Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Nov 20.
Published in final edited form as: Epigenetics. 2009 Jul 5;4(5):273–276. doi: 10.4161/epi.4.5.9212

Degrees make all the difference

The multifunctionality of histone H4 lysine 20 methylation

Yu Wang 1, Songtao Jia 1,*
PMCID: PMC5116398  NIHMSID: NIHMS827009  PMID: 19571682

Abstract

Residue and degree-specific methylation of histone lysines along with other epigenetic modifications organizes chromatin into distinct domains and regulates almost every aspect of DNA metabolism. Identification of histone methyltransferases and demethylases, as well as proteins that recognize methylated lysines, has clarified the role of each methylation event in regulating different biological pathways. Methylation of histone H4 lysine 20 (H4K20me) plays critical roles in diverse cellular processes such as gene expression, cell cycle progression and DNA damage repair, with each of the three degrees of methylation (mono-, di- and tri-methylation) making a unique contribution. Here we discuss recent studies of H4K20me that have greatly improved our understanding of the regulation and function of this fascinating histone modification.

Keywords: chromatin, histone, H4K20, methylation, epigenetics


Eukaryotic genomes are compacted in the form of chromatin in order to accommodate large amounts of DNA within the nucleus. The fundamental unit of chromatin is the nucleosome, which is composed of ~146 base pairs of DNA surrounding an octamer of histones (two copies each of H2A, H2B, H3 and H4). Chromatin structure and function can be modulated by post-translational modification of histones, such as acetylation, methylation, phosphorylation and ubiquitylation, which affect interactions between individual nucleosomes or between nucleosomes and regulatory proteins.1 Among histone modifications, lysine methylation shows the most remarkable specificity. The most extensively studied methylation sites are lysine 4, 9, 27, 36 and 79 of histone H3 and lysine 20 of histone H4, each of which can be found in one of three methylated degrees (mono-, di- or tri-methylation). Each residue and degree-specific lysine methylation requires specific enzymes for addition or removal of methyl groups,24 and genome-wide localization analyses indicate that each has a distinct distribution pattern, indicative of unique functions.59

Lysine 20 is the only lysine residue on histone H4 tail that can be methylated. However, among methylated histone lysines, the function of H4K20me is the most enigmatic. Each degree of methylation at this residue contributes distinctly to the regulation of diverse cellular processes. For example, H4K20me1 (H4K20 mono-methylation) regulates cell cycle progression as well as gene expression,1016 whereas H4K20me2 and H4K20me3 are required for DNA damage checkpoint activation and maintenance of heterochromatin structures, respectively1719 (Table 1). Better understanding of the mechanisms that regulate H4K20me will enable us to gain mechanistic insights into how different degrees of histone methylation are controlled and interpreted by the cellular machinery.

Table 1.

Three degrees of H4K20 methylation and their functions

Modification Methyltransferases Effectors Functions
H4K20me1 hs/mm PR-Set7/Set8 dm PR-Set7
sp Set9
hs/mm L3MBTL1
sp Pdp1
Cell cycle progression
Chromosome condensation
Transcription
H4K20me2 hs/mm Suv4-20h1/2 dm SUV4-20
sp Set9
hs/mm 53BP1
sp Crb2
DNA damage checkpoint
H4K20me3 hs/mm Suv4-20h1/2, dm SUV4-20
sp Set9
Heterochromatin maintenance
Telomere stability

hs: Homo sapiens; mm: Mus musculus; dm: Drosophila melanogaster; sp: Schizosaccharomyces pombe.

