We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Molecular coupling of DNA methylation and histone methylation

    Hideharu Hashimoto

    Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA

    ,
    Paula M Vertino

    Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA

    The Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA

    &
    Published Online:https://doi.org/10.2217/epi.10.44

    The combinatorial pattern of DNA and histone modifications constitutes an epigenetic ‘code’ that shapes gene-expression patterns by enabling or restricting the transcriptional potential of genomic domains. DNA methylation is associated with histone modifications, particularly the absence of histone H3 lysine 4 methylation (H3K4me0) and the presence of H3K9 methylation. This article focuses on three protein domains (ATRX–Dnmt3–Dnmt3L [ADD], Cys–X–X–Cys [CXXC] and the methyl-CpG-binding domain [MBD]) and the functional implications of domain architecture in the mechanisms linking histone methylation and DNA methylation in mammalian cells. The DNA methyltransferase DNMT3a and its accessory protein Dnmt3L contain a H3K4me0-interacting ADD domain that links the DNA methylation reaction with unmodified H3K4. The H3K4 methyltransferase MLL1 contains a CpG-interacting CXXC domain that may couple the H3K4 methylation reaction to unmethylated DNA. Another H3K4 methyltransferase, SET1, although lacking an intrinsic CXXC domain, interacts directly with an accessory protein CFP1 that contains the same domain. The H3K9 methyltransferase SETDB1 contains a putative MBD that potentially links the H3K4 methylation reaction to methylated DNA or may do so through the interaction with the MBD containing protein MBD1. Finally, we consider the domain structure of the DNA methyltransferase DNMT1, its accessory protein UHRF1 and their associated proteins, and propose a mechanism by which DNA methylation and histone methylation may be coordinately maintained through mitotic cell division, allowing for the transmission of parental DNA and for the histone methylation patterns to be copied to newly replicated chromatin.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Clapier CR, Chakravarthy S, Petosa C, Fernandez-Tornero C, Luger K, Muller CW: Structure of the Drosophila nucleosome core particle highlights evolutionary constraints on the H2A-H2B histone dimer. Proteins71,1–7 (2008).
    • Strahl BD, Allis CD: The language of covalent histone modifications. Nature403,41–45 (2000).
    • Margueron R, Reinberg D: Chromatin structure and the inheritance of epigenetic information. Nat. Rev. Genet.11,285–296 (2010).
    • Probst AV, Dunleavy E, Almouzni G: Epigenetic inheritance during the cell cycle. Nat. Rev. Mol. Cell Biol.10,192–206 (2009).
    • Barreto G, Schäfer A, Marhold J et al.: Gadd45a promotes epigenetic gene activation by repair-mediated DNA demethylation. Nature445,671–675 (2007).
    • Yuan P, Han J, Guo G et al.: Eset partners with Oct4 to restrict extraembryonic trophoblast lineage potential in embryonic stem cells. Genes Dev.23,2507–2520 (2009).
    • Schaefer CB, Ooi SK, Bestor TH, Bourc’his D: Epigenetic decisions in mammalian germ cells. Science316,398–399 (2007).
    • Ehrlich M: DNA methylation and cancer-associated genetic instability. Adv. Exp. Med. Biol.570,363–392 (2005).
    • Gronbaek K, Hother C, Jones PA: Epigenetic changes in cancer. APMIS115,1039–1059 (2007).
    • 10  Kurkjian C, Kummar S, Murgo AJ: DNA methylation: its role in cancer development and therapy. Curr. Probl. Cancer32,187–235 (2008).
    • 11  Gal-Yam EN, Saito Y, Egger G, Jones PA: Cancer epigenetics: modifications, screening, and therapy. Annu. Rev. Med.59,267–280 (2008).
    • 12  Dobrovic A, Kristensen LS: DNA methylation, epimutations and cancer predisposition. Int. J. Biochem. Cell Biol.41,34–39 (2009).
    • 13  McCabe MT, Brandes JC, Vertino PM: Cancer DNA methylation: molecular mechanisms and clinical implications. Clin. Cancer Res.15,3927–3937 (2009).
    • 14  Sharma S, Kelly TK, Jones PA: Epigenetics in cancer. Carcinogenesis31,27–36 (2010).
    • 15  Urdinguio RG, Sanchez-Mut JV, Esteller M: Epigenetic mechanisms in neurological diseases: genes, syndromes, and therapies. Lancet Neurol.8,1056–1072 (2009).
