cell 123, 593–605 (2005).

    CAS  PubMed  Google Scholar 

  22. Zhang, Y. Transcriptional regulation by histone ubiquitination and deubiquitination. Genes Dev. 17, 2733–2740 (2003).

    CAS  PubMed  Google Scholar 

  23. Fang, J., Chen, T., Chadwick, B., Li, E. & Zhang, Y. Ring1b-mediated H2A ubiquitination associates with inactive X chromosomes and is involved in initiation of X inactivation. J. Biol. Chem. 279, 52812–52815 (2004).

    CAS  PubMed  Google Scholar 

  24. Daniel, J.A. et al. Deubiquitination of histone H2B by a yeast acetyltransferase complex regulates transcription. J. Biol. Chem. 279, 1867–1871 (2004).

    CAS  PubMed  Google Scholar 

  25. Henry, K.W. et al. Transcriptional activation via sequential histone H2B ubiquitylation and deubiquitylation, mediated by SAGA-associated Ubp8. Genes Dev. 17, 2648–2663 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhu, P. et al. A histone H2A deubiquitinase complex coordinating histone acetylation and H1 dissociation in transcriptional regulation. Mol. Cell 27, 609–621 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Barber, C.M. et al. The enhancement of histone H4 and H2A serine 1 phosphorylation during mitosis and S-phase is evolutionarily conserved. Chromosoma 112, 360–371 (2004).

    CAS  PubMed  Google Scholar 

  28. Bradbury, E.M. Reversible histone modifications and the chromosome cell cycle. Bioessays 14, 9–16 (1992).

    CAS  PubMed  Google Scholar 

  29. Koshland, D. & Strunnikov, A. Mitotic chromosome condensation. Annu. Rev. Cell Dev. Biol. 12, 305–333 (1996).

    CAS  PubMed  Google Scholar 

  30. Foster, E.R. & Downs, J.A. Histone H2A phosphorylation in DNA double-strand break repair. FEBS J. 272, 3231–3240 (2005).

    CAS  PubMed  Google Scholar 

  31. Ahn, S.H., Henderson, K.A., Keeney, S. & Allis, C.D. H2B (Ser10) phosphorylation is induced during apoptosis and meiosis in S. cerevisiae. Cell Cycle 4, 780–783 (2005).

    CAS  PubMed  Google Scholar 

  32. Nowak, S.J. & Corces, V.G. Phosphorylation of histone H3: a balancing act between chromosome condensation and transcriptional activation. Trends Genet. 20, 214–220 (2004).

    CAS  PubMed  Google Scholar 

  33. Krishnamoorthy, T. et al. Phosphorylation of histone H4 Ser1 regulates sporulation in yeast and is conserved in fly and mouse spermatogenesis. Genes Dev. 20, 2580–2592 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Wendt, K.D. & Shilatifard, A. Packing for the germy: the role of histone H4 Ser1 phosphorylation in chromatin compaction and germ cell development. Genes Dev. 20, 2487–2491 (2006).

    CAS  PubMed  Google Scholar 

  35. Sims, R.J. III, & Reinberg, D. Histone H3 Lys 4 methylation: caught in a bind? Genes Dev. 20, 2779–2786 (2006).

    CAS  PubMed  Google Scholar 

  36. Trojer, P. & Reinberg, D. Histone lysine demethylases and their impact on epigenetics. Cell 125, 213–217 (2006).

    CAS  PubMed  Google Scholar 

  37. Feng, Q. et al. Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain. Curr. Biol. 12, 1052–1058 (2002).

    CAS  PubMed  Google Scholar 

  38. Lacoste, N., Utley, R.T., Hunter, J.M., Poirier, G.G. & Cote, J. Disruptor of telomeric silencing-1 is a chromatin-specific histone H3 methyltransferase. J. Biol. Chem. 277, 30421–30424 (2002).

    CAS  PubMed  Google Scholar 

  39. Ng, H.H. et al. Lysine methylation within the globular domain of histone H3 by Dot1 is important for telomeric silencing and Sir protein association. Genes Dev. 16, 1518–1527 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. van Leeuwen, F., Gafken, P.R. & Gottschling, D.E. Dot1p modulates silencing in yeast by methylation of the nucleosome core. Cell 109, 745–756 (2002).

    CAS  PubMed  Google Scholar 

  41. Klose, R.J. & Zhang, Y. Regulation of histone methylation by demethylimination and demethylation. Nat. Rev. Mol. Cell Biol. 8, 307–318 (2007).