The H4K20 Histone Methyltransferases

The majority of methyltransferases that catalyze histone lysine methylation contain an evolutionarily conserved SET domain.20 Two groups of SET domain proteins catalyze H4K20 methylation in higher eukaryotes, both of which require nucleosomes but not free histones as substrates in vitro.19,21,22 The first group, which includes mammalian PR-Set7/Set8 and Drosophila PR-Set7 (also known as KMT5A), is responsible for H4K20me1 in mammals and Drosophila.2124 Structural analysis demonstrates that a tyrosine residue in the active site of the PR-Set7/Set8 is responsible for preventing further addition of methyl groups beyond mono-methylation.23,24 Mutational analysis demonstrates that replacement of the tyrosine with phenylalanine facilitates the dissociation of a water molecule to provide enough room to accommodate higher methylated product.25 The second group, which includes mammalian Suv4-20h enzymes (Suv4-20h1/KMT5B and Suv4-20h2/KMT5C) and Drosophila Suv4-20 (KMT5B), is responsible for both H4K20me2 and H4K20me3.19,26,27 Although they are capable of catalyzing H4K20me2/3 with unmodified nucleosomes as substrates in vitro,19 higher degrees of H4K20me in vivo seem to require H4K20me1. This is supported by the fact that loss of PR-Set7/Set8 results in diminished levels of H4K20me2/3,12,28 and that knockout of Suv4-20hs in mice and Suv4-20 in Drosophila result in excessive accumulation of H4K20me1.26,29 The mechanism underlying the requirement of H4K20me1 for higher degrees of H4K20 methylation in these organisms is unknown, but the results suggest the sequential action of the two classes of H4K20 methyltransferases. Consistently, recent metabolic labeling coupled with mass spectrometry analysis demonstrates that newly synthesized histones are progressively methylated to higher degrees: histone H4 incorporated into chromatin in an unmodified form during S phase is first monomethylated at G2/M phase and then gradually converted to di- and tri-methylated forms over a few cell cycles.30 H4K20me3 also requires interaction of Suv4-20h with HP1 proteins, which are necessary for localization of Suv4-20hs to heterochromatin regions.19 This might help create high local concentrations of Suv4-20h for the addition of the third methyl group. Although H4K20me2 is the most abundant form of H4K20me in mammals and Drosophila,26,30 how it is specifically controlled is not well understood.

In Schizosaccharomyces pombe, Set9/KMT5 is the sole histone methyltransferase responsible for all three degrees of H4K20 methylation.18 Sequence analysis indicates that Set9 is similar to Suv4-20h and mutations of phenylalanines to tyrosines near the active site can modulate its specificity toward lower degrees of H4K20me.31 All three degrees of H4K20me in vivo also depend on an Set9-associated PWWP domain protein Pdp1, which is required for the recruitment of Set9 to chromatin.32 The PWWP domain of Pdp1 is a novel methyl-lysine binding motif with specificity towards H4K20me1. Mutations within the PWWP domain that disrupt the PWWP-H4K20me1 interaction delocalize Set9 from chromatin and result in the selective loss of H4K20me3 and a concomitant increase of H4K20me1.32 These results suggest that Pdp1 regulates Set9 function in two ways: first, the association of Pdp1 with Set9 might directly affect the ability of Set9 to perform H4K20me1, which is independent of the PWWP domain. Second, the PWWP domain of Pdp1 binds to H4K20me1 to increase the local concentration of Set9, enabling the enzyme to catalyze H4K20me3.32 Thus the H4K20me1 to H4K20me3 transition seems to be a conserved mechanism in controlling H4K20 methylation, which depends on the recruitment of high levels of histone methyltransferases to chromatin. Similar to higher eukaryotes, the selective regulation of H4K20me2 in fission yeast is not understood.

The Function of Different Degrees of H4K20me

The specificity of the various H4K20 methyltransferases has made it possible to dissect the functions of each degree of H4K20 methylation. For example, both the expression and activity of PR-Set7/Set8 are cell cycle regulated.13 The peak expression of PR-Set7/Set8 is during G2/M phase with the enzyme preferentially localizing to mitotic chromosomes.13 This corresponds to the conversion of unmethylated histone H4 deposited during S phase into H4K20me1 at G2/M transition, leading to increased levels of H4K20me1 at this stage.13,30 Consistently, removal of PR-Set7 in mice and Drosophila results in lethality, which can be attributed to defects in cell cycle progression.12,28,33 In addition, these mutant animals also show improper chromosome condensation, consistent with a role of H4K20me1 in regulating chromatin compaction.12,33

The Suv4-20 enzymes are localized preferentially to constitutive heterochromatin.19 This depends on the interaction between Suv4-20hs and HP1 proteins, which are hallmarks of heterochromatin structures.19 Consequently, H4K20me3 is enriched at heterochromatin and a mutation of Suv4-20 in Drosophila results in alternations in position effect variegation, indicative of defects in heterochromatin structures.19 Mice deficient for Suv4-20hs display loss of H4K20me2 and H4K20me3 as well as embryonic lethality.26 This can be attributed to defects in maintaining genome integrity, which in turn leads to chromosomal aberrations, consistent with the role of Suv4-20h in regulating cellular response to DNA damage.26 As a result, cells that require programmed DNA rearrangements, such as B cells and lymphoid progenitors, are severely depleted in Suv4-20hs knockout mice.26

