Skip to main content
Protein & Cell logoLink to Protein & Cell
. 2012 May 2;3(4):278–290. doi: 10.1007/s13238-012-2916-6

Epigenetic control on cell fate choice in neural stem cells

Xiao-Ling Hu 1, Yuping Wang 2, Qin Shen 1,
PMCID: PMC4729703  PMID: 22549586

Abstract

Derived from neural stem cells (NSCs) and progenitor cells originated from the neuroectoderm, the nervous system presents an unprecedented degree of cellular diversity, interwoven to ensure correct connections for propagating information and responding to environmental cues. NSCs and progenitor cells must integrate cell-intrinsic programs and environmental cues to achieve production of appropriate types of neurons and glia at appropriate times and places during development. These developmental dynamics are reflected in changes in gene expression, which is regulated by transcription factors and at the epigenetic level. From early commitment of neural lineage to functional plasticity in terminal differentiated neurons, epigenetic regulation is involved in every step of neural development. Here we focus on the recent advance in our understanding of epigenetic regulation on orderly generation of diverse neural cell types in the mammalian nervous system, an important aspect of neural development and regenerative medicine.

Keywords: neural stem cells (NSCs), epigenetic regulation, neurogenesis, gliogenesis, radial glial cell, cerebral cortex, subventricular zone (SVZ), DNA methylation, histone modification

References

  1. Allis C.D., Berger S.L., Cote J., Dent S., Jenuwien T., Kouzarides T., Pillus L., Reinberg D., Shi Y., Shiekhattar R., et al. New nomenclature for chromatin-modifying enzymes. Cell. 2007;131:633–636. doi: 10.1016/j.cell.2007.10.039. [DOI] [PubMed] [Google Scholar]
  2. Alvarez-Venegas R., Avramova Z. Methylation patterns of histone H3 Lys 4, Lys 9 and Lys 27 in transcriptionally active and inactive Arabidopsis genes and in atx1 mutants. Nucleic Acids Res. 2005;33:5199–5207. doi: 10.1093/nar/gki830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ambros V. MicroRNA pathways in flies and worms: growth, death, fat, stress, and timing. Cell. 2003;113:673–676. doi: 10.1016/s0092-8674(03)00428-8. [DOI] [PubMed] [Google Scholar]
  4. Ambros V. The functions of animal microRNAs. Nature. 2004;431:350–355. doi: 10.1038/nature02871. [DOI] [PubMed] [Google Scholar]
  5. Azuara V., Perry P., Sauer S., Spivakov M., Jørgensen H.F., John R.M., Gouti M., Casanova M., Warnes G., Merkenschlager M., et al. Chromatin signatures of pluripotent cell lines. Nat Cell Biol. 2006;8:532–538. doi: 10.1038/ncb1403. [DOI] [PubMed] [Google Scholar]
  6. Balazs R., Vernon J., Hardy J. Epigenetic mechanisms in Alzheimer’s disease: progress but much to do. Neurobiol Aging. 2011;32:1181–1187. doi: 10.1016/j.neurobiolaging.2011.02.024. [DOI] [PubMed] [Google Scholar]
  7. Bannister A.J., Kouzarides T. Reversing histone methylation. Nature. 2005;436:1103–1106. doi: 10.1038/nature04048. [DOI] [PubMed] [Google Scholar]
  8. Bannister A.J., Schneider R., Myers F.A., Thorne A.W., Crane-Robinson C., Kouzarides T. Spatial distribution of di- and tri-methyl lysine 36 of histone H3 at active genes. J Biol Chem. 2005;280:17732–17736. doi: 10.1074/jbc.M500796200. [DOI] [PubMed] [Google Scholar]
  9. Bartel D.P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
  10. Bartel D.P. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bernstein B.E., Meissner A., Lander E.S. The mammalian epigenome. Cell. 2007;128:669–681. doi: 10.1016/j.cell.2007.01.033. [DOI] [PubMed] [Google Scholar]
  12. Bernstein B.E., Mikkelsen T.S., Xie X., Kamal M., Huebert D.J., Cuff J., Fry B., Meissner A., Wernig M., Plath K., et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 2006;125:315–326. doi: 10.1016/j.cell.2006.02.041. [DOI] [PubMed] [Google Scholar]
  13. Bernstein E., Allis C.D. RNA meets chromatin. Genes Dev. 2005;19:1635–1655. doi: 10.1101/gad.1324305. [DOI] [PubMed] [Google Scholar]
  14. Bhutani N., Brady J.J., Damian M., Sacco A., Corbel S.Y., Blau H.M. Reprogramming towards pluripotency requires AID-dependent DNA demethylation. Nature. 2010;463:1042–1047. doi: 10.1038/nature08752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bhutani N., Burns D.M., Blau H.M. DNA demethylation dynamics. Cell. 2011;146:866–872. doi: 10.1016/j.cell.2011.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002;16:6–21. doi: 10.1101/gad.947102. [DOI] [PubMed] [Google Scholar]
  17. Bird A. Perceptions of epigenetics. Nature. 2007;447:396–398. doi: 10.1038/nature05913. [DOI] [PubMed] [Google Scholar]
  18. Burgold T., Spreafico F., De Santa F., Totaro M.G., Prosperini E., Natoli G., Testa G. The histone H3 lysine 27-specific demethylase Jmjd3 is required for neural commitment. PLoS One. 2008;3:e3034. doi: 10.1371/journal.pone.0003034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Cao R., Wang L., Wang H., Xia L., Erdjument-Bromage H., Tempst P., Jones R.S., Zhang Y. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science. 2002;298:1039–1043. doi: 10.1126/science.1076997. [DOI] [PubMed] [Google Scholar]
  20. Cao X., Yeo G., Muotri A.R., Kuwabara T., Gage F.H. Noncoding RNAs in the mammalian central nervous system. Annu Rev Neurosci. 2006;29:77–103. doi: 10.1146/annurev.neuro.29.051605.112839. [DOI] [PubMed] [Google Scholar]
  21. Cartagena J.A., Matsunaga S., Seki M., Kurihara D., Yokoyama M., Shinozaki K., Fujimoto S., Azumi Y., Uchiyama S., Fukui K. The Arabidopsis SDG4 contributes to the regulation of pollen tube growth by methylation of histone H3 lysines 4 and 36 in mature pollen. Dev Biol. 2008;315:355–368. doi: 10.1016/j.ydbio.2007.12.016. [DOI] [PubMed] [Google Scholar]