    CAS  PubMed  Google Scholar 

  42. Schneider, J. & Shilatifard, A. Histone demethylation by hydroxylation: chemistry in action. ACS Chem. Biol. 1, 75–81 (2006).

    CAS  PubMed  Google Scholar 

  43. Shi, Y. Histone lysine demethylases: emerging roles in development, physiology and disease. Nat. Rev. Genet. 8, 829–833 (2007).

    CAS  PubMed  Google Scholar 

  44. Eissenberg, J.C. et al. The trithorax-group gene in Drosophila little imaginal discs encodes a trimethylated histone H3 Lys4 demethylase. Nat. Struct. Mol. Biol. 14, 344–346 (2007).

    CAS  PubMed  Google Scholar 

  45. Iwase, S. et al. The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases. Cell 128, 1077–1088 (2007).

    CAS  PubMed  Google Scholar 

  46. Lee, N. et al. The trithorax-group protein Lid is a histone H3 trimethyl-Lys4 demethylase. Nat. Struct. Mol. Biol. 14, 341–343 (2007).

    CAS  PubMed  Google Scholar 

  47. Seward, D.J. et al. Demethylation of trimethylated histone H3 Lys4 in vivo by JARID1 JmjC proteins. Nat. Struct. Mol. Biol. 14, 240–242 (2007).

    CAS  PubMed  Google Scholar 

  48. Masumoto, H., Hawke, D., Kobayashi, R. & Verreault, A. A role for cell-cycle-regulated histone H3 lysine 56 acetylation in the DNA damage response. Nature 436, 294–298 (2005).

    CAS  PubMed  Google Scholar 

  49. Schneider, J., Bajwa, P., Johnson, F.C., Bhaumik, S.R. & Shilatifard, A. Rtt109 is required for proper H3K56 acetylation: a chromatin mark associated with the elongating RNA polymerase II. J. Biol. Chem. 281, 37270–37274 (2006).

    CAS  PubMed  Google Scholar 

  50. Xu, F., Zhang, K. & Grunstein, M. Acetylation in histone H3 globular domain regulates gene expression in yeast. Cell 121, 375–385 (2005).

    CAS  PubMed  Google Scholar 

  51. Dion, M.F., Altschuler, S.J., Wu, L.F. & Rando, O.J. Genomic characterization reveals a simple histone H4 acetylation code. Proc. Natl. Acad. Sci. USA 102, 5501–5506 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lucchesi, J.C., Kelly, W.G. & Panning, B. Chromatin remodeling in dosage compensation. Annu. Rev. Genet. 39, 615–651 (2005).

    CAS  PubMed  Google Scholar 

  53. Shogren-Knaak, M. et al. Histone H4–K16 acetylation controls chromatin structure and protein interactions. Science 311, 844–847 (2006).

    CAS  PubMed  Google Scholar 

  54. Braunstein, M., Sobel, R.E., Allis, C.D., Turner, B.M. & Broach, J.R. Efficient transcriptional silencing in Saccharomyces cerevisiae requires a heterochromatin histone acetylation pattern. Mol. Cell. Biol. 16, 4349–4356 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Turner, B.M., Birley, A.J. & Lavender, J. Histone H4 isoforms acetylated at specific lysine residues define individual chromosomes and chromatin domains in Drosophila polytene nuclei. Cell 69, 375–384 (1992).

    CAS  PubMed  Google Scholar 

  56. Kim, J., Hake, S.B. & Roeder, R.G. The human homolog of yeast BRE1 functions as a transcriptional coactivator through direct activator interactions. Mol. Cell 20, 759–770 (2005).

    CAS  PubMed  Google Scholar 

  57. Zhu, B. et al. Monoubiquitination of human histone H2B: the factors involved and their roles in HOX gene regulation. Mol. Cell 20, 601–611 (2005).

    CAS  PubMed  Google Scholar 

  58. Downs, J.A., Lowndes, N.F. & Jackson, S.P. A role for Saccharomyces cerevisiae histone H2A in DNA repair. Nature 408, 1001–1004 (2000).

    CAS  PubMed  Google Scholar 

  59. Fernandez-Capetillo, O. et al. DNA damage-induced G2–M checkpoint activation by histone H2AX and 53BP1. Nat. Cell Biol. 4, 993–997 (2002).