In fission yeast, loss of Set9 results in defective DNA damage checkpoint and cells that are sensitive to DNA damage treatment.18 The ability to alter the degree of methylation by mutation of the Tyr/Phe switch in Set9 or mutations in the PWWP domain of Pdp1 shows that H4K20me2 is the key modification responsible for checkpoint function.31,32 Interestingly, Set9 is not required for heterochromatin assembly and H4K20me3 is not enriched at heterochromatin regions in fission yeast,18 suggesting that H4K20me3 might have other functions in this organism.

How then do different degrees of H4K20me contribute to different cellular processes? An expanding body of evidence supports the idea that methylated lysines recruit effector proteins to regulate chromatin functions.34 Like many methylated histone lysines, H4K20me also recruits effector proteins, which might directly impact chromatin organization or engage other proteins with chromatin-modifying activities.

As discussed above, the PWWP domain of the fission yeast Pdp1 specifically recognizes H4K20me1 to strengthen the interaction between Set9 and chromatin, which in turn promotes H4K20me3.32 In addition, the MBT domain of mammalian L3MBTL1 was found to bind H4K20me1/2 as well as other mono- or di-methylated lysine residues in vitro11,16,35,36 and chromatin association of L3MBTL1 in vivo correlates with H4K20me1.11 Through interactions with H4K20me1/2 and H1K26me1/2, the MBT domains of L3MBTL1 concurrently bind at least two nucleosomes to compact nucleosome arrays in vitro16 and PR-Set7 promotes L3MBTL1-mediated repression in vivo,11 consistent with the role of H4K20me1 in repressing gene expression. However, it should be noted that H4K20me1 is also found at actively transcribed genes, although whether it is involved in transcription regulation is not understood.5,37 Interestingly, L3MBTL1 also associates with PR-Set7,38 which might be responsible for reestablishing H4K20me1 patterns after the incorporation of newly synthesized histones during S phase of the cell cycle, thus serving as a platform for the inheritance of H4K20me1.39 In addition, Drosophila dl(3)MBT recruits histone deacetylase dRPD3 to chromatin in an H4K20me1-dependent manner to facilitate chromatin maturation.38

While the regulatory mechanisms that enable H4K20 methyltransferases to specifically catalyze H4K20me2 remain unknown, the function of H4K20me2 is the most conserved among the three degrees of H4K20me. Biochemical and structural analysis demonstrate that 53BP1 in mammals and Crb2 in fission yeast selectively interact with H4K20me2 through a tandem Tudor domain.17,31,40 Both 53BP1 and Crb2 are DNA damage checkpoint proteins, which in the presence of DNA damage signal a delay of the cell cycle to allow sufficient time for repair and prevent the propagation of DNA damage.41 In both mammals and fission yeast, loss of H4K20me2 abrogates 53BP1/Crb2 recruitment to DNA damaged foci, thus blocking checkpoint signaling.18,31,32,42

It is interesting to note that bulk levels of H4K20me2 do not change upon DNA damage treatment.18 In fact, histone H4 containing K20me2 accounts for more than 80% of the total histone H4 in mammalian and Drosophila cells,26,27 which makes it unlikely to be the sole mark for the recruitment of 53BP1/Crb2 to sites of DNA damage. In fission yeast, the localization of Crb2 requires another DNA damage induced modification, the phosphorylation of H2A (γH2A.X in mammals), in addition to H4K20me2.42 As H4K20 is buried in the context of stacked nucleosomes,43 it has been proposed that DNA damage results in the opening up of chromatin to expose H4K20me2, which together with phosphorylated H2A enables high affinity binding of Crb2 to signal DNA damage checkpoint activation.18

H4K20me3 is enriched in constitutive heterochromatin in metazoans5,8,14,19,44 and regulates the stability of telomeres and heterochromatin structures.19,45 However, proteins specifically recognizing H4K20me3 to regulate heterochromatin assembly have not been found. Although in vitro studies demonstrate that the Tudor domain of histone demethylase Jmjd2A interacts with H4K20me2/3, the functional significance of this interaction is not clear.32,40