  22. Cheng L.C., Pastrana E., Tavazoie M., Doetsch F. miR-124 regulates adult neurogenesis in the subventricular zone stem cell niche. Nat Neurosci. 2009;12:399–408. doi: 10.1038/nn.2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Ciccone D.N., Su H., Hevi S., Gay F., Lei H., Bajko J., Xu G., Li E., Chen T. KDM1B is a histone H3K4 demethylase required to establish maternal genomic imprints. Nature. 2009;461:415–418. doi: 10.1038/nature08315. [DOI] [PubMed] [Google Scholar]
  24. Cloos P.A., Christensen J., Agger K., Maiolica A., Rappsilber J., Antal T., Hansen K.H., Helin K. The putative oncogene GASC1 demethylates tri- and dimethylated lysine 9 on histone H3. Nature. 2006;442:307–311. doi: 10.1038/nature04837. [DOI] [PubMed] [Google Scholar]
  25. Conaco C., Otto S., Han J.J., Mandel G. Reciprocal actions of REST and a microRNA promote neuronal identity. Proc Natl Acad Sci U S A. 2006;103:2422–2427. doi: 10.1073/pnas.0511041103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Coolen M., Bally-Cuif L. MicroRNAs in brain development and physiology. Curr Opin Neurobiol. 2009;19:461–470. doi: 10.1016/j.conb.2009.09.006. [DOI] [PubMed] [Google Scholar]
  27. Cortellino S., Xu J., Sannai M., Moore R., Caretti E., Cigliano A., Le Coz M., Devarajan K., Wessels A., Soprano D., et al. Thymine DNA glycosylase is essential for active DNA demethylation by linked deamination-base excision repair. Cell. 2011;146:67–79. doi: 10.1016/j.cell.2011.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. De Carvalho D.D., You J.S., Jones P.A. DNA methylation and cellular reprogramming. Trends Cell Biol. 2010;20:609–617. doi: 10.1016/j.tcb.2010.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. de la Serna I.L., Ohkawa Y., Imbalzano A.N. Chromatin remodelling in mammalian differentiation: lessons from ATP-dependent remodellers. Nat Rev Genet. 2006;7:461–473. doi: 10.1038/nrg1882. [DOI] [PubMed] [Google Scholar]
  30. De Pietri Tonelli D., Pulvers J.N., Haffner C., Murchison E.P., Hannon G.J., Huttner W.B. miRNAs are essential for survival and differentiation of newborn neurons but not for expansion of neural progenitors during early neurogenesis in the mouse embryonic neocortex. Development. 2008;135:3911–3921. doi: 10.1242/dev.025080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Deaton A.M., Bird A. CpG islands and the regulation of transcription. Genes Dev. 2011;25:1010–1022. doi: 10.1101/gad.2037511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Eden S., Hashimshony T., Keshet I., Cedar H., Thorne A.W. DNA methylation models histone acetylation. Nature. 1998;394:842. doi: 10.1038/29680. [DOI] [PubMed] [Google Scholar]
  33. Edmunds J.W., Mahadevan L.C., Clayton A.L. Dynamic histone H3 methylation during gene induction: HYPB/Setd2 mediates all H3K36 trimethylation. EMBO J. 2008;27:406–420. doi: 10.1038/sj.emboj.7601967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Eiraku M., Watanabe K., Matsuo-Takasaki M., Kawada M., Yonemura S., Matsumura M., Wataya T., Nishiyama A., Muguruma K., Sasai Y. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals. Cell Stem Cell. 2008;3:519–532. doi: 10.1016/j.stem.2008.09.002. [DOI] [PubMed] [Google Scholar]
  35. Fan G., Martinowich K., Chin M.H., He F., Fouse S.D., Hutnick L., Hattori D., Ge W., Shen Y., Wu H., et al. DNA methylation controls the timing of astrogliogenesis through regulation of JAK-STAT signaling. Development. 2005;132:3345–3356. doi: 10.1242/dev.01912. [DOI] [PubMed] [Google Scholar]
  36. Feng J., Chang H., Li E., Fan G. Dynamic expression of de novo DNA methyltransferases Dnmt3a and Dnmt3b in the central nervous system. J Neurosci Res. 2005;79:734–746. doi: 10.1002/jnr.20404. [DOI] [PubMed] [Google Scholar]
  37. Ferrón S.R., Charalambous M., Radford E., McEwen K., Wildner H., Hind E., Morante-Redolat J.M., Laborda J., Guillemot F., Bauer S.R., et al. Postnatal loss of Dlk1 imprinting in stem cells and niche astrocytes regulates neurogenesis. Nature. 2011;475:381–385. doi: 10.1038/nature10229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Ficz G., Branco M.R., Seisenberger S., Santos F., Krueger F., Hore T.A., Marques C.J., Andrews S., Reik W. Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature. 2011;473:398–402. doi: 10.1038/nature10008. [DOI] [PubMed] [Google Scholar]
  39. Gaspard N., Bouschet T., Hourez R., Dimidschstein J., Naeije G., van den Ameele J., Espuny-Camacho I., Herpoel A., Passante L., Schiffmann S.N., et al. An intrinsic mechanism of corticogenesis from embryonic stem cells. Nature. 2008;455:351–357. doi: 10.1038/nature07287. [DOI] [PubMed] [Google Scholar]
  40. Gatto S., Della Ragione F., Cimmino A., Strazzullo M., Fabbri M., Mutarelli M., Ferraro L., Weisz A., D’Esposito M., Matarazzo M.R. Epigenetic alteration of microRNAs in DNMT3B-mutated patients of ICF syndrome. Epigenetics. 2010;5:427–443. doi: 10.4161/epi.5.5.11999. [DOI] [PubMed] [Google Scholar]
  41. Giraldez A.J., Cinalli R.M., Glasner M.E., Enright A.J., Thomson J.M., Baskerville S., Hammond S.M., Bartel D.P., Schier A.F. MicroRNAs regulate brain morphogenesis in zebrafish. Science. 2005;308:833–838. doi: 10.1126/science.1109020. [DOI] [PubMed] [Google Scholar]
  42. Gjerset R.A., Martin D.W., Jr. Presence of a DNA demethylating activity in the nucleus of murine erythroleukemic cells. J Biol Chem. 1982;257:8581–8583. [PubMed] [Google Scholar]
  43. Goldberg A.D., Allis C.D., Bernstein E. Epigenetics: a landscape takes shape. Cell. 2007;128:635–638. doi: 10.1016/j.cell.2007.02.006. [DOI] [PubMed] [Google Scholar]
