6,140
Views
46
CrossRef citations to date
0
Altmetric
Research Paper

ATRX binds to atypical chromatin domains at the 3′ exons of zinc finger genes to preserve H3K9me3 enrichment

, , , , , , , & show all
Pages 398-414 | Received 21 Oct 2015, Accepted 17 Mar 2016, Published online: 13 May 2016

References

  • Sinha M, Peterson CL. Chromatin dynamics during repair of chromosomal DNA double-strand breaks. Epigenomics 2009; 1:371–85; PMID:20495614; http://dx.doi.org/10.2217/epi.09.22
  • Hargreaves DC, Crabtree GR. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res 2011; 21:396–420; PMID:21358755; http://dx.doi.org/10.1038/cr.2011.32
  • Euskirchen G, Auerbach RK, Snyder M. SWI/SNF chromatin-remodeling factors: Multiscale analyses and diverse functions. J Biol Chem 2012; 287:30897–905; PMID:22952240; http://dx.doi.org/10.1074/jbc.R111.309302
  • Gospodinov A, Herceg Z. Chromatin structure in double strand break repair. DNA Repair (Amst) 2013; 12:800–10; PMID:23919923; http://dx.doi.org/10.1016/j.dnarep.2013.07.006
  • Längst G, Manelyte L. Chromatin Remodelers: From Function to Dysfunction. Genes (Basel) 2015; 6:299–324; PMID:26075616; http://dx.doi.org/10.3390/genes6020299
  • Clynes D, Gibbons RJ. ATRX and the replication of structured DNA. Curr Opin Genet Dev 2013; 23:289–94; PMID:23453691; http://dx.doi.org/10.1016/j.gde.2013.01.005
  • Voon HPJ, Wong LH. New players in heterochromatin silencing: histone variant H3.3 and the ATRX/DAXX chaperone. Nucleic Acids Res 2016; 44:1496–501:gkw012; PMID:26773061; http://dx.doi.org/10.1093/nar/gkw012
  • Ratnakumar K, Bernstein E. ATRX: the case of a peculiar chromatin remodeler. Epigenetics 2013; 8:3–9; PMID:23249563; http://dx.doi.org/10.4161/epi.23271
  • Watson LA, Goldberg H, Bérubé NG. Emerging roles of ATRX in cancer. Epigenomics 2015; 7:1365–78; PMID:26646632; http://dx.doi.org/10.2217/epi.15.82
  • Park DJ, Pask AJ, Huynh K, Renfree MB, Harley VR, Marshall Graves J A. Comparative analysis of ATRX, a chromatin remodeling protein. Gene 2004; 339:39–48; PMID:15363844; http://dx.doi.org/10.1016/j.gene.2004.06.025
  • Eustermann S, Yang J-C, Law MJ, Amos R, Chapman LM, Jelinska C, Garrick D, Clynes D, Gibbons RJ, Rhodes D, et al. Combinatorial readout of histone H3 modifications specifies localization of ATRX to heterochromatin. Nat Struct Mol Biol 2011; 18:777–82; PMID:21666677; http://dx.doi.org/10.1038/nsmb.2070
  • Iwase S, Xiang B, Ghosh S, Ren T, Lewis PW, Cochrane JC, Allis CD, Picketts DJ, Patel DJ, Li H, et al. ATRX ADD domain links an atypical histone methylation recognition mechanism to human mental-retardation syndrome. Nat Struct Mol Biol 2011; 18:769–76; PMID:21666679; http://dx.doi.org/10.1038/nsmb.2062
  • Xue Y, Gibbons R, Yan Z, Yang D, McDowell TL, Sechi S, Qin J, Zhou S, Higgs D, Wang W. The ATRX syndrome protein forms a chromatin-remodeling complex with Daxx and localizes in promyelocytic leukemia nuclear bodies. Proc Natl Acad Sci USA 2003; 100:10635–40; PMID:12953102; http://dx.doi.org/10.1073/pnas.1937626100
