1,820
Views
18
CrossRef citations to date
0
Altmetric
Research Paper

Essential roles of HDAC1 and 2 in lineage development and genome-wide DNA methylation during mouse preimplantation development

ORCID Icon, , ORCID Icon, , , ORCID Icon, , , , ORCID Icon & ORCID Icon show all
Pages 369-385 | Received 27 Jul 2019, Accepted 12 Sep 2019, Published online: 24 Sep 2019

References

  • Eckersley-Maslin MA, Alda-Catalinas C, Reik W. Dynamics of the epigenetic landscape during the maternal-to-zygotic transition. Nat Rev Mol Cell Biol. 2018;19(7):436–450.
  • Schultz RM, Stein P, Svoboda P. The oocyte-to-embryo transition in mouse: past, present, and future. Biol Reprod. 2018;99(1):160–174.
  • Matoba S, Zhang Y. Somatic cell nuclear transfer reprogramming: mechanisms and applications. Cell Stem Cell. 2018;23(4):471–485.
  • Chen Z, Hagen DE, Elsik CG, et al. Characterization of global loss of imprinting in fetal overgrowth syndrome induced by assisted reproduction. Proc Natl Acad Sci U S A. 2015;112(15):4618–4623.
  • Liu W, Liu X, Wang C, et al. Identification of key factors conquering developmental arrest of somatic cell cloned embryos by combining embryo biopsy and single-cell sequencing. Cell Discov. 2016;2:16010.
  • Matoba S, Liu Y, Lu F, et al. Embryonic development following somatic cell nuclear transfer impeded by persisting histone methylation. Cell. 2014;159(4):884–895.
  • Gao R, Wang C, Gao Y, et al. Inhibition of aberrant DNA Re-methylation improves post-implantation development of somatic cell nuclear transfer embryos. Cell Stem Cell. 2018;23(3):426–435 e425.
  • Chung YG, Matoba S, Liu Y, et al. Histone demethylase expression enhances human somatic cell nuclear transfer efficiency and promotes derivation of pluripotent stem cells. Cell Stem Cell. 2015;17(6):758–766.
  • Zhou C, Wang Y, Zhang J, et al. H3K27me3 is an epigenetic barrier while KDM6A overexpression improves nuclear reprogramming efficiency. Faseb J. 2019;33(3):4638–4652.
  • Liu X, Wang Y, Gao Y, et al. H3K9 demethylase KDM4E is an epigenetic regulator for bovine embryonic development and a defective factor for nuclear reprogramming. Development. 2018;145(4):dev155317.
  • Liu Z, Cai Y, Wang Y, et al. Cloning of macaque monkeys by somatic cell nuclear transfer. Cell. 2018;174(1):245.
  • Xue Y, Wong J, Moreno GT, et al. NURD, a novel complex with both ATP-dependent chromatin-remodeling and histone deacetylase activities. Mol Cell. 1998;2(6):851–861.
  • Hassig CA, Fleischer TC, Billin AN, et al. Histone deacetylase activity is required for full transcriptional repression by mSin3A. Cell. 1997;89(3):341–347.
  • You A, Tong JK, Grozinger CM, et al. CoREST is an integral component of the CoREST-human histone deacetylase complex. Proc Natl Acad Sci U S A. 2001;98(4):1454–1458.
  • Ma P, Schultz RM. HDAC1 and HDAC2 in mouse oocytes and preimplantation embryos: specificity versus compensation. Cell Death Differ. 2016;23(7):1119–1127.
  • Sheikh BN, Akhtar A. The many lives of KATs – detectors, integrators and modulators of the cellular environment. Nat Rev Genet. 2019;20(1):7–23.
  • Robinson PJ, An W, Routh A, et al. 30 nm chromatin fibre decompaction requires both H4-K16 acetylation and linker histone eviction. J Mol Biol. 2008;381(4):816–825.
  • Kidder BL, Palmer S. HDAC1 regulates pluripotency and lineage specific transcriptional networks in embryonic and trophoblast stem cells. Nucleic Acids Res. 2012;40(7):2925–2939.
  • Wang Z, Zang C, Cui K, et al. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell. 2009;138(5):1019–1031.
  • Yamaguchi T, Cubizolles F, Zhang Y, et al. Histone deacetylases 1 and 2 act in concert to promote the G1-to-S progression. Gene Dev. 2010;24(5):455–469.
  • LeBoeuf M, Terrell A, Trivedi S, et al. Hdac1 and Hdac2 Act redundantly to control p63 and p53 functions in epidermal progenitor cells. Dev Cell. 2010;19(6):807–818.
  • Ma PP, Pan H, Montgomery RL, et al. Compensatory functions of histone deacetylase 1 (HDAC1) and HDAC2 regulate transcription and apoptosis during mouse oocyte development. Proc Natl Acad Sci U S A. 2012;109(8):E481–E489.
  • Montgomery RL, Davis CA, Potthoff MJ, et al. Histone deacetylases 1 and 2 redundantly regulate cardiac morphogenesis, growth, and contractility. Gene Dev. 2007;21(14):1790–1802.
  • Guan JS, Haggarty SJ, Giacometti E, et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature. 2009;459(7243):55–U58.
  • Lagger G, O’Carroll D, Rembold M, et al. Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression. Embo J. 2002;21(11):2672–2681.
