Skip to main content

Epigenetic Regulation of Pluripotency

  • Chapter
The Cell Biology of Stem Cells

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 695))

Abstract

Epigenetic regulation refers to the mechanisms that alter gene expression patterns in the absence of changes in the nucleotide sequence of the DNA molecule. The best understood epigenetic marks include posttranslational modifications of the histone tails and DNA methylation. Both play central roles in normal development and in diseases. Pluripotent stem cells have great promise for regenerative medicine and recent efforts have focused on identifying molecular networks that govern pluripotency. This chapter provides an overview of epigenetic regulation in embryonic stem cells. We present a brief introduction into epigenetic mechanisms and focus on their role in pluripotent cells.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Waddington C. The epigenotype. Endeavour 1942; 1:18–20.

    Google Scholar 

  2. Allis CD, Jenuwein T, Reinberg D. Epigenetics, x, 502 p. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 2007).

    Google Scholar 

  3. 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.

    CAS  PubMed  Google Scholar 

  4. Turner BM. Cellular memory and the histone code. Cell 2002; 111:285–91.

    CAS  PubMed  Google Scholar 

  5. Turner BM. Defining an epigenetic code. Nat Cell Biol 2007; 9:2–6.

    CAS  PubMed  Google Scholar 

  6. Spivakov M, Fisher AG. Epigenetic signatures of stem-cell identity. Nat Rev Genet 2007; 8:263–71.

    CAS  PubMed  Google Scholar 

  7. Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature 1981; 292:154–6.

    CAS  PubMed  Google Scholar 

  8. Martin GR. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad Sci USA 1981; 78:7634–8.

    CAS  PubMed  Google Scholar 

  9. Stojkovic M, Lako M, Stojkovic P et al. Derivation of human embryonic stem cells from day-8 blastocysts recovered after three-step in vitro culture. Stem Cells 2004; 22:790–7.

    PubMed  Google Scholar 

  10. Thomson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 1998; 282:1145–7.

    CAS  PubMed  Google Scholar 

  11. Yu J, Thomson JA. Pluripotent stem cell lines. Genes Dev 2008; 22:1987–97.

    CAS  PubMed  Google Scholar 

  12. Murry CE, Keller G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 2008; 132:661–80.

    CAS  PubMed  Google Scholar 

  13. Cowan CA, Klimanskaya I, McMahon J et al. Derivation of embryonic stem-cell lines from human blastocysts. N Engl J Med 2004; 350:1353–6.

    CAS  PubMed  Google Scholar 

  14. Sidhu KS, Ryan JP, Tuch BE. Derivation of a new human embryonic stem cell line, endeavour-1 and its clonal propagation. Stem Cells Dev 2008; 17:41–51.

    CAS  PubMed  Google Scholar 

  15. Bao S et al. Epigenetic reversion of post-implantation epiblast to pluripotent embryonic stem cells. Nature 2009; 461:1292–5.

    CAS  PubMed  Google Scholar 

  16. Matsui Y, Zsebo K, Hogan BL. Derivation of pluripotential embryonic stem cells from murine primordial germ cells in culture. Cell 1992; 70:841–7.

    CAS  PubMed  Google Scholar 

  17. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126:663–76.

    CAS  PubMed  Google Scholar 

  18. Jaenisch R, Young R. Stem cells, the molecular circuitry of pluripotency and nuclear reprogramming. Cell 2008; 132:567–82.

    CAS  PubMed  Google Scholar 

  19. Boyer LA, Lee TI, Cole MF et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 2005; 122:947–56.

    CAS  PubMed  Google Scholar 

  20. Wang J, Rao S, Chu J et al. A protein interaction network for pluripotency of embryonic stem cells. Nature 2006; 444:364–8.

    CAS  PubMed  Google Scholar 

  21. Luger K, Mader AW, Richmond RK et al. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 1997; 389:251–60.

    CAS  PubMed  Google Scholar 

  22. Fraser P, Bickmore W. Nuclear organization of the genome and the potential for gene regulation. Nature 2007; 447:413–7.

    CAS  PubMed  Google Scholar 

  23. Campos EI, Reinberg D. Histones: annotating chromatin. Annu Rev Genet 2009; 43:559–99.

    CAS  PubMed  Google Scholar 

  24. Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell 2007; 128:669–81.

    CAS  PubMed  Google Scholar 

  25. Morrison AJ, Shen X. Chromatin remodelling beyond transcription: the INO80 and SWR1 complexes. Nat Rev Mol Cell Biol 2009; 10:373–84.