    CAS  PubMed  Google Scholar 

  60. Rogakou, E.P., Pilch, D.R., Orr, A.H., Ivanova, V.S. & Bonner, W.M. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J. Biol. Chem. 273, 5858–5868 (1998).

    CAS  PubMed  Google Scholar 

  61. Lee, D.Y., Teyssier, C., Strahl, B.D. & Stallcup, M.R. Role of protein methylation in regulation of transcription. Endocr. Rev. 26, 147–170 (2005).

    CAS  PubMed  Google Scholar 

  62. Huang, S., Litt, M. & Felsenfeld, G. Methylation of histone H4 by arginine methyltransferase PRMT1 is essential in vivo for many subsequent histone modifications. Genes Dev. 19, 1885–1893 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Pavri, R. et al. Histone H2B monoubiquitination functions cooperatively with FACT to regulate elongation by RNA polymerase II. Cell 125, 703–717 (2006).

    CAS  PubMed  Google Scholar 

  64. Shi, X. et al. ING2 PHD domain links histone H3 lysine 4 methylation to active gene repression. Nature 442, 96–99 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Ayyanathan, K. et al. Regulated recruitment of HP1 to a euchromatic gene induces mitotically heritable, epigenetic gene silencing: a mammalian cell culture model of gene variegation. Genes Dev. 17, 1855–1869 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Grewal, S.I. & Moazed, D. Heterochromatin and epigenetic control of gene expression. Science 301, 798–802 (2003).

    CAS  PubMed  Google Scholar 

  67. Richards, E.J. & Elgin, S.C. Epigenetic codes for heterochromatin formation and silencing: rounding up the usual suspects. Cell 108, 489–500 (2002).

    CAS  PubMed  Google Scholar 

  68. Sims, R.J. III, Nishioka, K. & Reinberg, D. Histone lysine methylation: a signature for chromatin function. Trends Genet. 19, 629–639 (2003).

    CAS  PubMed  Google Scholar 

  69. Nielsen, S.J. et al. Rb targets histone H3 methylation and HP1 to promoters. Nature 412, 561–565 (2001).

    CAS  PubMed  Google Scholar 

  70. Vandel, L. et al. Transcriptional repression by the retinoblastoma protein through the recruitment of a histone methyltransferase. Mol. Cell. Biol. 21, 6484–6494 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. van Leeuwen, F. & Gottschling, D.E. Genome-wide histone modifications: gaining specificity by preventing promiscuity. Curr. Opin. Cell Biol. 14, 756–762 (2002).

    CAS  PubMed  Google Scholar 

  72. Huyen, Y. et al. Methylated lysine 79 of histone H3 targets 53BP1 to DNA double-strand breaks. Nature 432, 406–411 (2004).

    CAS  PubMed  Google Scholar 

  73. Wysocki, R. et al. Role of Dot1-dependent histone H3 methylation in G1 and S phase DNA damage checkpoint functions of Rad9. Mol. Cell. Biol. 25, 8430–8443 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Paik, W.K., Paik, D.C. & Kim, S. Historical review: the field of protein methylation. Trends Biochem. Sci. 32, 146–152 (2007).

    CAS  PubMed  Google Scholar 

  75. Trewick, S.C., McLaughlin, P.J. & Allshire, R.C. Methylation: lost in hydroxylation? EMBO Rep. 6, 315–320 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Bannister, A.J., Schneider, R. & Kouzarides, T. Histone methylation: dynamic or static? Cell 109, 801–806 (2002).

    CAS  PubMed  Google Scholar 

  77. Bannister, A.J. & Kouzarides, T. Reversing histone methylation. Nature 436, 1103–1106 (2005).

    CAS  PubMed  Google Scholar 

  78. Lee, M.G., Wynder, C., Cooch, N. & Shiekhattar, R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature 437, 432–435 (2005).

    CAS  PubMed  Google Scholar 

  79. Rudolph, T. et al. Heterochromatin formation in Drosophila is initiated through active removal of H3K4 methylation by the LSD1 homolog SU(VAR)3–3. Mol. Cell 26, 103–115 (2007).

    CAS  PubMed  Google Scholar 

  80. Chosed, R. & Dent, S.Y. A two-way street: LSD1 regulates chromatin boundary formation in S. pombe and Drosophila. Mol. Cell 26, 160–162 (2007).