  • McDowell TL, Gibbons RJ, Sutherland H, O'Rourke DM, Bickmore W A, Pombo A, Turley H, Gatter K, Picketts DJ, Buckle VJ, et al. Localization of a putative transcriptional regulator (ATRX) at pericentromeric heterochromatin and the short arms of acrocentric chromosomes. Proc Natl Acad Sci USA 1999; 96:13983–8; PMID:10570185; http://dx.doi.org/10.1073/pnas.96.24.13983
  • Law MJ, Lower KM, Voon HPJJ, Hughes JR, Garrick D, Viprakasit V, Mitson M, De Gobbi M, Marra M, Morris A, et al. ATR-X Syndrome Protein Targets Tandem Repeats and Influences Allele-Specific Expression in a Size-Dependent Manner. Cell 2010; 143:367–78; PMID:21029860; http://dx.doi.org/10.1016/j.cell.2010.09.023
  • Drané P, Ouararhni K, Depaux A, Shuaib M, Hamiche A. The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev 2010; 24:1253–65; PMID:Can't; http://dx.doi.org/10.1101/gad.566910
  • Goldberg AD, Banaszynski L A, Noh K-M, Lewis PW, Elsaesser SJ, Stadler S, Dewell S, Law M, Guo X, Li X, et al. Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 2010; 140:678–91; PMID:20211137; http://dx.doi.org/10.1016/j.cell.2010.01.003
  • Wong LH, McGhie JD, Sim M, Anderson M A, Ahn S, Hannan RD, George AJ, Morgan K A, Mann JR, Choo KHA. ATRX interacts with H3.3 in maintaining telomere structural integrity in pluripotent embryonic stem cells. Genome Res 2010; 20:351–60; PMID:20110566; http://dx.doi.org/10.1101/gr.101477.109
  • Lewis PW, Elsaesser SJ, Noh K-M, Stadler SC, Allis CD. Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc Natl Acad Sci USA 2010; 107:14075–80; PMID:20651253; http://dx.doi.org/10.1073/pnas.1008850107
  • Sadic D, Schmidt K, Groh S, Kondofersky I, Ellwart J, Fuchs C, Theis FJ, Schotta G. Atrx promotes heterochromatin formation at retrotransposons. EMBO Rep 2015; 16:836 -50; PMID:26012739; http://dx.doi.org/10.15252/embr.201439937
  • Elsässer SJ, Noh K-M, Diaz N, Allis CD, Banaszynski LA. Histone H3.3 is required for endogenous retroviral element silencing in embryonic stem cells. Nature 2015; 522:240–4; PMID:25938714; http://dx.doi.org/10.1038/nature14345
  • He Q, Kim H, Huang R, Lu W, Tang M, Shi F, Yang D, Zhang X, Huang J, Liu D, et al. The Daxx/Atrx Complex Protects Tandem Repetitive Elements during DNA Hypomethylation by Promoting H3K9 Trimethylation. Cell Stem Cell 2015; 17:273–86; PMID:26340527; http://dx.doi.org/10.1016/j.stem.2015.07.022
  • Bérubé NG, Smeenk CA, Picketts DJ. Cell cycle-dependent phosphorylation of the ATRX protein correlates with changes in nuclear matrix and chromatin association. Hum Mol Genet 2000; 9:539–47; PMID:10699177; http://dx.doi.org/10.1093/hmg/9.4.539
  • Lechner MS, Schultz DC, Negorev D, Maul GG, Rauscher FJ. The mammalian heterochromatin protein 1 binds diverse nuclear proteins through a common motif that targets the chromoshadow domain. Biochem Biophys Res Commun 2005; 331:929–37; PMID:15882967; http://dx.doi.org/10.1016/j.bbrc.2005.04.016
  • Voon HPJ, Hughes JR, Rode C, De La Rosa-Velázquez IA, Jenuwein T, Feil R, Higgs DR, Gibbons RJ. ATRX Plays a Key Role in Maintaining Silencing at Interstitial Heterochromatic Loci and Imprinted Genes. Cell Rep 2015; 11:405–18; PMID:25865896; http://dx.doi.org/10.1016/j.celrep.2015.03.036
