2,756
Views
18
CrossRef citations to date
0
Altmetric
Research Paper

Dynamics of histone H3 acetylation in the nucleosome core during mouse pre-implantation development

, , &
Pages 553-562 | Received 03 Sep 2015, Accepted 27 Sep 2015, Published online: 27 Jun 2016

References

  • Kouzarides T. Chromatin modifications and their function. Cell 2007; 128:693-705; PMID:17320507; http://dx.doi.org/10.1016/j.cell.2007.02.005
  • Strahl BD, Allis CD. The language of covalent histone modifications. Nature 2000; 403:41-5; PMID:10638745; http://dx.doi.org/10.1038/47412
  • Vermeulen M, Mulder KW, Denissov S, Pijnappel WW, van Schaik FM, Varier RA, Baltissen MP, Stunnenberg HG, Mann M, Timmers HT. Selective anchoring of TFIID to nucleosomes by trimethylation of histone H3 lysine 4. Cell 2007; 131:58-69; PMID:17884155; http://dx.doi.org/10.1016/j.cell.2007.08.016
  • Kamieniarz K, Izzo A, Dundr M, Tropberger P, Ozretic L, Kirfel J, Scheer E, Tropel P, Wisniewski JR, Tora L, et al. A dual role of linker histone H1.4 Lys 34 acetylation in transcriptional activation. Genes Dev 2011; 26:797-802; http://dx.doi.org/10.1101/gad.182014.111
  • Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 1997; 389:251-60; PMID:9305837; http://dx.doi.org/10.1038/38444
  • Cosgrove MS, Boeke JD, Wolberger C. Regulated nucleosome mobility and the histone code. Nat Struct Mol Biol 2004; 11:1037-43; PMID:15523479; http://dx.doi.org/10.1038/nsmb851
  • Tropberger P, Schneider R. Scratching the (lateral) surface of chromatin regulation by histone modifications. Nat Struct Mol Biol 2013; 20:657-61; PMID:23739170; http://dx.doi.org/10.1038/nsmb.2581
  • Tessarz P, Santos-Rosa H, Robson SC, Sylvestersen KB, Nelson CJ, Nielsen ML, Kouzarides T. Glutamine methylation in histone H2A is an RNA-polymerase-I-dedicated modification. Nature 2014; 505:564-8; PMID:24352239; http://dx.doi.org/10.1038/nature12819
  • Di Cerbo V, Mohn F, Ryan DP, Montellier E, Kacem S, Tropberger P, Kallis E, Holzner M, Hoerner L, Feldmann A, et al. Acetylation of histone H3 at lysine 64 regulates nucleosome dynamics and facilitates transcription. Elife 2014; 3, e01632; PMID:24668167; http://dx.doi.org/10.7554/eLife.01632
  • Simon M, North JA, Shimko JC, Forties RA, Ferdinand MB, Manohar M, Zhang M, Fishel R, Ottesen JJ, Poirier MG. Histone fold modifications control nucleosome unwrapping and disassembly. Proc Natl Acad Sci U S A 2011; 108:12711-6; PMID:21768347; http://dx.doi.org/10.1073/pnas.1106264108
  • Tropberger P, Pott S, Keller C, Kamieniarz-Gdula K, Caron M, Richter F, Li G, Mittler G, Liu ET, Bühler M, et al. Regulation of transcription through acetylation of H3K122 on the lateral surface of the histone octamer. Cell 2013; 152:859-72; PMID:23415232; http://dx.doi.org/10.1016/j.cell.2013.01.032
  • Xu F, Zhang K, Grunstein M. Acetylation in histone H3 globular domain regulates gene expression in yeast. Cell 2005; 121:375-85; PMID:15882620; http://dx.doi.org/10.1016/j.cell.2005.03.011
  • Xie W, Song C, Young NL, Sperling AS, Xu F, Sridharan R, Conway AE, Garcia BA, Plath K, Clark AT, et al. Histone h3 lysine 56 acetylation is linked to the core transcriptional network in human embryonic stem cells. Mol Cell 2009; 33:417-27; PMID:19250903; http://dx.doi.org/10.1016/j.molcel.2009.02.004
  • Li Q, Zhou H, Wurtele H, Davies B, Horazdovsky B, Verreault A, Zhang Z. Acetylation of histone H3 lysine 56 regulates replication-coupled nucleosome assembly. Cell 2008; 134:244-55; PMID:18662540; http://dx.doi.org/10.1016/j.cell.2008.06.018