  44. Golebiewska A., Atkinson S.P., Lako M., Armstrong L. Epigenetic landscaping during hESC differentiation to neural cells. Stem Cells. 2009;27:1298–1308. doi: 10.1002/stem.59. [DOI] [PubMed] [Google Scholar]
  45. Goll M.G., Bestor T.H. Eukaryotic cytosine methyltransferases. Annu Rev Biochem. 2005;74:481–514. doi: 10.1146/annurev.biochem.74.010904.153721. [DOI] [PubMed] [Google Scholar]
  46. Goll M.G., Halpern M.E. DNA methylation in zebrafish. Prog Mol Biol Transl Sci. 2011;101:193–218. doi: 10.1016/B978-0-12-387685-0.00005-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Griffiths-Jones S., Saini H.K., van Dongen S., Enright A.J. miRBase: tools for microRNA genomics. Nucleic Acids Res. 2008;36:D154–D158. doi: 10.1093/nar/gkm952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Grimson A., Farh K.K., Johnston W.K., Garrett-Engele P., Lim L.P., Bartel D.P. MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell. 2007;27:91–105. doi: 10.1016/j.molcel.2007.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Groth A., Rocha W., Verreault A., Almouzni G. Chromatin challenges during DNA replication and repair. Cell. 2007;128:721–733. doi: 10.1016/j.cell.2007.01.030. [DOI] [PubMed] [Google Scholar]
  50. Guo J.U., Su Y., Zhong C., Ming G.L., Song H. Hydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brain. Cell. 2011;145:423–434. doi: 10.1016/j.cell.2011.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Habibi E., Masoudi-Nejad A., Abdolmaleky H.M., Haggarty S.J. Emerging roles of epigenetic mechanisms in Parkinson’s disease. Funct Integr Genomics. 2011;11:523–537. doi: 10.1007/s10142-011-0246-z. [DOI] [PubMed] [Google Scholar]
  52. Hargreaves D.C., Crabtree G.R. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res. 2011;21:396–420. doi: 10.1038/cr.2011.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Hashimshony T., Zhang J., Keshet I., Bustin M., Cedar H. The role of DNA methylation in setting up chromatin structure during development. Nat Genet. 2003;34:187–192. doi: 10.1038/ng1158. [DOI] [PubMed] [Google Scholar]
  54. He L., Hannon G.J. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5:522–531. doi: 10.1038/nrg1379. [DOI] [PubMed] [Google Scholar]
  55. He X.B., Yi S.H., Rhee Y.H., Kim H., Han Y.M., Lee S.H., Lee H., Park C.H., Lee Y.S., Richardson E., et al. Prolonged membrane depolarization enhances midbrain dopamine neuron differentiation via epigenetic histone modifications. Stem Cells. 2011;29:1861–1873. doi: 10.1002/stem.739. [DOI] [PubMed] [Google Scholar]
  56. He Y.F., Li B.Z., Li Z., Liu P., Wang Y., Tang Q., Ding J., Jia Y., Chen Z., Li L., et al. Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science. 2011;333:1303–1307. doi: 10.1126/science.1210944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hevner R.F., Daza R.A., Rubenstein J.L., Stunnenberg H., Olavarria J.F., Englund C. Beyond laminar fate: toward a molecular classification of cortical projection/pyramidal neurons. Dev Neurosci. 2003;25:139–151. doi: 10.1159/000072263. [DOI] [PubMed] [Google Scholar]
  58. Hitoshi S., Alexson T., Tropepe V., Donoviel D., Elia A.J., Nye J.S., Conlon R.A., Mak T.W., Bernstein A., van der Kooy D. Notch pathway molecules are essential for the maintenance, but not the generation, of mammalian neural stem cells. Genes Dev. 2002;16:846–858. doi: 10.1101/gad.975202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hitoshi S., Ishino Y., Kumar A., Jasmine S., Tanaka K.F., Kondo T., Kato S., Hosoya T., Hotta Y., Ikenaka K. Mammalian Gcm genes induce Hes5 expression by active DNA demethylation and induce neural stem cells. Nat Neurosci. 2011;14:957–964. doi: 10.1038/nn.2875. [DOI] [PubMed] [Google Scholar]
  60. Hitoshi S., Seaberg R.M., Koscik C., Alexson T., Kusunoki S., Kanazawa I., Tsuji S., van der Kooy D. Primitive neural stem cells from the mammalian epiblast differentiate to definitive neural stem cells under the control of Notch signaling. Genes Dev. 2004;18:1806–1811. doi: 10.1101/gad.1208404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Holliday R. Epigenetics: a historical overview. Epigenetics. 2006;1:76–80. doi: 10.4161/epi.1.2.2762. [DOI] [PubMed] [Google Scholar]
  62. Hsieh J., Nakashima K., Kuwabara T., Mejia E., Gage F.H. Histone deacetylase inhibition-mediated neuronal differentiation of multipotent adult neural progenitor cells. Proc Natl Acad Sci U S A. 2004;101:16659–16664. doi: 10.1073/pnas.0407643101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Huarte M., Lan F., Kim T., Vaughn M.W., Zaratiegui M., Martienssen R.A., Buratowski S., Shi Y. The fission yeast Jmj2 reverses histone H3 Lysine 4 trimethylation. J Biol Chem. 2007;282:21662–21670. doi: 10.1074/jbc.M703897200. [DOI] [PubMed] [Google Scholar]
  64. Iorio M.V., Piovan C., Croce C.M. Interplay between microRNAs and the epigenetic machinery: an intricate network. Biochim Biophys Acta. 2010;1799:694–701. doi: 10.1016/j.bbagrm.2010.05.005. [DOI] [PubMed] [Google Scholar]
  65. Irmady K., Zechel S., Unsicker K. Fibroblast growth factor 2 regulates astrocyte differentiation in a region-specific manner in the hindbrain. Glia. 2011;59:708–719. doi: 10.1002/glia.21141. [DOI] [PubMed] [Google Scholar]
  66. Iwase S., Lan F., Bayliss P., de la Torre-Ubieta L., Huarte M., Qi H.H., Whetstine J.R., Bonni A., Roberts T.M., Shi Y. The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases. Cell. 2007;128:1077–1088. doi: 10.1016/j.cell.2007.02.017. [DOI] [PubMed] [Google Scholar]
  67. Jacobson M. Developmental neurobiology. 3rd edn. New York: Plenum Press; 1991. [Google Scholar]