Among methylated lysines of histone tails, H4K20 is uniquely positioned close to the nucleosome core and buried inside the nucleosome-nucleosome interface,43 raising the possibility that H4K20me can directly impact chromatin organization. Earlier studies demonstrated that H4K20me competes with H4K16 acetylation,13,22 a modification that is critical for chromatin compaction,46 although mass spectrometry studies showed that H4K16 acetylation and H4K20me coexist.27,30,47 Interestingly, recent structural studies show that the presence of H4K20me3 results in local structural alterations within nucleosomes.48 Further biochemical analysis of nucleosome arrays reconstituted with recombinant H4K20me2/3-containing histones demonstrates that H4K20me directly promotes chromatin compaction, which is consistent with its role in regulating higher-order chromatin assembly.48

Is there a Possible H4K20 Demethylase?

Recently, two classes of proteins that demethylate histones were identified.2,4 LSD1/KDM1 (lysine specific demethylase 1) contains a SWIRM domain that reverses histone H3K4me by an oxidative demethylation reaction in which flavin serves as a cofactor, whereas JmjC domain-containing proteins catalyze lysine demethylation via an oxidative reaction that requires Fe(II) and α-ketoglutarate. The last two years have seen a growing number of JmjC domain proteins identified as histone demethylases that target different lysine residues.2,4 Their complexity and specificity rivals that of SET domain proteins. However, a demethylase for H4K20me has not yet been identified. Although mass spectrometry analysis indicates that this mark is relatively stable in vivo,27 a similar argument was used against the existence of histone demethylases before their identification. The difficulty in the identification of an H4K20 demethylase might be due to technical challenges, and it remains an area that needs to be further explored.49

Concluding Remarks

Since the identification of H4K20 methylation and the H4K20 methyltransferases, we have gained more and more insights into the regulation of the distinct degrees of H4K20me and the molecular mechanisms underlying their functions. Being uniquely located at the nucleosome interaction interface, H4K20me has the ability to directly affect chromatin compaction. As with other methylated histone lysines, different degrees of H4K20me serve as binding sites for many effector proteins that can recruit other chromatin associated activities. Identification of novel proteins that either regulate H4K20 methyltransferases or recognize different degrees of H4K20me will further improve our understanding of the role of H4K20 methylation in the regulation and maintenance of epigenetic states.

H4K20me is essential for development of multi-cellular organisms as removal of H4K20 methyltransferases results in lethality.12,22,26 The loss of H4K20me3 is also a hallmark of many human cancers.50,51 Further study into the mechanisms that regulate H4K20 methylation and its biological function will provide new avenues to develop therapeutic treatment for human diseases.

Acknowledgments

We thank Scott Kallgren, Bharat Reddy and Hengbin Wang for comments on the manuscript. We apologize to those whose work was not cited due to space limitations. Work in the Jia laboratory is supported by the National Institutes of Health grant R01 GM085145 and a Basil O’Connor Starter Scholar Award from the March of Dimes foundation.

Footnotes

Previously published online as an Epigenetics E-publication: http://www.landesbioscience.com/journals/epigenetics/article/9212