    • 16  Sananbenesi F, Fischer A: The epigenetic bottleneck of neurodegenerative and psychiatric diseases. Biol. Chem.390,1145–1153 (2009).
    • 17  Bernstein BE, Meissner A, Lander ES: The mammalian epigenome. Cell128,669–681 (2007).
    • 18  Meissner A, Mikkelsen TS, Gu H et al.: Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature454,766–770 (2008).▪▪ One of the first studies to describe genome-scale DNA methylation patterns at base pair resolution and to compare DNA methylation and histone methylation patterns as they change during cellular differentiation.
    • 19  Fan S, Zhang MQ, Zhang X: Histone methylation marks play important roles in predicting the methylation status of CpG islands. Biochem. Biophys. Res. Commun.374,559–564 (2008).
    • 20  Cedar H, Bergman Y: Linking DNA methylation and histone modification: patterns and paradigms. Nat. Rev. Genet.10,295–304 (2009).
    • 21  Dindot SV, Person R, Strivens M, Garcia R, Beaudet AL: Epigenetic profiling at mouse imprinted gene clusters reveals novel epigenetic and genetic features at differentially methylated regions. Genome Res.19,1374–1383 (2009).
    • 22  Laurent L, Wong E, Li G et al.: Dynamic changes in the human methylome during differentiation. Genome Res.20,320–331 (2010).
    • 23  Cheng X, Blumenthal RM: Coordinated chromatin control: structural and functional linkage of DNA and histone methylation. Biochemistry49,2999–3008 (2010).
    • 24  Tamaru H, Selker EU: A histone H3 methyltransferase controls DNA methylation in Neurospora crassa.Nature414,277–283 (2001).
    • 25  Tamaru H, Zhang X, Debra McMillen et al.: Trimethylated lysine 9 of histone H3 is a mark for DNA methylation in Neurospora crassa.Nat. Genet.34,75–79 (2003).
    • 26  Jackson JP, Lindroth AM, Cao X, Jacobsen SE: Control of CpNpG DNA methylation by the KRYPTONITE histone H3 methyltransferase. Nature416,556–560 (2002).
    • 27  Lehnertz B, Ueda Y, Derijck AA et al.: Suv39h-mediated histone H3 lysine 9 methylation directs DNA methylation to major satellite repeats at pericentric heterochromatin. Curr. Biol.13,1192–1200 (2003).
    • 28  Matsui T, Leung D, Miyashita H et al.: Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET. Nature464,927–931 (2010).▪▪ Suggests a DNA methylation-independent H3K9 methylation pathway involving ESET (also known as SETDB1).
    • 29  Nguyen CT, Weisenberger DJ, Velicescu M et al.: Histone H3-lysine 9 methylation is associated with aberrant gene silencing in cancer cells and is rapidly reversed by 5-aza-2´-deoxycytidine. Cancer Res.62,6456–6461 (2002).
    • 30  Chen T, Ueda Y, Xie S, Li E: A novel Dnmt3a isoform produced from an alternative promoter localizes to euchromatin and its expression correlates with active de novo methylation. J. Biol. Chem.277,38746–38754 (2002).
    • 31  Ooi SK, Qiu C, Bernstein E et al.: DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature448,714–717 (2007).▪▪ Suggests a model whereby absence of H3K4 methylation acts as a signal to recruit DNMT3L and induce DNA methylation by DNMT3a.
    • 32  Hu JL, Zhou BO, Zhang RR, Zhang KL, Zhou JQ, Xu GL: The N-terminus of histone H3 is required for de novo DNA methylation in chromatin. Proc. Natl Acad. Sci. USA106,22187–22192 (2009).▪▪ Discusses how the authors tested histone–Dnmt3L–Dnmt3a–DNA interactions in the budding yeast Saccharomyces cerevisiae, and found that induced DNA methylation in yeast preferentially occurs in heterochromatic regions where H3K4 methylation is rare.
    • 33  Proffitt JH, Davie JR, Swinton D, Hattman S: 5-methylcytosine is not detectable in Saccharomyces cerevisiae DNA. Mol. Cell Biol.4,985–988 (1984).
    • 34  Bulkowska U, Ishikawa T, Kurlandzka A, Trzcinska-Danielewicz J, Derlacz R, Fronk J: Expression of murine DNA methyltransferases Dnmt1 and Dnmt3a in the yeast Saccharomyces cerevisiae.Yeast24,871–882 (2007).