  68. Jaenisch R., Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33:245–254. doi: 10.1038/ng1089. [DOI] [PubMed] [Google Scholar]
  69. Jamniczky H.A., Boughner J.C., Rolian C., Gonzalez P.N., Powell C.D., Schmidt E.J., Parsons T.E., Bookstein F.L., Hallgrímsson B. Rediscovering Waddington in the post-genomic age: Operationalising Waddington’s epigenetics reveals new ways to investigate the generation and modulation of phenotypic variation. Bioessays. 2010;32:553–558. doi: 10.1002/bies.200900189. [DOI] [PubMed] [Google Scholar]
  70. Jiang H., Shukla A., Wang X., Chen W.Y., Bernstein B.E., Roeder R.G. Role for Dpy-30 in ES cell-fate specification by regulation of H3K4 methylation within bivalent domains. Cell. 2011;144:513–525. doi: 10.1016/j.cell.2011.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Joglekar M.V., Joglekar V.M., Hardikar A.A. Expression of islet-specific microRNAs during human pancreatic development. Gene Expr Patterns. 2009;9:109–113. doi: 10.1016/j.gep.2008.10.001. [DOI] [PubMed] [Google Scholar]
  72. Juliandi B., Abematsu M., Nakashima K. Chromatin remodeling in neural stem cell differentiation. Curr Opin Neurobiol. 2010;20:408–415. doi: 10.1016/j.conb.2010.04.001. [DOI] [PubMed] [Google Scholar]
  73. Juliandi B., Abematsu M., Nakashima K. Epigenetic regulation in neural stem cell differentiation. Dev Growth Differ. 2010;52:493–504. doi: 10.1111/j.1440-169X.2010.01175.x. [DOI] [PubMed] [Google Scholar]
  74. Kapoor A., Agius F., Zhu J.K. Preventing transcriptional gene silencing by active DNA demethylation. FEBS Lett. 2005;579:5889–5898. doi: 10.1016/j.febslet.2005.08.039. [DOI] [PubMed] [Google Scholar]
  75. Kapsimali M., Kloosterman W.P., de Bruijn E., Rosa F., Plasterk R.H., Wilson S.W. MicroRNAs show a wide diversity of expression profiles in the developing and mature central nervous system. Genome Biol. 2007;8:R173. doi: 10.1186/gb-2007-8-8-r173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Kawaguchi A., Miyata T., Sawamoto K., Takashita N., Murayama A., Akamatsu W., Ogawa M., Okabe M., Tano Y., Goldman S.A., et al. Nestin-EGFP transgenic mice: visualization of the self-renewal and multipotency of CNS stem cells. Mol Cell Neurosci. 2001;17:259–273. doi: 10.1006/mcne.2000.0925. [DOI] [PubMed] [Google Scholar]
  77. Kawase-Koga Y., Low R., Otaegi G., Pollock A., Deng H., Eisenhaber F., Maurer-Stroh S., Sun T. RNAase-III enzyme Dicer maintains signaling pathways for differentiation and survival in mouse cortical neural stem cells. J Cell Sci. 2010;123:586–594. doi: 10.1242/jcs.059659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Kennison J.A. The Polycomb and trithorax group proteins of Drosophila: trans-regulators of homeotic gene function. Annu Rev Genet. 1995;29:289–303. doi: 10.1146/annurev.ge.29.120195.001445. [DOI] [PubMed] [Google Scholar]
  79. Kohyama J., Kojima T., Takatsuka E., Yamashita T., Namiki J., Hsieh J., Gage F.H., Namihira M., Okano H., Sawamoto K., et al. Epigenetic regulation of neural cell differentiation plasticity in the adult mammalian brain. Proc Natl Acad Sci U S A. 2008;105:18012–18017. doi: 10.1073/pnas.0808417105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Kornberg R.D. Chromatin structure: a repeating unit of histones and DNA. Science. 1974;184:868–871. doi: 10.1126/science.184.4139.868. [DOI] [PubMed] [Google Scholar]
  81. Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705. doi: 10.1016/j.cell.2007.02.005. [DOI] [PubMed] [Google Scholar]
  82. Ku M., Koche R.P., Rheinbay E., Mendenhall E.M., Endoh M., Mikkelsen T.S., Presser A., Nusbaum C., Xie X., Chi A.S., et al. Genomewide analysis of PRC1 and PRC2 occupancy identifies two classes of bivalent domains. PLoS Genet. 2008;4:e1000242. doi: 10.1371/journal.pgen.1000242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Kuo M.H., Allis C.D. Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays. 1998;20:615–626. doi: 10.1002/(SICI)1521-1878(199808)20:8<615::AID-BIES4>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
  84. Lan F., Bayliss P.E., Rinn J.L., Whetstine J.R., Wang J.K., Chen S., Iwase S., Alpatov R., Issaeva I., Canaani E., et al. A histone H3 lysine 27 demethylase regulates animal posterior development. Nature. 2007;449:689–694. doi: 10.1038/nature06192. [DOI] [PubMed] [Google Scholar]
  85. Lee M.G., Norman J., Shilatifard A., Shiekhattar R. Physical and functional association of a trimethyl H3K4 demethylase and Ring6a/MBLR, a polycomb-like protein. Cell. 2007;128:877–887. doi: 10.1016/j.cell.2007.02.004. [DOI] [PubMed] [Google Scholar]
  86. Lee M.G., Villa R., Trojer P., Norman J., Yan K.P., Reinberg D., Di Croce L., Shiekhattar R. Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science. 2007;318:447–450. doi: 10.1126/science.1149042. [DOI] [PubMed] [Google Scholar]
  87. Lee Y.S., Nakahara K., Pham J.W., Kim K., He Z., Sontheimer E.J., Carthew R.W. Distinct roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing pathways. Cell. 2004;117:69–81. doi: 10.1016/s0092-8674(04)00261-2. [DOI] [PubMed] [Google Scholar]
  88. Lessard J., Wu J.I., Ranish J.A., Wan M., Winslow M.M., Staahl B.T., Wu H., Aebersold R., Graef I.A., Crabtree G.R. An essential switch in subunit composition of a chromatin remodeling complex during neural development. Neuron. 2007;55:201–215. doi: 10.1016/j.neuron.2007.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Leucht C., Stigloher C., Wizenmann A., Klafke R., Folchert A., Bally-Cuif L. MicroRNA-9 directs late organizer activity of the midbrain-hindbrain boundary. Nat Neurosci. 2008;11:641–648. doi: 10.1038/nn.2115. [DOI] [PubMed] [Google Scholar]