References

  • 1.Allis C, Jenuwein T, Reinberg D. Epigenetics. Cold Spring Harbor, NY: Cold Spring Harbor laboratory press; 2007. [Google Scholar]
  • 2.Klose RJ, Zhang Y. Regulation of histone methylation by demethylimination and demethylation. Nat Rev Mol Cell Biol. 2007;8:307–18. doi: 10.1038/nrm2143. [DOI] [PubMed] [Google Scholar]
  • 3.Martin C, Zhang Y. The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol. 2005;6:838–49. doi: 10.1038/nrm1761. [DOI] [PubMed] [Google Scholar]
  • 4.Shi Y, Whetstine JR. Dynamic regulation of histone lysine methylation by demethylases. Mol Cell. 2007;25:1–14. doi: 10.1016/j.molcel.2006.12.010. [DOI] [PubMed] [Google Scholar]
  • 5.Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823–37. doi: 10.1016/j.cell.2007.05.009. [DOI] [PubMed] [Google Scholar]
  • 6.Bernstein BE, Kamal M, Lindblad-Toh K, Bekiranov S, Bailey DK, Huebert DJ, et al. Genomic maps and comparative analysis of histone modifications in human and mouse. Cell. 2005;120:169–81. doi: 10.1016/j.cell.2005.01.001. [DOI] [PubMed] [Google Scholar]
  • 7.Liu CL, Kaplan T, Kim M, Buratowski S, Schreiber SL, Friedman N, Rando OJ. Single-nucleosome mapping of histone modifications in S. cerevisiae. PLoS Biol. 2005;3:328. doi: 10.1371/journal.pbio.0030328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448:553–60. doi: 10.1038/nature06008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Pokholok DK, Harbison CT, Levine S, Cole M, Hannett NM, Lee TI, et al. Genome-wide map of nucleosome acetylation and methylation in yeast. Cell. 2005;122:517–27. doi: 10.1016/j.cell.2005.06.026. [DOI] [PubMed] [Google Scholar]
  • 10.Houston SI, McManus KJ, Adams MM, Sims JK, Carpenter PB, Hendzel MJ, Rice JC. Catalytic function of the PR-Set7 histone H4 lysine 20 monomethyltransferase is essential for mitotic entry and genomic stability. J Biol Chem. 2008;283:19478–88. doi: 10.1074/jbc.M710579200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kalakonda N, Fischle W, Boccuni P, Gurvich N, Hoya-Arias R, Zhao X, et al. Histone H4 lysine 20 monomethylation promotes transcriptional repression by L3MBTL1. Oncogene. 2008;27:4293–304. doi: 10.1038/onc.2008.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Oda H, Okamoto I, Murphy N, Chu J, Price SM, Shen MM, et al. Monomethylation of Histone H4-Lysine 20 is involved in Chromosome Structure and Stability and is essential for Mouse Development. Mol Cell Biol. 2009 doi: 10.1128/MCB.01768-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rice JC, Nishioka K, Sarma K, Steward R, Reinberg D, Allis CD. Mitotic-specific methylation of histone H4 Lys 20 follows increased PR-Set7 expression and its localization to mitotic chromosomes. Genes Dev. 2002;16:2225–30. doi: 10.1101/gad.1014902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sims JK, Houston SI, Magazinnik T, Rice JC. A trans-tail histone code defined by monomethylated H4 Lys-20 and H3 Lys-9 demarcates distinct regions of silent chromatin. J Biol Chem. 2006;281:12760–6. doi: 10.1074/jbc.M513462200. [DOI] [PubMed] [Google Scholar]
  • 15.Sims JK, Rice JC. PR-Set7 establishes a repressive trans-tail histone code that regulates differentiation. Mol Cell Biol. 2008;28:4459–68. doi: 10.1128/MCB.00410-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Trojer P, Li G, Sims RJ, 3rd, Vaquero A, Kalakonda N, Boccuni P, et al. L3MBTL1, a histone-methylation-dependent chromatin lock. Cell. 2007;129:915–28. doi: 10.1016/j.cell.2007.03.048. [DOI] [PubMed] [Google Scholar]
  • 17.Botuyan MV, Lee J, Ward IM, Kim JE, Thompson JR, Chen J, Mer G. Structural basis for the methylation state-specific recognition of histone H4-K20 by 53BP1 and Crb2 in DNA repair. Cell. 2006;127:1361–73. doi: 10.1016/j.cell.2006.10.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sanders SL, Portoso M, Mata J, Bahler J, Allshire RC, Kouzarides T. Methylation of histone H4 lysine 20 controls recruitment of Crb2 to sites of DNA damage. Cell. 2004;119:603–14. doi: 10.1016/j.cell.2004.11.009. [DOI] [PubMed] [Google Scholar]