    • 35  Wang J, Hevi S, Kurash JK et al.: The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat. Genet.41,125–129 (2009).▪ Along with [36], demonstrates that LSD1 and LSD2, two related mammalian H3K4me1/2 demethylases, are essential in maintaining global DNA methylation or establishing maternal DNA genomic imprints, respectively.
    • 36  Ciccone DN, Su H, Hevi S et al.: KDM1B is a histone H3K4 demethylase required to establish maternal genomic imprints. Nature461,415–418 (2009).▪ Along with [35], demonstrates that LSD1 and LSD2, two related mammalian H3K4me1/2 demethylases, are essential in maintaining global DNA methylation or establishing maternal DNA genomic imprints, respectively.
    • 37  Kato Y, Kaneda M, Hata K et al.: Role of the Dnmt3 family in de novo methylation of imprinted and repetitive sequences during male germ cell development in the mouse. Hum. Mol. Genet.16,2272–2280 (2007).
    • 38  Jeong S, Liang G, Sharma S et al.: Selective anchoring of DNA methyltransferases 3A and 3B to nucleosomes containing methylated DNA. Mol. Cell Biol.29,5366–5376 (2009).▪ Revealed that almost all of the cellular DNMT3a and -3b was associated with nucleosomes, primarily associated with methylated short interspersed repetitive nuclear elements and long interspersed repetitive nuclear elements as well as some single-copy methylated CpG islands.
    • 39  Zhang Y, Jurkowska R, Soeroes S et al.: Chromatin methylation activity of Dnmt3a and Dnmt3a/3L is guided by interaction of the ADD domain with the histone H3 tail. Nucleic Acids Res.38(13),4246–4253 (2010).
    • 40  Otani J, Nankumo T, Arita K, Inamoto S, Ariyoshi M, Shirakawa M: Structural basis for recognition of H3K4 methylation status by the DNA methyltransferase 3A ATRX-DNMT3-DNMT3L domain. EMBO Rep.10,1235–1241 (2009).
    • 41  Argentaro A, Yang JC, Chapman L et al.: Structural consequences of disease-causing mutations in the ATRX-DNMT3-DNMT3L (ADD) domain of the chromatin-associated protein ATRX. Proc. Natl Acad. Sci. USA104,11939–11944 (2007).
    • 42  Bostick M, Kim JK, Esteve PO, Clark A, Pradhan S, Jacobsen SE: UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science317,1760–1764 (2007).
    • 43  Sharif J, Muto M, Takebayashi S et al.: The SRA protein Np95 mediates epigenetic inheritance by recruiting Dnmt1 to methylated DNA. Nature450,908–912 (2007).
    • 44  Hashimoto H, Horton JR, Zhang X, Cheng X: UHRF1, a modular multi-domain protein, regulates replication-coupled crosstalk between DNA methylation and histone modifications. Epigenetics4,8–14 (2009).
    • 45  Unoki M, Brunet J, Mousli M: Drug discovery targeting epigenetic codes: the great potential of UHRF1, which links DNA methylation and histone modifications, as a drug target in cancers and toxoplasmosis. Biochem. Pharmacol.78,1279–1288 (2009).
    • 46  Citterio E, Papait R, Nicassio F et al.: Np95 is a histone-binding protein endowed with ubiquitin ligase activity. Mol. Cell Biol.24,2526–2535 (2004).
    • 47  Karagianni P, Amazit L, Qin J, Wong, J: ICBP90, a novel methyl K9 H3 binding protein linking protein ubiquitination with heterochromatin formation. Mol. Cell Biol.28,705–717 (2008).
    • 48  Lan F, Collins RE, De Cegli R et al.: Recognition of unmethylated histone H3 lysine 4 links BHC80 to LSD1-mediated gene repression. Nature448,718–722 (2007).
    • 49  Iwase S, Januma A, Miyamoto K et al.: Characterization of BHC80 in BRAF–HDAC complex, involved in neuron-specific gene repression. Biochem. Biophys. Res. Commun.322,601–608 (2004).
    • 50  Org T, Chignola F, Hetényi C et al.: The autoimmune regulator PHD finger binds to non-methylated histone H3K4 to activate gene expression. EMBO Rep.9,370–376 (2008).