  90. Li E., Bestor T.H., Jaenisch R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell. 1992;69:915–926. doi: 10.1016/0092-8674(92)90611-f. [DOI] [PubMed] [Google Scholar]
  91. Li J.Y., Pu M.T., Hirasawa R., Li B.Z., Huang Y.N., Zeng R., Jing N.H., Chen T., Li E., Sasaki H., et al. Synergistic function of DNA methyltransferases Dnmt3a and Dnmt3b in the methylation of Oct4 and Nanog. Mol Cell Biol. 2007;27:8748–8759. doi: 10.1128/MCB.01380-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Li X., Barkho B.Z., Luo Y., Smrt R.D., Santistevan N.J., Liu C., Kuwabara T., Gage F.H., Zhao X. Epigenetic regulation of the stem cell mitogen Fgf-2 by Mbd1 in adult neural stem/progenitor cells. J Biol Chem. 2008;283:27644–27652. doi: 10.1074/jbc.M804899200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Liu C., Teng Z.Q., Santistevan N.J., Szulwach K.E., Guo W., Jin P., Zhao X. Epigenetic regulation of miR-184 by MBD1 governs neural stem cell proliferation and differentiation. Cell Stem Cell. 2010;6:433–444. doi: 10.1016/j.stem.2010.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Lunyak V.V., Rosenfeld M.G. No rest for REST: REST/NRSF regulation of neurogenesis. Cell. 2005;121:499–501. doi: 10.1016/j.cell.2005.05.003. [DOI] [PubMed] [Google Scholar]
  95. Lyle R., Watanabe D., te Vruchte D., Lerchner W., Smrzka O.W., Wutz A., Schageman J., Hahner L., Davies C., Barlow D.P. The imprinted antisense RNA at the Igf2r locus overlaps but does not imprint Mas1. Nat Genet. 2000;25:19–21. doi: 10.1038/75546. [DOI] [PubMed] [Google Scholar]
  96. Ma D.K., Jang M.H., Guo J.U., Kitabatake Y., Chang M.L., Pow-Anpongkul N., Flavell R.A., Lu B., Ming G.L., Song H. Neuronal activity-induced Gadd45b promotes epigenetic DNA demethylation and adult neurogenesis. Science. 2009;323:1074–1077. doi: 10.1126/science.1166859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Ma D.K., Marchetto M.C., Guo J.U., Ming G.L., Gage F.H., Song H. Epigenetic choreographers of neurogenesis in the adult mammalian brain. Nat Neurosci. 2010;13:1338–1344. doi: 10.1038/nn.2672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Martens J.A., Winston F. Recent advances in understanding chromatin remodeling by Swi/Snf complexes. Curr Opin Genet Dev. 2003;13:136–142. doi: 10.1016/s0959-437x(03)00022-4. [DOI] [PubMed] [Google Scholar]
  99. Mastroeni D., Grover A., Delvaux E., Whiteside C., Coleman P.D., Rogers J. Epigenetic mechanisms in Alzheimer’s disease. Neurobiol Aging. 2011;32:1161–1180. doi: 10.1016/j.neurobiolaging.2010.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Matsumoto S., Banine F., Struve J., Xing R., Adams C., Liu Y., Metzger D., Chambon P., Rao M.S., Sherman L.S. Brg1 is required for murine neural stem cell maintenance and gliogenesis. Dev Biol. 2006;289:372–383. doi: 10.1016/j.ydbio.2005.10.044. [DOI] [PubMed] [Google Scholar]
  101. Mattick J.S., Makunin I.V. Small regulatory RNAs in mammals. Hum Mol Genet. 2005;14:R121–R132. doi: 10.1093/hmg/ddi101. [DOI] [PubMed] [Google Scholar]
  102. Mattick J.S., Makunin I.V. Non-coding RNA. Hum Mol Genet. 2006;15:R17–R29. doi: 10.1093/hmg/ddl046. [DOI] [PubMed] [Google Scholar]
  103. Mehler M.F. Epigenetic principles and mechanisms underlying nervous system functions in health and disease. Prog Neurobiol. 2008;86:305–341. doi: 10.1016/j.pneurobio.2008.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Mehler M.F., Mattick J.S. Non-coding RNAs in the nervous system. J Physiol. 2006;575:333–341. doi: 10.1113/jphysiol.2006.113191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Mehler M.F., Mattick J.S. Noncoding RNAs and RNA editing in brain development, functional diversification, and neurological disease. Physiol Rev. 2007;87:799–823. doi: 10.1152/physrev.00036.2006. [DOI] [PubMed] [Google Scholar]
  106. Meissner A., Mikkelsen T.S., Gu H., Wernig M., Hanna J., Sivachenko A., Zhang X., Bernstein B.E., Nusbaum C., Jaffe D.B., et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature. 2008;454:766–770. doi: 10.1038/nature07107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Métivier R., Gallais R., Tiffoche C., Le Péron C., Jurkowska R.Z., Carmouche R.P., Ibberson D., Barath P., Demay F., Reid G., et al. Cyclical DNA methylation of a transcriptionally active promoter. Nature. 2008;452:45–50. doi: 10.1038/nature06544. [DOI] [PubMed] [Google Scholar]
  108. Moazed D. Small RNAs in transcriptional gene silencing and genome defence. Nature. 2009;457:413–420. doi: 10.1038/nature07756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Mohn F., Weber M., Rebhan M., Roloff T.C., Richter J., Stadler M.B., Bibel M., Schübeler D. Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol Cell. 2008;30:755–766. doi: 10.1016/j.molcel.2008.05.007. [DOI] [PubMed] [Google Scholar]
  110. Montgomery R.L., Hsieh J., Barbosa A.C., Richardson J.A., Olson E.N. Histone deacetylases 1 and 2 control the progression of neural precursors to neurons during brain development. Proc Natl Acad Sci U S A. 2009;106:7876–7881. doi: 10.1073/pnas.0902750106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Morange M. The relations between genetics and epigenetics: a historical point of view. Ann N Y Acad Sci. 2002;981:50–60. doi: 10.1111/j.1749-6632.2002.tb04911.x. [DOI] [PubMed] [Google Scholar]
  112. Mosammaparast N., Shi Y. Reversal of histone methylation: biochemical and molecular mechanisms of histone demethylases. Annu Rev Biochem. 2010;79:155–179. doi: 10.1146/annurev.biochem.78.070907.103946. [DOI] [PubMed] [Google Scholar]