    CAS  PubMed  Google Scholar 

  81. Nicolas, E. et al. Fission yeast homologs of human histone H3 lysine 4 demethylase regulate a common set of genes with diverse functions. J. Biol. Chem. 281, 35983–35988 (2006).

    CAS  PubMed  Google Scholar 

  82. Opel, M. et al. Genome-wide studies of histone demethylation catalysed by the fission yeast homologues of mammalian LSD1. PLoS ONE 2, e386 (2007).

    PubMed  PubMed Central  Google Scholar 

  83. Wang, J. et al. Opposing LSD1 complexes function in developmental gene activation and repression programmes. Nature 446, 882–887 (2007).

    CAS  PubMed  Google Scholar 

  84. Di Stefano, L., Ji, J.-Y., Moon, N.-S., Herr, A. & Dyson, N. Mutation of Drosophila Lsd1 disrupts H3–K4 methylation, resulting in tissue-specific defects during development. Curr. Biol. 17, 808–812 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Christensen, J. et al. RBP2 belongs to a family of demethylases, specific for tri-and dimethylated lysine 4 on histone 3. Cell 128, 1063–1076 (2007).

    CAS  PubMed  Google Scholar 

  86. Klose, R.J. et al. The retinoblastoma binding protein RBP2 is an H3K4 demethylase. Cell 128, 889–900 (2007).

    CAS  PubMed  Google Scholar 

  87. Lee, M.G., Norman, J., Shilatifard, A. & Shiekhattar, R. Physical and functional association of a trimethyl H3K4 demethylase and Ring6a/MBLR, a polycomb-like protein. Cell 128, 877–887 (2007).

    CAS  PubMed  Google Scholar 

  88. Liang, G., Klose, R.J., Gardner, K.E. & Zhang, Y. Yeast Jhd2p is a histone H3 Lys4 trimethyl demethylase. Nat. Struct. Mol. Biol. 14, 243–245 (2007).

    CAS  PubMed  Google Scholar 

  89. Secombe, J., Li, L., Carlos, L. & Eisenman, R.N. The Trithorax group protein Lid is a trimethyl histone H3K4 demethylase required for dMyc-induced cell growth. Genes Dev. 21, 537–551 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Gildea, J.J., Lopez, R. & Shearn, A. A screen for new trithorax group genes identified little imaginal discs, the Drosophila melanogaster homologue of human retinoblastoma binding protein 2. Genetics 156, 645–663 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Secombe, J. & Eisenman, R.N. The function and regulation of the JARID1 family of histone H3 lysine 4 demethylases: the Myc connection. Cell Cycle 6, 1324–1328 (2007).

    CAS  PubMed  Google Scholar 

  92. Yamane, K. et al. PLU-1 is an H3K4 demethylase involved in transcriptional repression and breast cancer cell proliferation. Mol. Cell 25, 801–812 (2007).

    CAS  PubMed  Google Scholar 

  93. Agger, K. et al. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature 449, 731–734 (2007).

    CAS  PubMed  Google Scholar 

  94. Lan, F. et al. A histone H3 lysine 27 demethylase regulates animal posterior development. Nature 449, 689–694 (2007).

    CAS  PubMed  Google Scholar 

  95. Lee, M.G. et al. Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science published online, 30 August 2007, doi: 10.1126/science1149042.

  96. Cho, Y.W. et al. PTIP associates with MLL3- and MLL4-containing histone H3 lysine 4 methyltransferase complex. J. Biol. Chem. 282, 20395–20406 (2007).

    CAS  PubMed  Google Scholar 

  97. Issaeva, I. et al. Knockdown of ALR (MLL2) reveals ALR target genes and leads to alterations in cell adhesion and growth. Mol. Cell. Biol. 27, 1889–1903 (2007).

    CAS  PubMed  Google Scholar 

  98. Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007).

    CAS  PubMed  Google Scholar 

  99. Bernstein, B.E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    CAS  PubMed  Google Scholar 

  100. Lee, J.H., Hart, S.R. & Skalnik, D.G. Histone deacetylase activity is required for embryonic stem cell differentiation. Genesis 38, 32–38 (2004).

    CAS  PubMed  Google Scholar 

  101. Kimura, H., Tada, M., Nakatsuji, N. & Tada, T. Histone code modifications on pluripotential nuclei of reprogrammed somatic cells. Mol. Cell. Biol. 24, 5710–5720 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Vakoc, C.R., Mandat, S.A., Olenchock, B.A. & Blobel, G.A. Histone H3 lysine 9 methylation and HP1γ are associated with transcription elongation through mammalian chromatin. Mol. Cell 19, 381–391 (2005).