  • 19.Schotta G, Lachner M, Sarma K, Ebert A, Sengupta R, Reuter G, et al. A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev. 2004;18:1251–62. doi: 10.1101/gad.300704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jenuwein T, Laible G, Dorn R, Reuter G. SET domain proteins modulate chromatin domains in eu- and heterochromatin. Cell Mol Life Sci. 1998;54:80–93. doi: 10.1007/s000180050127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fang J, Feng Q, Ketel CS, Wang H, Cao R, Xia L, et al. Purification and functional characterization of SET8, a nucleosomal histone H4-lysine 20-specific methyltransferase. Curr Biol. 2002;12:1086–99. doi: 10.1016/s0960-9822(02)00924-7. [DOI] [PubMed] [Google Scholar]
  • 22.Nishioka K, Rice JC, Sarma K, Erdjument-Bromage H, Werner J, Wang Y, et al. PR-Set7 is a nucleosome-specific methyltransferase that modifies lysine 20 of histone H4 and is associated with silent chromatin. Mol Cell. 2002;9:1201–13. doi: 10.1016/s1097-2765(02)00548-8. [DOI] [PubMed] [Google Scholar]
  • 23.Couture JF, Collazo E, Brunzelle JS, Trievel RC. Structural and functional analysis of SET8, a histone H4 Lys-20 methyltransferase. Genes Dev. 2005;19:1455–65. doi: 10.1101/gad.1318405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Xiao B, Jing C, Kelly G, Walker PA, Muskett FW, Frenkiel TA, et al. Specificity and mechanism of the histone methyltransferase Pr-Set7. Genes Dev. 2005;19:1444–54. doi: 10.1101/gad.1315905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Couture JF, Dirk LM, Brunzelle JS, Houtz RL, Trievel RC. Structural origins for the product specificity of SET domain protein methyltransferases. Proc Natl Acad Sci USA. 2008;105:20659–64. doi: 10.1073/pnas.0806712105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Schotta G, Sengupta R, Kubicek S, Malin S, Kauer M, Callen E, et al. A chromatin-wide transition to H4K20 monomethylation impairs genome integrity and programmed DNA rearrangements in the mouse. Genes Dev. 2008;22:2048–61. doi: 10.1101/gad.476008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Yang H, Pesavento JJ, Starnes TW, Cryderman DE, Wallrath LL, Kelleher NL, Mizzen CA. Preferential dimethylation of histone H4 lysine 20 by Suv4-20. J Biol Chem. 2008;283:12085–92. doi: 10.1074/jbc.M707974200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Karachentsev D, Sarma K, Reinberg D, Steward R. PR-Set7-dependent methylation of histone H4 Lys 20 functions in repression of gene expression and is essential for mitosis. Genes Dev. 2005;19:431–5. doi: 10.1101/gad.1263005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sakaguchi A, Karachentsev D, Seth-Pasricha M, Druzhinina M, Steward R. Functional characterization of the Drosophila Hmt4-20/Suv4-20histone methyltransferase. Genetics. 2008;179:317–22. doi: 10.1534/genetics.108.087650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pesavento JJ, Yang H, Kelleher NL, Mizzen CA. Certain and progressive methylation of histone H4 at lysine 20 during the cell cycle. Mol Cell Biol. 2008;28:468–86. doi: 10.1128/MCB.01517-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Greeson NT, Sengupta R, Arida AR, Jenuwein T, Sanders SL. Di-methyl H4 lysine 20 targets the checkpoint protein Crb2 to sites of DNA damage. J Biol Chem. 2008;283:33168–74. doi: 10.1074/jbc.M806857200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wang Y, Reddy B, Thompson J, Wang H, Noma K, Yates JR, 3rd, Jia S. Regulation of Set9-mediated H4K20 methylation by a PWWP domain protein. Mol Cell. 2009;33:428–37. doi: 10.1016/j.molcel.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sakaguchi A, Steward R. Aberrant monomethylation of histone H4 lysine 20 activates the DNA damage checkpoint in Drosophila melanogaster. J Cell Biol. 2007;176:155–62. doi: 10.1083/jcb.200607178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ. How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers. Nat Struct Mol Biol. 2007;14:1025–40. doi: 10.1038/nsmb1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Li H, Fischle W, Wang W, Duncan EM, Liang L, Murakami-Ishibe S, et al. Structural basis for lower lysine methylation state-specific readout by MBT repeats of L3MBTL1 and an engineered PHD finger. Mol Cell. 2007;28:677–91. doi: 10.1016/j.molcel.2007.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Min J, Allali-Hassani A, Nady N, Qi C, Ouyang H, Liu Y, et al. L3MBTL1 recognition of mono- and dimethylated histones. Nat Struct Mol Biol. 2007;14:1229–30. doi: 10.1038/nsmb1340. [DOI] [PubMed] [Google Scholar]