  113. Nakamura Y., Sakakibara S., Miyata T., Ogawa M., Shimazaki T., Weiss S., Kageyama R., Okano H. The bHLH gene hes1 as a repressor of the neuronal commitment of CNS stem cells. J Neurosci. 2000;20:283–293. doi: 10.1523/JNEUROSCI.20-01-00283.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Namihira M., Kohyama J., Abematsu M., Nakashima K. Epigenetic mechanisms regulating fate specification of neural stem cells. Philos Trans R Soc Lond B Biol Sci. 2008;363:2099–2109. doi: 10.1098/rstb.2008.2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Nekrasov M., Wild B., Müller J. Nucleosome binding and histone methyltransferase activity of Drosophila PRC2. EMBO Rep. 2005;6:348–353. doi: 10.1038/sj.embor.7400376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Nguyen S., Meletis K., Fu D., Jhaveri S., Jaenisch R. Ablation of de novo DNA methyltransferase Dnmt3a in the nervous system leads to neuromuscular defects and shortened lifespan. Dev Dyn. 2007;236:1663–1676. doi: 10.1002/dvdy.21176. [DOI] [PubMed] [Google Scholar]
  117. Niehrs C. Active DNA demethylation and DNA repair. Differentiation. 2009;77:1–11. doi: 10.1016/j.diff.2008.09.004. [DOI] [PubMed] [Google Scholar]
  118. Okano M., Bell D.W., Haber D.A., Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99:247–257. doi: 10.1016/s0092-8674(00)81656-6. [DOI] [PubMed] [Google Scholar]
  119. Ooi S.K., Bestor T.H. The colorful history of active DNA demethylation. Cell. 2008;133:1145–1148. doi: 10.1016/j.cell.2008.06.009. [DOI] [PubMed] [Google Scholar]
  120. Ooi S.K., Bestor T.H. Cytosine methylation: remaining faithful. Curr Biol. 2008;18:R174–R176. doi: 10.1016/j.cub.2007.12.045. [DOI] [PubMed] [Google Scholar]
  121. Pastor W.A., Pape U.J., Huang Y., Henderson H.R., Lister R., Ko M., McLoughlin E.M., Brudno Y., Mahapatra S., Kapranov P., et al. Genome-wide mapping of 5-hydroxymethylcytosine in embryonic stem cells. Nature. 2011;473:394–397. doi: 10.1038/nature10102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Pazin M.J., Kadonaga J.T. What’s up and down with histone deacetylation and transcription? Cell. 1997;89:325–328. doi: 10.1016/s0092-8674(00)80211-1. [DOI] [PubMed] [Google Scholar]
  123. Qian X., Goderie S.K., Shen Q., Stern J.H., Temple S. Intrinsic programs of patterned cell lineages in isolated vertebrate CNS ventricular zone cells. Development. 1998;125:3143–3152. doi: 10.1242/dev.125.16.3143. [DOI] [PubMed] [Google Scholar]
  124. Qian X., Shen Q., Goderie S.K., He W., Capela A., Davis A.A., Temple S. Timing of CNS cell generation: a programmed sequence of neuron and glial cell production from isolated murine cortical stem cells. Neuron. 2000;28:69–80. doi: 10.1016/s0896-6273(00)00086-6. [DOI] [PubMed] [Google Scholar]
  125. Rai K., Huggins I.J., James S.R., Karpf A.R., Jones D.A., Cairns B.R. DNA demethylation in zebrafish involves the coupling of a deaminase, a glycosylase, and gadd45. Cell. 2008;135:1201–1212. doi: 10.1016/j.cell.2008.11.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Randazzo F.M., Khavari P., Crabtree G., Tamkun J., Rossant J. brg1: a putative murine homologue of the Drosophila brahma gene, a homeotic gene regulator. Dev Biol. 1994;161:229–242. doi: 10.1006/dbio.1994.1023. [DOI] [PubMed] [Google Scholar]
  127. Ravin R., Hoeppner D.J., Munno D.M., Carmel L., Sullivan J., Levitt D.L., Miller J.L., Athaide C., Panchision D.M., McKay R.D. Potency and fate specification in CNS stem cell populations in vitro. Cell Stem Cell. 2008;3:670–680. doi: 10.1016/j.stem.2008.09.012. [DOI] [PubMed] [Google Scholar]
  128. Reynolds B.A., Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 1992;255:1707–1710. doi: 10.1126/science.1553558. [DOI] [PubMed] [Google Scholar]
  129. Ringrose L., Paro R. Epigenetic regulation of cellular memory by the Polycomb and Trithorax group proteins. Annu Rev Genet. 2004;38:413–443. doi: 10.1146/annurev.genet.38.072902.091907. [DOI] [PubMed] [Google Scholar]
  130. Robertson K.D., Ait-Si-Ali S., Yokochi T., Wade P.A., Jones P.L., Wolffe A.P. DNMT1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F-responsive promoters. Nat Genet. 2000;25:338–342. doi: 10.1038/77124. [DOI] [PubMed] [Google Scholar]
  131. Robertson K.D., Wolffe A.P. DNA methylation in health and disease. Nat Rev Genet. 2000;1:11–19. doi: 10.1038/35049533. [DOI] [PubMed] [Google Scholar]
  132. Roth S.Y., Allis C.D. Histone acetylation and chromatin assembly: a single escort, multiple dances? Cell. 1996;87:5–8. doi: 10.1016/s0092-8674(00)81316-1. [DOI] [PubMed] [Google Scholar]
  133. Rougeulle C., Chaumeil J., Sarma K., Allis C.D., Reinberg D., Avner P., Heard E. Differential histone H3 Lys-9 and Lys-27 methylation profiles on the X chromosome. Mol Cell Biol. 2004;24:5475–5484. doi: 10.1128/MCB.24.12.5475-5484.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Sato S., Yagi S., Arai Y., Hirabayashi K., Hattori N., Iwatani M., Okita K., Ohgane J., Tanaka S., Wakayama T., et al. Genome-wide DNA methylation profile of tissue-dependent and differentially methylated regions (T-DMRs) residing in mouse pluripotent stem cells. Genes Cells. 2010;15:607–618. doi: 10.1111/j.1365-2443.2010.01404.x. [DOI] [PubMed] [Google Scholar]
  135. Schmitz S.U., Albert M., Malatesta M., Morey L., Johansen J.V., Bak M., Tommerup N., Abarrategui I., Helin K. Jarid1b targets genes regulating development and is involved in neural differentiation. EMBO J. 2011;30:4586–4600. doi: 10.1038/emboj.2011.383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Setoguchi H., Namihira M., Kohyama J., Asano H., Sanosaka T., Nakashima K. Methyl-CpG binding proteins are involved in restricting differentiation plasticity in neurons. J Neurosci Res. 2006;84:969–979. doi: 10.1002/jnr.21001. [DOI] [PubMed] [Google Scholar]