    CAS  PubMed  Google Scholar 

  103. Smith, E. & Shilatifard, A. The A, B, Gs of silencing. Genes Dev. 21, 1141–1144 (2007).

    CAS  PubMed  Google Scholar 

  104. Su, R.C. et al. Dynamic assembly of silent chromatin during thymocyte maturation. Nat. Genet. 36, 502–506 (2004).

    CAS  PubMed  Google Scholar 

  105. Szutorisz, H. et al. Formation of an active tissue-specific chromatin domain initiated by epigenetic marking at the embryonic stem cell stage. Mol. Cell. Biol. 25, 1804–1820 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Govin, J., Caron, C., Lestrat, C., Rousseaux, S. & Khochbin, S. The role of histones in chromatin remodelling during mammalian spermiogenesis. Eur. J. Biochem. 271, 3459–3469 (2004).

    CAS  PubMed  Google Scholar 

  107. Lewis, J.D., Abbott, D.W. & Ausio, J. A haploid affair: core histone transitions during spermatogenesis. Biochem. Cell Biol. 81, 131–140 (2003).

    CAS  PubMed  Google Scholar 

  108. O'Carroll, D. et al. Isolation and characterization of Suv39h2, a second histone H3 methyltransferase gene that displays testis-specific expression. Mol. Cell. Biol. 20, 9423–9433 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Peters, A.H. et al. Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell 107, 323–337 (2001).

    CAS  PubMed  Google Scholar 

  110. Payne, C. & Braun, R.E. Histone lysine trimethylation exhibits a distinct perinuclear distribution in Plzf-expressing spermatogonia. Dev. Biol. 293, 461–472 (2006).

    CAS  PubMed  Google Scholar 

  111. Gu, L. et al. Distribution and expression of phosphorylated histone H3 during porcine oocyte maturation. Mol. Reprod. Dev. published online 6 March 2007 (doi: 10.1002/mrd.20706).

    CAS  Google Scholar 

  112. Chang, S. et al. Histone deacetylases 5 and 9 govern responsiveness of the heart to a subset of stress signals and play redundant roles in heart development. Mol. Cell. Biol. 24, 8467–8476 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Tou, L., Liu, Q. & Shivdasani, R.A. Regulation of mammalian epithelial differentiation and intestine development by class I histone deacetylases. Mol. Cell. Biol. 24, 3132–3139 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Ringrose, L. & Paro, R. Epigenetic regulation of cellular memory by the Polycomb and Trithorax group proteins. Annu. Rev. Genet. 38, 413–443 (2004).

    CAS  PubMed  Google Scholar 

  115. Klymenko, T. & Muller, J. The histone methyltransferases Trithorax and Ash1 prevent transcriptional silencing by Polycomb group proteins. EMBO Rep. 5, 373–377 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Hsieh, J.J.-D., Cheng, E.H.-Y. & Korsmeyer, S.J. Taspase1: a threonine aspartase required for cleavage of MLL and proper HOX gene expression. Cell 115, 293–303 (2003).

    CAS  PubMed  Google Scholar 

  117. Tan, X., Rotllant, J., Li, H., De Deyne, P. & Du, S.J. SmyD1, a histone methyltransferase, is required for myofibril organization and muscle contraction in zebrafish embryos. Proc. Natl. Acad. Sci. USA 103, 2713–2718 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Blais, A. et al. An initial blueprint for myogenic differentiation. Genes Dev. 19, 553–569 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Phan, D. et al. BOP, a regulator of right ventricular heart development, is a direct trans-criptional target of MEF2C in the developing heart. Development 132, 2669–2678 (2005).

    CAS  PubMed  Google Scholar 

  120. Gottlieb, P.D. et al. Bop encodes a muscle-restricted protein containing MYND and SET domains and is essential for cardiac differentiation and morphogenesis. Nat. Genet. 31, 25–32 (2002).

    CAS  PubMed  Google Scholar 

  121. Caretti, G., Di Padova, M., Micales, B., Lyons, G.E. & Sartorelli, V. The Polycomb Ezh2 methyltransferase regulates muscle gene expression and skeletal muscle differentiation. Genes Dev. 18, 2627–2638 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Baxendale, S. et al. The B-cell maturation factor Blimp-1 specifies vertebrate slow-twitch muscle fiber identity in response to Hedgehog signaling. Nat. Genet. 36, 88–93 (2004).