  • 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 2005; 436:294-8; PMID:16015338; http://dx.doi.org/10.1038/nature03714
  • Neumann H, Hancock SM, Buning R, Routh A, Chapman L, Somers J, Owen-Hughes T, van Noort J, Rhodes D, Chin JW. A method for genetically installing site-specific acetylation in recombinant histones defines the effects of H3 K56 acetylation. Mol Cell 2009; 36:153-63; PMID:19818718; http://dx.doi.org/10.1016/j.molcel.2009.07.027
  • North JA, Shimko JC, Javaid S, Mooney AM, Shoffner MA, Rose SD, Bundschuh R, Fishel R, Ottesen JJ, Poirier MG. Regulation of the nucleosome unwrapping rate controls DNA accessibility. Nucleic Acids Res 2012; 40:10215-27; PMID:22965129; http://dx.doi.org/10.1093/nar/gks747
  • Burton A, Torres-Padilla ME. Chromatin dynamics in the regulation of cell fate allocation during early embryogenesis. Nat Rev Mol Cell Biol 2014; 15:723-34; PMID:25303116; http://dx.doi.org/10.1038/nrm3885
  • Hemberger M, Dean W, Reik W. Epigenetic dynamics of stem cells and cell lineage commitment: digging Waddington's canal. Nat Rev Mol Cell Biol 2009; 10:526-37; PMID:19603040; http://dx.doi.org/10.1038/nrm2727
  • Adenot PG, Mercier Y, Renard JP, Thompson EM. Differential H4 acetylation of paternal and maternal chromatin precedes DNA replication and differential transcriptional activity in pronuclei of 1-cell mouse embryos. Development 1997; 124:4615-25; PMID:9409678
  • Bouniol-Baly C, Nguyen E, Besombes D, Debey P. Dynamic organization of DNA replication in one-cell mouse embryos: relationship to transcriptional activation. Exp Cell Res 1997; 236:201-11; PMID:9344600; http://dx.doi.org/10.1006/excr.1997.3708
  • Santenard A, Ziegler-Birling C, Koch M, Tora L, Bannister AJ, Torres-Padilla ME. Heterochromatin formation in the mouse embryo requires critical residues of the histone variant H3.3. Nat Cell Biol 2010; 12:853-62; PMID:20676102; http://dx.doi.org/10.1038/ncb2089
  • Boskovic A, Bender A, Gall L, Ziegler-Birling C, Beaujean N, Torres-Padilla ME. Analysis of active chromatin modifications in early mammalian embryos reveals uncoupling of H2A.Z acetylation and H3K36 trimethylation from embryonic genome activation. Epigenetics 2012; 7:747-57; PMID:22647320; http://dx.doi.org/10.4161/epi.20584
  • Daujat S, Weiss T, Mohn F, Lange UC, Ziegler-Birling C, Zeissler U, Lappe M, Schübeler D, Torres-Padilla ME, Schneider R. H3K64 trimethylation marks heterochromatin and is dynamically remodeled during developmental reprogramming. Nat Struct Mol Biol 2009; 16:777-81; PMID:19561610; http://dx.doi.org/10.1038/nsmb.1629
  • Kourmouli N, Jeppesen P, Mahadevhaiah S, Burgoyne P, Wu R, Gilbert DM, Bongiorni S, Prantera G, Fanti L, Pimpinelli S, et al. Heterochromatin and tri-methylated lysine 20 of histone H4 in animals. J Cell Sci 2004; 117:2491-501; PMID:15128874; http://dx.doi.org/10.1242/jcs.01238
  • Kwok RP, Liu XT, Smith GD. Distribution of co-activators CBP and p300 during mouse oocyte and embryo development. Mol Reprod Dev 2006; 73:885-94; PMID:16596650; http://dx.doi.org/10.1002/mrd.20440
  • Wen D, Banaszynski LA, Rosenwaks Z, Allis CD, Rafii S. H3.3 replacement facilitates epigenetic reprogramming of donor nuclei in somatic cell nuclear transfer embryos. Nucleus 2014; 5:369-75; PMID:25482190; http://dx.doi.org/10.4161/nucl.36231
  • Erhardt S, Su IH, Schneider R, Barton S, Bannister AJ, Perez-Burgos L, Jenuwein T, Kouzarides T, Tarakhovsky A, Surani MA. Consequences of the depletion of zygotic and embryonic enhancer of zeste 2 during preimplantation mouse development. Development 2003; 130:4235-48; PMID:12900441; http://dx.doi.org/10.1242/dev.00625