    • 51  Chignola F, Gaetani M, Rebane A et al.: The solution structure of the first PHD finger of autoimmune regulator in complex with non-modified histone H3 tail reveals the antagonistic role of H3R2 methylation. Nucleic Acids Res.37,2951–2961 (2009).
    • 52  Helbling Chadwick L, Chadwick BP, Jaye DL, Wade PA: The Mi-2/NuRD complex associates with pericentromeric heterochromatin during S phase in rapidly proliferating lymphoid cells. Chromosoma118,445–457 (2009).
    • 53  Zegerman P, Canas B, Pappin D, Kouzarides T: Histone H3 lysine 4 methylation disrupts binding of nucleosome remodeling and deacetylase (NuRD) repressor complex. J. Biol. Chem.277,11621–11624 (2002).
    • 54  Nishioka K, Chuikov S, Sarma K et al.: Set9, a novel histone H3 methyltransferase that facilitates transcription by precluding histone tail modifications required for heterochromatin formation. Genes Dev.16,479–489 (2002).
    • 55  Musselman CA, Mansfield RE, Garske AL et al.: Binding of the CHD4 PHD2 finger to histone H3 is modulated by covalent modifications. Biochem. J.423,179–187 (2009).
    • 56  Cosgrove MS, Patel A: Mixed lineage leukemia: a structure-function perspective of the MLL1 protein. FEBS J.277,1832–1842 (2010).
    • 57  Ansari KI, Mandal SS: Mixed lineage leukemia: roles in gene expression, hormone signaling and mRNA processing. FEBS J.277,1790–1804 (2010).
    • 58  Terranova R, Agherbi H, Boned A, Meresse S, Djabali M: Histone and DNA methylation defects at Hox genes in mice expressing a SET domain-truncated form of MLL. Proc. Natl Acad. Sci. USA103,6629–6634 (2006).
    • 59  Milne TA, Briggs SD, Brock HW et al.: MLL targets SET domain methyltransferase activity to Hox gene promoters. Mol. Cell10,1107–1117 (2002).
    • 60  Birke M, Schreiner S, Garcia-Cuellar MP, Mahr K, Titgemeyer F, Slany RK: The MT domain of the proto-oncoprotein MLL binds to CpG-containing DNA and discriminates against methylation. Nucleic Acids Res.30,958–965 (2002).
    • 61  Ayton PM, Chen EH, Cleary ML: Binding to nonmethylated CpG DNA is essential for target recognition, transactivation, and myeloid transformation by an MLL oncoprotein. Mol. Cell Biol.24,10470–10478 (2004).
    • 62  Allen MD, Grummitt CG, Hilcenko C et al.: Solution structure of the nonmethyl-CpG-binding CXXC domain of the leukaemia-associated MLL histone methyltransferase. EMBO J.25,4503–4512 (2006).
    • 63  Cierpicki T, Risner LE, Grembecka J et al.: Structure of the MLL CXXC domain–DNA complex and its functional role in MLL-AF9 leukemia. Nat. Struct. Mol. Biol.17,62–68 (2010).
    • 64  Lee JH, Skalnik DG: CpG-binding protein (CXXC finger protein 1) is a component of the mammalian Set1 histone H3–Lys4 methyltransferase complex, the analogue of the yeast Set1/COMPASS complex. J. Biol. Chem.280,41725–41731 (2005).
    • 65  Lee JH, Tate CM, You JS, Skalnik DG: Identification and characterization of the human Set1B histone H3–Lys4 methyltransferase complex. J. Biol. Chem.282,13419–13428 (2007).
    • 66  Thomson JP, Skene PJ, Selfridge J et al.: CpG islands influence chromatin structure via the CpG-binding protein Cfp1. Nature464,1082–1086 (2010).▪▪ Suggests that unmethylated CpGs recruit Cfp1 and the associated methyltransferase Set1 creates new marks of H3K4me3 on the local chromatin.
    • 67  Tsukada Y, Fang J, Erdjument-Bromage H et al.: Histone demethylation by a family of JmjC domain-containing proteins. Nature439,811–816 (2006).
    • 68  Blackledge NP, Zhou JC, Tolstorukov MY, Farcas AM, Park PJ, Klose RJ: CpG islands recruit a histone H3 lysine 36 demethylase. Mol. Cell38,179–190 (2010).▪▪ Demonstrates that Jumonji-domain containing histone demethylase, a H3K36me2/1 demethylase, is recruited to the nonmethylated CpG island DNA on a genome-wide scale and that this is dependent on the intrinsic Cys–X–X–Cys domain.