  • Ray-Gallet D, Woolfe A, Vassias I, Pellentz C, Lacoste N, Puri A, Schultz DC, Pchelintsev NA. Adams PD, Jansen LET, et al. Dynamics of Histone H3 Deposition In Vivo Reveal a Nucleosome Gap-Filling Mechanism for H3.3 to Maintain Chromatin Integrity. Mol Cell 2011; 44:928–41; PMID:22195966; http://dx.doi.org/10.1016/j.molcel.2011.12.006
  • Schneiderman JI, Orsi GA, Hughes KT, Loppin B, Ahmad K. Nucleosome-depleted chromatin gaps recruit assembly factors for the H3.3 histone variant. Proc Natl Acad Sci USA 2012; 109:19721–6; PMID:23150573; http://dx.doi.org/10.1073/pnas.1206629109
  • Filipescu D, Szenker E, Almouzni G. Developmental roles of histone H3 variants and their chaperones. Trends Genet 2013; 29:630–40; PMID:23830582; http://dx.doi.org/10.1016/j.tig.2013.06.002
  • Ratnakumar K, Duarte LF, LeRoy G, Hasson D, Smeets D, Vardabasso C, Bönisch C, Zeng T, Xiang B, Zhang DY, et al. ATRX-mediated chromatin association of histone variant macroH2A1 regulates α-globin expression. Genes Dev 2012; 26:433–8; PMID:22391447; http://dx.doi.org/10.1101/gad.179416.111
  • Clynes D, Higgs DR, Gibbons RJ. The chromatin remodeller ATRX: a repeat offender in human disease. Trends Biochem Sci 2013; 38:461–6; PMID:23916100; http://dx.doi.org/10.1016/j.tibs.2013.06.011
  • Levy M a., Kernohan KD, Jiang Y, Bérubé NG. ATRX promotes gene expression by facilitating transcriptional elongation through guanine-rich coding regions. Hum Mol Genet 2015; 24:1824–35; PMID:25452430; http://dx.doi.org/10.1093/hmg/ddu596
  • Heaphy CM, de Wilde RF, Jiao Y, Klein AP, Edil BH, Shi C, Bettegowda C, Rodriguez FJ, Eberhart CG, Hebbar S, et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science 2011; 333:425; PMID:21719641; http://dx.doi.org/10.1126/science.1207313
  • Schwartzentruber J, Korshunov A, Liu X-Y, Jones DTW, Pfaff E, Jacob K, Sturm D, Fontebasso AM, Quang D-AK, Tönjes M, et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 2012; 484:130–130; PMID:22286061; http://dx.doi.org/10.1038/nature11026
  • Lovejoy CA, Li W, Reisenweber S, Thongthip S, Bruno J, de Lange T, De S, Petrini JHJ, Sung P A, Jasin M, et al. Loss of ATRX, genome instability, and an altered DNA damage response are hallmarks of the alternative lengthening of telomeres pathway. PLoS Genet 2012; 8:e1002772; PMID:22829774; http://dx.doi.org/10.1371/journal.pgen.1002772
  • Bower K, Napier CE, Cole SL, Dagg RA, Lau LMS, Duncan EL, Moy EL, Reddel RR. Loss of wild-type ATRX expression in somatic cell hybrids segregates with activation of Alternative Lengthening of Telomeres. PLoS One 2012; 7:e50062; PMID:23185534; http://dx.doi.org/10.1371/journal.pone.0050062
  • Clynes D, Jelinska C, Xella B, Ayyub H, Scott C, Mitson M, Taylor S, Higgs DR, Gibbons RJ. Suppression of the alternative lengthening of telomere pathway by the chromatin remodelling factor ATRX. Nat Commun 2015; 6:7538; PMID:26143912; http://dx.doi.org/10.1038/ncomms8538