  • Santos F, Hendrich B, Reik W, Dean W. Dynamic reprogramming of DNA methylation in the early mouse embryo. Dev Biol 2002; 241:172-82; PMID:11784103; http://dx.doi.org/10.1006/dbio.2001.0501
  • Ooga M, Inoue A, Kageyama S, Akiyama T, Nagata M, Aoki F. Changes in H3K79 methylation during preimplantation development in mice. Biol Reprod 2008; 78:413-24; PMID:18003948; http://dx.doi.org/10.1095/biolreprod.107.063453
  • Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, Steine EJ, Hanna J, Lodato MA, Frampton GM, Sharp PA, et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci U S A 2010; 107:21931-6; PMID:21106759; http://dx.doi.org/10.1073/pnas.1016071107
  • Wiekowski M, Miranda M, DePamphilis ML. Regulation of gene expression in preimplantation mouse embryos: effects of the zygotic clock and the first mitosis on promoter and enhancer activities. Dev Biol 1991; 147:403-14; PMID:1916016; http://dx.doi.org/10.1016/0012-1606(91)90298-H
  • Bevilacqua A, Fiorenza MT, Mangia F. Developmental activation of an episomic hsp70 gene promoter in two-cell mouse embryos by transcription factor Sp1. Nucleic Acids Res 1997; 25:1333-8; PMID:9060426; http://dx.doi.org/10.1093/nar/25.7.1333
  • Latham KE, Solter D, Schultz RM. Acquisition of a transcriptionally permissive state during the 1-cell stage of mouse embryogenesis. Dev Biol 1992; 149:457-62; PMID:1309712; http://dx.doi.org/10.1016/0012-1606(92)90300-6
  • Schultz RM, Worrad DM. Role of chromatin structure in zygotic gene activation in the mammalian embryo. Semin Cell Biol 1995; 6:201-8; PMID:8562912; http://dx.doi.org/10.1006/scel.1995.0028
  • Boskovic A, Eid A, Pontabry J, Ishiuchi T, Spiegelhalter C, Raghu Ram EV, Meshorer E, Torres-Padilla ME. Higher chromatin mobility supports totipotency and precedes pluripotency in vivo. Genes Dev 2014; 28:1042-7; PMID:24831699; http://dx.doi.org/10.1101/gad.238881.114
  • Bouniol C, Nguyen E, Debey P. Endogenous transcription occurs at the 1-cell stage in the mouse embryo. Exp Cell Res 1995; 218:57-62; PMID:7537698; http://dx.doi.org/10.1006/excr.1995.1130
  • Adenot PG, Campion E, Legouy E, Allis CD, Dimitrov S, Renard J, Thompson EM. Somatic linker histone H1 is present throughout mouse embryogenesis and is not replaced by variant H1 degrees. J Cell Sci 2000; 113 (Pt 16):2897-907; PMID:10910774
  • Wiekowski M, Miranda M, DePamphilis ML. Requirements for promoter activity in mouse oocytes and embryos distinguish paternal pronuclei from maternal and zygotic nuclei. Dev Biol 1993; 159:366-78; PMID:8365573; http://dx.doi.org/10.1006/dbio.1993.1248
  • Brehove M, Wang T, North J, Luo Y, Dreher SJ, Shimko JC, Ottesen JJ, Luger K, Poirier MG. Histone Core Phosphorylation Regulates DNA Accessibility. J Biol Chem 2015; 290(37):22612-21; PMID:26175159; http://dx.doi.org/10.1074/jbc.M115.661363
  • Manohar M, Mooney AM, North JA, Nakkula RJ, Picking JW, Edon A, Fishel R, Poirier MG, Ottesen JJ. Acetylation of histone H3 at the nucleosome dyad alters DNA-histone binding. J Biol Chem 2009; 284:23312-21; PMID:19520870; http://dx.doi.org/10.1074/jbc.M109.003202
  • Hogan BL, Beddington R, Costantini F, Lacy E. Manipulating the Mouse Embryo. 2nd. edn, (Cold Spring Harbor Laboratory Press, 1994).
  • Torres-Padilla ME, Bannister AJ, Hurd PJ, Kouzarides T, Zernicka-Goetz M. Dynamic distribution of the replacement histone variant H3.3 in the mouse oocyte and preimplantation embryos. Int J Dev Biol 2006; 50:455-61; PMID:16586346; http://dx.doi.org/10.1387/ijdb.052073mt