  • Ma PP, Schultz RM. Histone deacetylase 1 (HDAC1) regulates histone acetylation, development, and gene expression in preimplantation mouse embryos. Dev Biol. 2008;319(1):110–120.
  • Ito A, Kawaguchi Y, Lai C-H, et al. MDM2-HDAC1-mediated deacetylation of p53 is required for its degradation. Embo J. 2002;21(22):6236–6245.
  • Cao ZB, Carey TS, Ganguly A, et al. Transcription factor AP-2 gamma induces early Cdx2 expression and represses HIPPO signaling to specify the trophectoderm lineage. Development. 2015;142(9):1606–1615.
  • Tatsuta T, Mukaisho K-I, Sugihara H, et al. Expression of Cdx2 in early GRCL of Barrett’s esophagus induced in rats by duodenal reflux. Digest Dis Sci. 2005;50(3):425–431.
  • Suzuki S, Nozawa Y, Tsukamoto S, et al. CHD1 acts via the Hmgpi pathway to regulate mouse early embryogenesis. Development. 2015;142(13):2375–2384.
  • Zhao J, Ohsumi TK, Kung JT, et al. Genome-wide identification of polycomb-associated RNAs by RIP-seq. Mol Cell. 2010;40(6):939–953.
  • Wang JL, Rao S, Chu J, et al. A protein interaction network for pluripotency of embryonic stem cells. Nature. 2006;444(7117):364–368.
  • Pengpeng MA, Schultz RM. Histone deacetylase 1 (HDAC1) regulates histone acetylation, development, and gene expression in preimplantation mouse embryos. Dev Biol. 2008;319(2):485–486.
  • Xue ZG, Huang K, Cai C, et al. Genetic programs in human and mouse early embryos revealed by single-cell RNA sequencing. Nature. 2013;500(7464):593.
  • Furumai R, Matsuyama A, Kobashi N, et al. FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Res. 2002;62(17):4916–4921.
  • Zhang K, Dai XP, Wallingford MC, et al. Depletion of Suds3 reveals an essential role in early lineage specification. Dev Biol. 2013;373(2):359–372.
  • Zenker J, White MD, Templin RM, et al. A microtubule-organizing center directing intracellular transport in the early mouse embryo. Science. 2017;357(6354):925.
  • Rossant J. Genetic control of early cell lineages in the mammalian embryo. Annu Rev Genet. 2018;52:185–201.
  • Nishioka N, Inoue K-I, Adachi K, et al. The hippo signaling pathway components lats and yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell. 2009;16(3):398–410.
  • Robertson KD, Ait-Si-Ali S, Yokochi T, et al. DNMT1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F-responsive promoters. Nat Genet. 2000;25(3):338–342.
  • Rountree MR, Bachman KE, Baylin SB. DNMT1 binds HDAC2 and a new co-repressor, DMAP1, to form a complex at replication foci. Nat Genet. 2000;25(3):269–277.
  • Fuks F, Burgers WA, Godin N, et al. Dnmt3a binds deacetylases and is recruited by a sequence-specific repressor to silence transcription. Embo J. 2001;20(10):2536–2544.
  • Ma P, de Waal E, Weaver JR, et al. A DNMT3A2-HDAC2 complex is essential for genomic imprinting and genome integrity in mouse oocytes. Cell Rep. 2015;13(8):1552–1560.
  • Bostick M, Kim JK, Estève P-O, et al. UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science. 2007;317(5845):1760–1764.
  • Yang WM, Inouye C, Zeng YY, et al. Transcriptional repression by YY1 is mediated by interaction with a mammalian homolog of the yeast global regulator RPD3. Proc Natl Acad Sci U S A. 1996;93(23):12845–12850.
  • Magnaghi-Jaulin L, Groisman R, Naguibneva I, et al. Retinoblastoma protein represses transcription by recruiting a histone deacetylase. Nature. 1998;391(6667):601–605.
  • Juan LJ, Shia WJ, Chen MH, et al. Histone deacetylases specifically down-regulate p53-dependent gene activation. J Biol Chem. 2000;275(27):20436–20443.
  • Narita T, Weinert BT, Choudhary C. Functions and mechanisms of non-histone protein acetylation. Nat Rev Mol Cell Bio. 2019;20(3):156–174.
  • Kruiswijk F, Labuschagne CF, Vousden KH. p53 in survival, death and metabolic health: a lifeguard with a licence to kill. Nat Rev Mol Cell Bio. 2015;16(7):393–405.
  • Jamaladdin S, Kelly RDW, O’Regan L, et al. Histone deacetylase (HDAC) 1 and 2 are essential for accurate cell division and the pluripotency of embryonic stem cells. Proc Natl Acad Sci U S A. 2014;111(27):9840–9845.
  • Wilting RH, Yanover E, Heideman MR, et al. Overlapping functions of Hdac1 and Hdac2 in cell cycle regulation and haematopoiesis. Embo J. 2010;29(15):2586–2597.
  • Hainer SJ, Boskovic A, McCannell KN, et al. Profiling of pluripotency factors in single cells and early embryos. Cell. 2019;177(5):1319-1329.
  • Yagi R, Kohn MJ, Karavanova I, et al. Transcription factor TEAD4 specifies the trophectoderm lineage at the beginning of mammalian development. Development. 2007;134(21):3827–3836.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.