  • Huh MS, Price O'Dea T, Ouazia D, McKay BC, Parise G, Parks RJ, Rudnicki MA, Picketts DJ. Compromised genomic integrity impedes muscle growth after Atrx inactivation. J Clin Invest 2012; 122:4412–23; PMID:23114596; http://dx.doi.org/10.1172/JCI63765
  • Watson LA, Solomon LA, Li JR, Jiang Y, Edwards M, Shin-Ya K, Beier F, Bérubé NG. Atrx deficiency induces telomere dysfunction, endocrine defects, and reduced life span. J Clin Invest 2013; 123:2049–63; PMID:23563309; http://dx.doi.org/10.1172/JCI65634
  • Leung JW-CC, Ghosal G, Wang W, Shen X, Wang J, Li L, Chen J. Alpha thalassemia/mental retardation syndrome X-linked gene product ATRX is required for proper replication restart and cellular resistance to replication stress. J Biol Chem 2013; 288:6342–50; PMID:23329831; http://dx.doi.org/10.1074/jbc.M112.411603
  • Clynes D, Jelinska C, Xella B, Ayyub H, Taylor S, Mitson M, Bachrati CZ, Higgs DR, Gibbons RJ. ATRX dysfunction induces replication defects in primary mouse cells. PLoS One 2014; 9:e92915; PMID:24651726; http://dx.doi.org/10.1371/journal.pone.0092915
  • Emerson RO, Thomas JH. Adaptive evolution in zinc finger transcription factors. PLoS Genet 2009; 5; PMID:19119423; http://dx.doi.org/10.1371/journal.pgen.1000325
  • Dhwani Tadepally H, Aubry M. Evolution of C2H2 Zinc-finger Gene Families in Mammals. In: Encyclopedia of Life Sciences. Chichester, UK: John Wiley & Sons, Ltd; 2010; 1–9; http://dx.doi.org/10.1002/9780470015902.a0021738
  • Nowick K, Fields C, Gernat T, Caetano-Anolles D, Kholina N, Stubbs L. Gain, loss and divergence in primate zinc-finger genes: a rich resource for evolution of gene regulatory differences between species. PLoS One 2011; 6:e21553; PMID:21738707; http://dx.doi.org/10.1371/journal.pone.0021553
  • Blahnik KR, Dou L, Echipare L, Iyengar S, O'Geen H, Sanchez E, Zhao Y, Marra MA, Hirst M, Costello JF, et al. Characterization of the contradictory chromatin signatures at the 3′ exons of zinc finger genes. PLoS One 2011; 6:e17121; PMID:21347206; http://dx.doi.org/10.1371/journal.pone.0017121
  • Dunham I, Kundaje A, Aldred SF, Collins PJ, Davis C A, Doyle F, Epstein CB, Frietze S, Harrow J, Kaul R, et al. An integrated encyclopedia of DNA elements in the human genome. Nature 2012; 489:57–74; PMID:22955616; http://dx.doi.org/10.1038/nature11247
  • Sarma K, Cifuentes-Rojas C, Ergun A, Del Rosario A, Jeon Y, White F, Sadreyev R, Lee JT. ATRX directs binding of PRC2 to Xist RNA and Polycomb targets. Cell 2014; 159:869–83; PMID:25417162; http://dx.doi.org/10.1016/j.cell.2014.10.019
  • Ramamoorthy M, Smith S. Loss of ATRX Suppresses Resolution of Telomere Cohesion to Control Recombination in ALT Cancer Cells. Cancer Cell 2015; 28:357–69; PMID:26373281; http://dx.doi.org/10.1016/j.ccell.2015.08.003
  • Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, Epstein CB, Zhang X, Wang L, Issner R, Coyne M, et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 2011; 473:43–9; PMID:21441907; http://dx.doi.org/10.1038/nature09906