    CAS  PubMed  Google Scholar 

  123. Roy, S., Wolff, C. & Ingham, P.W. The u-boot mutation identifies a Hedgehog-regulated myogenic switch for fiber-type diversification in the zebrafish embryo. Genes Dev. 15, 1563–1576 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Reik, W. & Lewis, A. Co-evolution of X-chromosome inactivation and imprinting in mammals. Nat. Rev. Genet. 6, 403–410 (2005).

    CAS  PubMed  Google Scholar 

  125. Mak, W. et al. Reactivation of the paternal X chromosome in early mouse embryos. Science 303, 666–669 (2004).

    CAS  PubMed  Google Scholar 

  126. Okamoto, I., Otte, A.P., Allis, C.D., Reinberg, D. & Heard, E. Epigenetic dynamics of imprinted X inactivation during early mouse development. Science 303, 644–649 (2004).

    CAS  PubMed  Google Scholar 

  127. Huynh, K.D. & Lee, J.T. Inheritance of a pre-inactivated paternal X chromosome in early mouse embryos. Nature 426, 857–862 (2003).

    CAS  PubMed  Google Scholar 

  128. Kohlmaier, A. et al. A chromosomal memory triggered by Xist regulates histone methylation in X inactivation. PLoS Biol. [online] 2, E171 (2004).

    PubMed  PubMed Central  Google Scholar 

  129. Lee, J.T. & Lu, N. Targeted mutagenesis of Tsix leads to nonrandom X inactivation. Cell 99, 47–57 (1999).

    CAS  PubMed  Google Scholar 

  130. Liu, H., Kim, J.M. & Aoki, F. Regulation of histone H3 lysine 9 methylation in oocytes and early pre-implantation embryos. Development 131, 2269–2280 (2004).

    CAS  PubMed  Google Scholar 

  131. Fraga, M.F. et al. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat. Genet. 37, 391–400 (2005).

    CAS  PubMed  Google Scholar 

  132. Fraga, M.F. & Esteller, M. Towards the human cancer epigenome: a first draft of histone modifications. Cell Cycle 4, 1377–1381 (2005).

    CAS  PubMed  Google Scholar 

  133. Yang, X.J. & Ullah, M. MOZ and MORF, two large MYSTic HATs in normal and cancer stem cells. Oncogene 26, 5408–5419 (2007).

    CAS  PubMed  Google Scholar 

  134. Gibbons, R.J. Histone modifying and chromatin remodelling enzymes in cancer and dysplastic syndromes. Hum. Mol. Genet. 14 (review issue 1), R85–R92 (2005).

    CAS  PubMed  Google Scholar 

  135. Faravelli, F. NSD1 mutations in Sotos syndrome. Am. J. Med. Genet. C. Semin. Med. Genet. 137, 24–31 (2005).

    Google Scholar 

  136. Jaju, R.J. et al. A novel gene, NSD1, is fused to NUP98 in the t(5;11)(q35;p15.5) in de novo childhood acute myeloid leukemia. Blood 98, 1264–1267 (2001).

    CAS  PubMed  Google Scholar 

  137. Schneider, R., Bannister, A.J. & Kouzarides, T. Unsafe SETs: histone lysine methyltransferases and cancer. Trends Biochem. Sci. 27, 396–402 (2002).

    CAS  PubMed  Google Scholar 

  138. Schneider, J., Dover, J., Johnston, M. & Shilatifard, A. Global proteomic analysis of S. cerevisiae (GPS) to identify proteins required for histone modifications. Methods Enzymol. 377, 227–234 (2004).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are thankful to J. Cote for discussions while writing this review. We are grateful for L. Shilatifard's editorial assistance. The work in S.R.B.'s laboratory is supported by the American Heart Association, Scientist Development Grant (0635008N) and the American Cancer Society, Research Scholar Grant (06-52). The research in A.S.'s laboratory is supported by grants from the US National Institutes of Health (2R01CA089455 and 1R01GM069905).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ali Shilatifard.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bhaumik, S., Smith, E. & Shilatifard, A. Covalent modifications of histones during development and disease pathogenesis. Nat Struct Mol Biol 14, 1008–1016 (2007). https://doi.org/10.1038/nsmb1337

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb1337

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