    • 69  Jurkowska RZ, Anspach N, Urbanke C et al.: Formation of nucleoprotein filaments by mammalian DNA methyltransferase Dnmt3a in complex with regulator Dnmt3L. Nucleic Acids Res.36,6656–6663 (2008).
    • 70  Pradhan M, Esteve PO, Chin HG, Samaranayke M, Kim GD, Pradhan S: CXXC domain of human DNMT1 is essential for enzymatic activity. Biochemistry47,10000–10009 (2008).
    • 71  Jorgensen HF, Ben-Porath I, Bird AP: Mbd1 is recruited to both methylated and nonmethylated CpGs via distinct DNA binding domains. Mol. Cell Biol.24,3387–3395 (2004).
    • 72  Tahiliani M, Koh KP, Shen Y et al.: Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science324,930–935 (2009).▪▪ Among the first studies to demonstrate that 5-methylcytosine can be converted to hydroxymethylcytosine and explain the relevance of this modified base in the human genome.
    • 73  Ono R, Taki T, Taketani T, Taniwaki M, Kobayashi H, Hayashi Y: LCX, leukemia-associated protein with a CXXC domain, is fused to MLL in acute myeloid leukemia with trilineage dysplasia having t(10;11)(q22;q23). Cancer Res.62,4075–4080 (2002).
    • 74  Lorsbach RB, Moore J, Mathew S, Raimondi SC, Mukatira ST, Downing JR: TET1, a member of a novel protein family, is fused to MLL in acute myeloid leukemia containing the t(10;11)(q22;q23). Leukemia17,637–641 (2003).
    • 75  Dhasarathy A, Wade PA: The MBD protein family-reading an epigenetic mark? Mutat. Res.647,39–43 (2008).
    • 76  Hendrich B, Hardeland U, Ng HH, Jiricny J, Bird A: The thymine glycosylase MBD4 can bind to the product of deamination at methylated CpG sites. Nature401,301–304 (1999).
    • 77  Rai K, Huggins IJ, James SR, Karpf AR, Jones DA, Cairns BR: DNA demethylation in zebrafish involves the coupling of a deaminase, a glycosylase, and gadd45. Cell135,1201–1212 (2008).▪ Suggests that in zebrafish, methyl-CpG-binding domain 4 (containing both a N-terminal methyl-CpG-binding domain and a C-terminal thymine glycosylase domain) is involved in demethylating DNA.
    • 78  Popp C, Dean W, Feng S et al.: Genome-wide erasure of DNA methylation in mouse primordial germ cells is affected by AID deficiency. Nature463,1101–1105 (2010).▪▪ Together with [79], demonstrates that the cytidine deaminase AID has a role in DNA demethylation.
    • 79  Bhutani N, Brady JJ, Damian M, Sacco A, Corbel SY, Blau HM: Reprogramming towards pluripotency requires AID-dependent DNA demethylation. Nature463,1042–1047 (2010).▪▪ Together with [78], demonstrates that the cytidine deaminase AID has a role in DNA demethylation.
    • 80  Kim MS, Kondo T, Takada I et al.: DNA demethylation in hormone-induced transcriptional derepression. Nature461,1007–1012 (2009).
    • 81  Sarraf SA, Stancheva I: Methyl-CpG binding protein MBD1 couples histone H3 methylation at lysine 9 by SETDB1 to DNA replication and chromatin assembly. Mol. Cell15,595–605 (2004).
    • 82  Lyst MJ, Nan X, Stancheva I: Regulation of MBD1-mediated transcriptional repression by SUMO and PIAS proteins. EMBO J.25,5317–5328 (2006).
    • 83  Schultz DC, Ayyanathan K, Negorev D, Maul GG, Rauscher FJ 3rd: SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev.16,919–932 (2002).
    • 84  Rowe HM, Jakobsson J, Mesnard D et al.: KAP1 controls endogenous retroviruses in embryonic stem cells. Nature463,237–240 (2010).
    • 85  Fujita N, Watanabe S, Ichimura T et al.: Methyl-CpG binding domain 1 (MBD1) interacts with the Suv39h1-HP1 heterochromatic complex for DNA methylation-based transcriptional repression. J. Biol. Chem.278,24132–24138 (2003).