  137. Shen Q., Temple S. Fine control: microRNA regulation of adult neurogenesis. Nat Neurosci. 2009;12:369–370. doi: 10.1038/nn0409-369. [DOI] [PubMed] [Google Scholar]
  138. Shen Q., Wang Y., Dimos J.T., Fasano C.A., Phoenix T.N., Lemischka I.R., Ivanova N.B., Stifani S., Morrisey E.E., Temple S. The timing of cortical neurogenesis is encoded within lineages of individual progenitor cells. Nat Neurosci. 2006;9:743–751. doi: 10.1038/nn1694. [DOI] [PubMed] [Google Scholar]
  139. Shi Y., Lan F., Matson C., Mulligan P., Whetstine J.R., Cole P.A., Casero R.A., Shi Y. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 2004;119:941–953. doi: 10.1016/j.cell.2004.12.012. [DOI] [PubMed] [Google Scholar]
  140. Shibata M., Kurokawa D., Nakao H., Ohmura T., Aizawa S. MicroRNA-9 modulates Cajal-Retzius cell differentiation by suppressing Foxg1 expression in mouse medial pallium. J Neurosci. 2008;28:10415–10421. doi: 10.1523/JNEUROSCI.3219-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Smith C.L., Peterson C.L. A conserved Swi2/Snf2 ATPase motif couples ATP hydrolysis to chromatin remodeling. Mol Cell Biol. 2005;25:5880–5892. doi: 10.1128/MCB.25.14.5880-5892.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Smith J.L., Schoenwolf G.C. Neurulation: coming to closure. Trends Neurosci. 1997;20:510–517. doi: 10.1016/s0166-2236(97)01121-1. [DOI] [PubMed] [Google Scholar]
  143. So A.Y., Jung J.W., Lee S., Kim H.S., Kang K.S. DNA methyltransferase controls stem cell aging by regulating BMI1 and EZH2 through microRNAs. PLoS One. 2011;6:e19503. doi: 10.1371/journal.pone.0019503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Song C.X., Szulwach K.E., Fu Y., Dai Q., Yi C., Li X., Li Y., Chen C.H., Zhang W., Jian X., et al. Selective chemical labeling reveals the genome-wide distribution of 5-hydroxymethylcytosine. Nat Biotechnol. 2011;29:68–72. doi: 10.1038/nbt.1732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Song M.R., Ghosh A. FGF2-induced chromatin remodeling regulates CNTF-mediated gene expression and astrocyte differentiation. Nat Neurosci. 2004;7:229–235. doi: 10.1038/nn1192. [DOI] [PubMed] [Google Scholar]
  146. Sterner D.E., Berger S.L. Acetylation of histones and transcription-related factors. Microbiol Mol Biol Rev. 2000;64:435–459. doi: 10.1128/mmbr.64.2.435-459.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Storz G. An expanding universe of noncoding RNAs. Science. 2002;296:1260–1263. doi: 10.1126/science.1072249. [DOI] [PubMed] [Google Scholar]
  148. Sun, G., Fu, C., Shen, C., and Shi, Y. (2011). Histone deacetylases in neural stem cells and induced pluripotent stem cells. J Biomed Biotechnol 2011, 835968. [DOI] [PMC free article] [PubMed]
  149. Sun G., Yu R.T., Evans R.M., Shi Y. Orphan nuclear receptor TLX recruits histone deacetylases to repress transcription and regulate neural stem cell proliferation. Proc Natl Acad Sci U S A. 2007;104:15282–15287. doi: 10.1073/pnas.0704089104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Surani M.A., Hayashi K., Hajkova P. Genetic and epigenetic regulators of pluripotency. Cell. 2007;128:747–762. doi: 10.1016/j.cell.2007.02.010. [DOI] [PubMed] [Google Scholar]
  151. Suzuki M.M., Bird A. DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Genet. 2008;9:465–476. doi: 10.1038/nrg2341. [DOI] [PubMed] [Google Scholar]
  152. Temple S. The development of neural stem cells. Nature. 2001;414:112–117. doi: 10.1038/35102174. [DOI] [PubMed] [Google Scholar]
  153. Testa G. The time of timing: how Polycomb proteins regulate neurogenesis. Bioessays. 2011;33:519–528. doi: 10.1002/bies.201100021. [DOI] [PubMed] [Google Scholar]
  154. Tropepe V., Sibilia M., Ciruna B.G., Rossant J., Wagner E.F., van der Kooy D. Distinct neural stem cells proliferate in response to EGF and FGF in the developing mouse telencephalon. Dev Biol. 1999;208:166–188. doi: 10.1006/dbio.1998.9192. [DOI] [PubMed] [Google Scholar]
  155. Tsujimura K., Abematsu M., Kohyama J., Namihira M., Nakashima K. Neuronal differentiation of neural precursor cells is promoted by the methyl-CpG-binding protein MeCP2. Exp Neurol. 2009;219:104–111. doi: 10.1016/j.expneurol.2009.05.001. [DOI] [PubMed] [Google Scholar]
  156. Viré E., Brenner C., Deplus R., Blanchon L., Fraga M., Didelot C., Morey L., Van Eynde A., Bernard D., Vanderwinden J.M., et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature. 2006;439:871–874. doi: 10.1038/nature04431. [DOI] [PubMed] [Google Scholar]
  157. Wade P.A., Pruss D., Wolffe A.P. Histone acetylation: chromatin in action. Trends Biochem Sci. 1997;22:128–132. doi: 10.1016/s0968-0004(97)01016-5. [DOI] [PubMed] [Google Scholar]
  158. Walsh C.P., Bestor T.H. Cytosine methylation and mammalian development. Genes Dev. 1999;13:26–34. doi: 10.1101/gad.13.1.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Wang H., Wang L., Erdjument-Bromage H., Vidal M., Tempst P., Jones R.S., Zhang Y. Role of histone H2A ubiquitination in Polycomb silencing. Nature. 2004;431:873–878. doi: 10.1038/nature02985. [DOI] [PubMed] [Google Scholar]
  160. Weiss A., Cedar H. The role of DNA demethylation during development. Genes Cells. 1997;2:481–486. doi: 10.1046/j.1365-2443.1997.1390337.x. [DOI] [PubMed] [Google Scholar]
  161. Whetstine J.R., Nottke A., Lan F., Huarte M., Smolikov S., Chen Z., Spooner E., Li E., Zhang G., Colaiacovo M., et al. Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell. 2006;125:467–481. doi: 10.1016/j.cell.2006.03.028. [DOI] [PubMed] [Google Scholar]