  • Vogel MJ, Guelen L, De Wit E, Peric-Hupkes D, Lodén M, Talhout W, Feenstra M, Abbas B, Classen AK, Van Steensel B. Human heterochromatin proteins form large domains containing KRAB-ZNF genes. Genome Res 2006; 16:1493–504; PMID:17038565; http://dx.doi.org/10.1101/gr.5391806
  • Julienne H, Zoufir A, Audit B, Arneodo A. Human genome replication proceeds through four chromatin states. PLoS Comput Biol 2013; 9:e1003233; PMID:24130466; http://dx.doi.org/10.1371/journal.pcbi.1003233
  • Hoffman MM, Ernst J, Wilder SP, Kundaje A, Harris RS, Libbrecht M, Giardine B, Ellenbogen PM, Bilmes J A, Birney E, et al. Integrative annotation of chromatin elements from ENCODE data. Nucleic Acids Res 2013; 41:827–41; PMID:23221638; http://dx.doi.org/10.1093/nar/gks1284
  • Iyengar S, Farnham PJ. KAP1 protein: An enigmatic master regulator of the genome. J Biol Chem 2011; 286:26267–76; PMID:21652716; http://dx.doi.org/10.1074/jbc.R111.252569
  • Frietze S, O'Geen H, Blahnik KR, Jin VX, Farnham PJ. ZNF274 recruits the histone methyltransferase SETDB1 to the 3′ ends of ZNF genes. PLoS One 2010; 5:e15082; PMID:21170338; http://dx.doi.org/10.1371/journal.pone.0015082
  • Bérubé NG. ATRX in chromatin assembly and genome architecture during development and disease. Biochem Cell Biol 2011; 89:435–44; PMID:Can't; http://dx.doi.org/10.1139/o11-038
  • Najafabadi HS, Mnaimneh S, Schmitges FW, Garton M, Lam KN, Yang A, Albu M, Weirauch MT, Radovani E, Kim PM, et al. C2H2 zinc finger proteins greatly expand the human regulatory lexicon. Nat Biotechnol 2015; 33:555–62; PMID:25690854; http://dx.doi.org/10.1038/nbt.3128
  • Iyengar S, Ivanov A V, Jin VX, Rauscher FJ, Farnham PJ. Functional analysis of KAP1 genomic recruitment. Mol Cell Biol 2011; 31:1833–47; PMID:21343339; http://dx.doi.org/10.1128/MCB.01331-10
  • Aymard F, Bugler B, Schmidt CK, Guillou E, Caron P, Briois S, Iacovoni JS, Daburon V, Miller KM, Jackson SP, et al. Transcriptionally active chromatin recruits homologous recombination at DNA double-strand breaks. Nat Struct Mol Biol 2014; 21:366–74; PMID:24658350; http://dx.doi.org/10.1038/nsmb.2796
  • Pfister SX, Ahrabi S, Zalmas L-P, Sarkar S, Aymard F, Bachrati CZ, Helleday T, Legube G, La Thangue NB, Porter ACG, et al. SETD2-dependent histone H3K36 trimethylation is required for homologous recombination repair and genome stability. Cell Rep 2014; 7:2006–18; PMID:24931610; http://dx.doi.org/10.1016/j.celrep.2014.05.026
  • Murray JM, Stiff T, Jeggo P A. DNA double-strand break repair within heterochromatic regions. Biochem Soc Trans 2012; 40:173–8; PMID:22260685; http://dx.doi.org/10.1042/BST20110631
  • Kalousi A, Hoffbeck A-S, Selemenakis PN, Pinder J, Savage KI, Khanna KK, Brino L, Dellaire G, Gorgoulis VG, Soutoglou E. The Nuclear Oncogene SET Controls DNA Repair by KAP1 and HP1 Retention to Chromatin. Cell Rep 2015; 11:149–63; PMID:25818296; http://dx.doi.org/10.1016/j.celrep.2015.03.005
  • Napier CE, Huschtscha LI, Harvey A, Bower K, Noble JR, Hendrickson EA, Reddel RR. ATRX represses alternative lengthening of telomeres. Oncotarget 2015; 6:16543–58; PMID:26001292; http://dx.doi.org/10.18632/oncotarget.3846