    • 86  Ohki I, Shimotake N, Fujita N et al.: Solution structure of the methyl-CpG binding domain of human MBD1 in complex with methylated DNA. Cell105,487–497 (2001).
    • 87  Ho KL, McNae IW, Schmiedeberg L, Klose RJ, Bird AP, Walkinshaw MD: MeCP2 binding to DNA depends upon hydration at methyl-CpG. Mol. Cell29,525–531 (2008).
    • 88  McGarvey KM, Fahrner JA, Greene E, Martens J, Jenuwein T, Baylin SB: Silenced tumor suppressor genes reactivated by DNA demethylation do not return to a fully euchromatic chromatin state. Cancer Res.66,3541–3549 (2006).
    • 89  Egger G, Aparicio AM, Escobar SG, Jones PA: Inhibition of histone deacetylation does not block resilencing of p16 after 5-aza-2’-deoxycytidine treatment. Cancer Res.67,346–353 (2007).
    • 90  Collins R, Cheng, X: A case study in cross-talk: the histone lysine methyltransferases G9a and GLP. Nucleic Acids Res.38,3503–3511 (2010).
    • 91  Clouaire T, de Las Heras JI, Merusi C, Stancheva I: Recruitment of MBD1 to target genes requires sequence-specific interaction of the MBD domain with methylated DNA. Nucleic Acids Res.38(14),4620–4634 (2010).
    • 92  Walker JR, Avvakumov GV, Xue S et al.: Crystal structure of the tandem Tudor domains of the E3 ubiquitin-protein ligase UHRF1 in complex with trimethylated histone H3-K9 peptide. doi:10.2210/pdb3db3/pdb. (2008) (Epub ahead of print).
    • 93  Rottach A, Frauer C, Pichler G, Bonapace IM, Spada F, Leonhardt H: The multi-domain protein Np95 connects DNA methylation and histone modification. Nucleic Acids Res.38,1796–1804 (2009).
    • 94  Hashimoto H, Horton JR, Zhang X, Bostick M, Jacobsen SE, Cheng X: The SRA domain of UHRF1 flips 5-methylcytosine out of the DNA helix. Nature455,826–829 (2008).
    • 95  Arita K, Ariyoshi M, Tochio H, Nakamura Y, Shirakawa M: Recognition of hemi-methylated DNA by the SRA protein UHRF1 by a base-flipping mechanism. Nature455,818–821 (2008).
    • 96  Avvakumov GV, Walker JR, Xue S et al.: Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1. Nature455,822–825 (2008).
    • 97  Tubbs JL, Latypov V, Kanugula S et al.: Flipping of alkylated DNA damage bridges base and nucleotide excision repair. Nature459,808–813 (2009).
    • 98  Kim JK, Esteve PO, Jacobsen SE, Pradhan S: UHRF1 binds G9a and participates in p21 transcriptional regulation in mammalian cells. Nucleic Acids Res.37,493–505 (2009).
    • 99  Achour M, Fuhrmann G, Alhosin M et al.: UHRF1 recruits the histone acetyltransferase Tip60 and controls its expression and activity. Biochem. Biophys. Res. Commun.390,523–528 (2009).
    • 100  Xu M, Long C, Chen X, Huang C, Chen S, Zhu B: Partitioning of histone H3-H4 tetramers during DNA replication-dependent chromatin assembly. Science328,94–98 (2010).▪▪ Demonstrates that histone H3.1–H4 tetramers remain intact and are partitioned evenly during DNA replication-dependent chromatin assembly.
    • 101  Collins RE, Northrop JP, Horton JR et al.: The ankyrin repeats of G9a and GLP histone methyltransferases are mono- and dimethyllysine binding modules. Nat. Struct. Mol. Biol.15,245–250 (2008).
    • 102  Grewal SI, Jia S: Heterochromatin revisited. Nat. Rev. Genet.8,35–46 (2007).
    • 103  Zhang K, Mosch K, Fischle W, Grewal SI: Roles of the Clr4 methyltransferase complex in nucleation, spreading and maintenance of heterochromatin. Nat. Struct. Mol. Biol.15,381–388 (2008).
    • 104  Peisajovich SG, Garbarino JE, Wei P, Lim WA: Rapid diversification of cell signaling phenotypes by modular domain recombination. Science328,368–372 (2010).