  162. Wilks A., Seldran M., Jost J.P. An estrogen-dependent demethylation at the 5′ end of the chicken vitellogenin gene is independent of DNA synthesis. Nucleic Acids Res. 1984;12:1163–1177. doi: 10.1093/nar/12.2.1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Wood H.B., Episkopou V. Comparative expression of the mouse Sox1, Sox2 and Sox3 genes from pre-gastrulation to early somite stages. Mech Dev. 1999;86:197–201. doi: 10.1016/s0925-4773(99)00116-1. [DOI] [PubMed] [Google Scholar]
  164. Wu H., Coskun V., Tao J., Xie W., Ge W., Yoshikawa K., Li E., Zhang Y., Sun Y.E. Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science. 2010;329:444–448. doi: 10.1126/science.1190485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Wu H., D’Alessio A.C., Ito S., Wang Z., Cui K., Zhao K., Sun Y.E., Zhang Y. Genome-wide analysis of 5-hydroxymethylcytosine distribution reveals its dual function in transcriptional regulation in mouse embryonic stem cells. Genes Dev. 2011;25:679–684. doi: 10.1101/gad.2036011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Wu H., D’Alessio A.C., Ito S., Xia K., Wang Z., Cui K., Zhao K., Sun Y.E., Zhang Y. Dual functions of Tet1 in transcriptional regulation in mouse embryonic stem cells. Nature. 2011;473:389–393. doi: 10.1038/nature09934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Wu H., Sun Y.E. Reversing DNA methylation: new insights from neuronal activity-induced Gadd45b in adult neurogenesis. Sci Signal. 2009;2:pe17. doi: 10.1126/scisignal.264pe17. [DOI] [PubMed] [Google Scholar]
  168. Wu H., Tao J., Chen P.J., Shahab A., Ge W., Hart R.P., Ruan X., Ruan Y., Sun Y.E. Genome-wide analysis reveals methyl-CpG-binding protein 2-dependent regulation of microRNAs in a mouse model of Rett syndrome. Proc Natl Acad Sci U S A. 2010;107:18161–18166. doi: 10.1073/pnas.1005595107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Wu J., Xie X. Comparative sequence analysis reveals an intricate network among REST, CREB and miRNA in mediating neuronal gene expression. Genome Biol. 2006;7:R85. doi: 10.1186/gb-2006-7-9-r85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Wu J.I., Lessard J., Olave I.A., Qiu Z., Ghosh A., Graef I.A., Crabtree G.R. Regulation of dendritic development by neuron-specific chromatin remodeling complexes. Neuron. 2007;56:94–108. doi: 10.1016/j.neuron.2007.08.021. [DOI] [PubMed] [Google Scholar]
  171. Xu Y., Wu F., Tan L., Kong L., Xiong L., Deng J., Barbera A.J., Zheng L., Zhang H., Huang S., et al. Genome-wide regulation of 5hmC, 5mC, and gene expression by Tet1 hydroxylase in mouse embryonic stem cells. Mol Cell. 2011;42:451–464. doi: 10.1016/j.molcel.2011.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Yamane K., Toumazou C., Tsukada Y., Erdjument-Bromage H., Tempst P., Wong J., Zhang Y. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell. 2006;125:483–495. doi: 10.1016/j.cell.2006.03.027. [DOI] [PubMed] [Google Scholar]
  173. Ye F., Chen Y., Hoang T., Montgomery R.L., Zhao X.H., Bu H., Hu T., Taketo M.M., van Es J.H., Clevers H., et al. HDAC1 and HDAC2 regulate oligodendrocyte differentiation by disrupting the beta-catenin-TCF interaction. Nat Neurosci. 2009;12:829–838. doi: 10.1038/nn.2333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Yoder J.A., Walsh C.P., Bestor T.H. Cytosine methylation and the ecology of intragenomic parasites. Trends Genet. 1997;13:335–340. doi: 10.1016/s0168-9525(97)01181-5. [DOI] [PubMed] [Google Scholar]
  175. Yoo A.S., Staahl B.T., Chen L., Crabtree G.R. MicroRNA-mediated switching of chromatin-remodelling complexes in neural development. Nature. 2009;460:642–646. doi: 10.1038/nature08139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Yoo A.S., Sun A.X., Li L., Shcheglovitov A., Portmann T., Li Y., Lee-Messer C., Dolmetsch R.E., Tsien R.W., Crabtree G.R. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature. 2011;476:228–231. doi: 10.1038/nature10323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Yu I.T., Park J.Y., Kim S.H., Lee J.S., Kim Y.S., Son H. Valproic acid promotes neuronal differentiation by induction of proneural factors in association with H4 acetylation. Neuropharmacology. 2009;56:473–480. doi: 10.1016/j.neuropharm.2008.09.019. [DOI] [PubMed] [Google Scholar]
  178. Zhan X., Shi X., Zhang Z., Chen Y., Wu J.I. Dual role of Brg chromatin remodeling factor in Sonic hedgehog signaling during neural development. Proc Natl Acad Sci U S A. 2011;108:12758–12763. doi: 10.1073/pnas.1018510108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Zhang Y., Reinberg D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. 2001;15:2343–2360. doi: 10.1101/gad.927301. [DOI] [PubMed] [Google Scholar]
  180. Zhao C., Sun G., Li S., Shi Y. A feedback regulatory loop involving microRNA-9 and nuclear receptor TLX in neural stem cell fate determination. Nat Struct Mol Biol. 2009;16:365–371. doi: 10.1038/nsmb.1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Zhao X., Ueba T., Christie B.R., Barkho B., McConnell M.J., Nakashima K., Lein E.S., Eadie B.D., Willhoite A.R., Muotri A.R., et al. Mice lacking methyl-CpG binding protein 1 have deficits in adult neurogenesis and hippocampal function. Proc Natl Acad Sci U S A. 2003;100:6777–6782. doi: 10.1073/pnas.1131928100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Zhao Z., Yu Y., Meyer D., Wu C., Shen W.H. Prevention of early flowering by expression of FLOWERING LOCUS C requires methylation of histone H3 K36. Nat Cell Biol. 2005;7:1256–1260. doi: 10.1038/ncb1329. [DOI] [PubMed] [Google Scholar]

Articles from Protein & Cell are provided here courtesy of Oxford University Press

RESOURCES