  • Naftelberg S, Schor IE, Ast G, Kornblihtt AR. Regulation of Alternative Splicing Through Coupling with Transcription and Chromatin Structure. Annu Rev Biochem 2015; 84:165–98; PMID:26034889; http://dx.doi.org/10.1146/annurev-biochem-060614-034242
  • Hasson D, Panchenko T, Salimian KJ, Salman MU, Sekulic N, Alonso A, Warburton PE, Black BE. The octamer is the major form of CENP-A nucleosomes at human centromeres. Nat Struct Mol Biol 2013; 20:687–95; PMID:23644596; http://dx.doi.org/10.1038/nsmb.2562
  • Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 2009; 10:R25; PMID:19261174; http://dx.doi.org/10.1186/gb-2009-10-3-r25
  • Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum C, Myers RM, Brown M, Li W, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol 2008; 9:R137; PMID:18798982; http://dx.doi.org/10.1186/gb-2008-9-9-r137
  • Liang K, Keleş S. Normalization of ChIP-seq data with control. BMC Bioinformatics 2012; 13:199; PMID:22883957; http://dx.doi.org/10.1186/1471-2105-13-199
  • Zang C, Schones DE, Zeng C, Cui K, Zhao K, Peng W. A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 2009; 25:1952–8; PMID:19505939; http://dx.doi.org/10.1093/bioinformatics/btp340
  • Quinlan AR, Hall IM. BEDTools: A flexible suite of utilities for comparing genomic features. Bioinformatics 2010; 26:841–2; PMID:20110278; http://dx.doi.org/10.1093/bioinformatics/btq033
  • Ramírez F, Dündar F, Diehl S, Grüning B a., Manke T. DeepTools: A flexible platform for exploring deep-sequencing data. Nucleic Acids Res 2014; 42:187–91; PMID:24799436; http://dx.doi.org/10.1093/nar/gku365
  • Sandve GK, Gundersen S, Johansen M, Glad IK, Gunathasan K, Holden L, Holden M, Liestøl K, Nygård S, Nygaard V, et al. The Genomic HyperBrowser: an analysis web server for genome-scale data. Nucleic Acids Res 2013; 41:W133–41; PMID:23632163; http://dx.doi.org/10.1093/nar/gkt342
  • Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 2009; 4:44–57; PMID:19131956; http://dx.doi.org/10.1038/nprot.2008.211
  • Huppert JL, Balasubramanian S. Prevalence of quadruplexes in the human genome. Nucleic Acids Res 2005; 33:2908–16; PMID:15914667; http://dx.doi.org/10.1093/nar/gki609
  • Machanick P, Bailey TL. MEME-ChIP: Motif analysis of large DNA datasets. Bioinformatics 2011; 27:1696–7; PMID:21486936; http://dx.doi.org/10.1093/bioinformatics/btr189
  • Ran F, Hsu P, Wright J, Agarwala V. Genome engineering using the CRISPR-Cas9 system. Nat Protoc 2013; 8:2281–308; PMID:24157548; http://dx.doi.org/10.1038/nprot.2013.143
  • Singh NP, McCoy MT, Tice RR, Schneider EL. A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res 1988; 175:184–91; PMID:3345800; http://dx.doi.org/10.1016/0014-4827(88)90265-0
  • Vardabasso C, Gaspar-Maia A, Hasson D, Pünzeler S, Valle-Garcia D, Straub T, Keilhauer EC, Strub T, Dong J, Panda T, et al. Histone Variant H2A.Z.2 Mediates Proliferation and Drug Sensitivity of Malignant Melanoma. Mol Cell 2015; 75–88; PMID:26051178; http://dx.doi.org/10.1016/j.molcel.2015.05.009