  • 37.Vakoc CR, Sachdeva MM, Wang H, Blobel GA. Profile of histone lysine methylation across transcribed mammalian chromatin. Mol Cell Biol. 2006;26:9185–95. doi: 10.1128/MCB.01529-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Scharf AN, Meier K, Seitz V, Kremmer E, Brehm A, Imhof A. Monomethylation of lysine 20 on histone H4 facilitates chromatin maturation. Mol Cell Biol. 2009;29:57–67. doi: 10.1128/MCB.00989-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Trojer P, Reinberg D. Histone lysine demethylases and their impact on epigenetics. Cell. 2006;125:213–7. doi: 10.1016/j.cell.2006.04.003. [DOI] [PubMed] [Google Scholar]
  • 40.Kim J, Daniel J, Espejo A, Lake A, Krishna M, Xia L, et al. Tudor, MBT and chromo domains gauge the degree of lysine methylation. EMBO Rep. 2006;7:397–403. doi: 10.1038/sj.embor.7400625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Zhou BB, Elledge SJ. The DNA damage response: putting checkpoints in perspective. Nature. 2000;408:433–9. doi: 10.1038/35044005. [DOI] [PubMed] [Google Scholar]
  • 42.Du LL, Nakamura TM, Russell P. Histone modification-dependent and -independent pathways for recruitment of checkpoint protein Crb2 to double-strand breaks. Genes Dev. 2006;20:1583–96. doi: 10.1101/gad.1422606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Dorigo B, Schalch T, Bystricky K, Richmond TJ. Chromatin fiber folding: requirement for the histone H4 N-terminal tail. J Mol Biol. 2003;327:85–96. doi: 10.1016/s0022-2836(03)00025-1. [DOI] [PubMed] [Google Scholar]
  • 44.Martens JH, O’Sullivan RJ, Braunschweig U, Opravil S, Radolf M, Steinlein P, Jenuwein T. The profile of repeat-associated histone lysine methylation states in the mouse epigenome. EMBO J. 2005;24:800–12. doi: 10.1038/sj.emboj.7600545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Benetti R, Gonzalo S, Jaco I, Schotta G, Klatt P, Jenuwein T, Blasco MA. Suv4-20h deficiency results in telomere elongation and derepression of telomere recombination. J Cell Biol. 2007;178:925–36. doi: 10.1083/jcb.200703081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Shogren-Knaak M, Ishii H, Sun JM, Pazin MJ, Davie JR, Peterson CL. Histone H4-K16 acetylation controls chromatin structure and protein interactions. Science. 2006;311:844–7. doi: 10.1126/science.1124000. [DOI] [PubMed] [Google Scholar]
  • 47.Phanstiel D, Brumbaugh J, Berggren WT, Conard K, Feng X, Levenstein ME, et al. Mass spectrometry identifies and quantifies 74 unique histone H4 isoforms in differentiating human embryonic stem cells. Proc Natl Acad Sci USA. 2008;105:4093–8. doi: 10.1073/pnas.0710515105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lu X, Simon MD, Chodaparambil JV, Hansen JC, Shokat KM, Luger K. The effect of H3K79 dimethylation and H4K20 trimethylation on nucleosome and chromatin structure. Nat Struct Mol Biol. 2008;15:1122–4. doi: 10.1038/nsmb.1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lan F, Shi Y. Epigenetic regulation: methylation of histone and non-histone proteins. Sci China C Life Sci. 2009;52:311–22. doi: 10.1007/s11427-009-0054-z. [DOI] [PubMed] [Google Scholar]
  • 50.Fraga MF, Ballestar E, Villar-Garea A, Boix-Chornet M, Espada J, Schotta G, et al. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet. 2005;37:391–400. doi: 10.1038/ng1531. [DOI] [PubMed] [Google Scholar]
  • 51.Merdzhanova G, Edmond V, De Seranno S, Van den Broeck A, Corcos L, Brambilla C, et al. E2F1 controls alternative splicing pattern of genes involved in apoptosis through upregulation of the splicing factor SC35. Cell Death Differ. 2008;15:1815–23. doi: 10.1038/cdd.2008.135. [DOI] [PubMed] [Google Scholar]

RESOURCES