    CAS  PubMed  Google Scholar 

  26. Li B, Carey M, Workman JL. The role of chromatin during transcription. Cell 2007; 128:707–19.

    CAS  PubMed  Google Scholar 

  27. Gutierrez J et al. Chromatin remodeling by SWI/SNF results in nucleosome mobilization to preferential positions in the rat osteocalcin gene promoter. J Biol Chem 2007; 282:9445–57.

    CAS  PubMed  Google Scholar 

  28. Kouzarides T. Chromatin modifications and their function. Cell 2007; 128:693–705.

    CAS  PubMed  Google Scholar 

  29. Bernstein BE, Mikkelsen TS, Xie X et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 2006; 125:315–26.

    CAS  PubMed  Google Scholar 

  30. Mikkelsen TS, Ku M, Jaffe DB et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 2007; 448:553–60.

    CAS  PubMed  Google Scholar 

  31. Pietersen AM, van Lohuizen M. Stem cell regulation by polycomb repressors: postponing commitment. Curr Opin Cell Biol 2008; 20:201–7.

    CAS  PubMed  Google Scholar 

  32. Henikoff S. Nucleosome destabilization in the epigenetic regulation of gene expression. Nat Rev Genet 2008; 9:15–26.

    CAS  PubMed  Google Scholar 

  33. Sterner DE, Berger SL. Acetylation of histones and transcription-related factors. Microbiol Mol Biol Rev 2000; 64:435–59.

    CAS  PubMed  Google Scholar 

  34. Sims RJ, 3rd, Nishioka K, Reinberg D. Histone lysine methylation: a signature for chromatin function. Trends Genet 2003; 19:629–39.

    CAS  PubMed  Google Scholar 

  35. Nowak SJ, Corces VG. Phosphorylation of histone H3: a balancing act between chromosome condensation and transcriptional activation. Trends Genet 2004; 20:214–20.

    CAS  PubMed  Google Scholar 

  36. Shilatifard A. Chromatin modifications by methylation and ubiquitination: implications in the regulation of gene expression. Annu Rev Biochem 2006; 75:243–69.

    CAS  PubMed  Google Scholar 

  37. Nathan D, Ingvarsdottir K, Sterner DE et al. Histone sumoylation is a negative regulator in Saccharomyces cerevisiae and shows dynamic interplay with positive-acting histone modifications. Genes Dev 2006; 20:966–76.

    CAS  PubMed  Google Scholar 

  38. Hassa PO, Haenni SS, Elser M et al. Nuclear ADP-ribosylation reactions in mammalian cells: where are we today and where are we going? Microbiol Mol Biol Rev 2006; 70:789–829.

    CAS  PubMed  Google Scholar 

  39. Cuthbert GL, Daujat S, Snowden AW et al. Histone deimination antagonizes arginine methylation. Cell 2004; 118:545–53.

    CAS  PubMed  Google Scholar 

  40. Nelson CJ, Santos-Rosa H, Kouzarides T. Proline isomerization of histone H3 regulates lysine methylation and gene expression. Cell 2006; 126:905–16.

    CAS  PubMed  Google Scholar 

  41. Schuettengruber B, Chourrout D, Vervoort M et al. Genome regulation by polycomb and trithorax proteins. Cell 2007; 128:735–45.

    CAS  PubMed  Google Scholar 

  42. Elderkin S, Maertens GN, Endoh M et al. A phosphorylated form of Mel-18 targets the Ring1B histone H2A ubiquitin ligase to chromatin. Mol Cell 2007; 28:107–20.

    CAS  PubMed  Google Scholar 

  43. Ku M, Koche RP, Rheinbay E et al. Genomewide analysis of PRC1 and PRC2 occupancy identifies two classes of bivalent domains. PLoS Genet 2008; 4:e1000242.

    PubMed  Google Scholar 

  44. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002; 16:6–21.

    CAS  PubMed  Google Scholar 

  45. Grandjean V, Yaman R, Cuzin F et al. Inheritance of an epigenetic mark: the CpG DNA methyltransferase 1 is required for de novo establishment of a complex pattern of nonCpG methylation. PLoS One 2007; 2:e1136.

    PubMed  Google Scholar 

  46. Ramsahoye BH, Biniszkiewicz D, Lyko F, Clark V et al. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc Natl Acad Sci USA 2000; 97:5237–42.

    CAS  PubMed  Google Scholar 

  47. Finnegan EJ, Peacock WJ, Dennis ES. DNA methylation, a key regulator of plant development and other processes. Curr Opin Genet Dev 2000; 10:217–23.

    CAS  PubMed  Google Scholar 

  48. Lister R, Pelizzola M, Dowen RH et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009; 462:315–22.

    CAS  PubMed  Google Scholar 

  49. Meissner A, Gnirke A, Bell GW et al. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res 2005; 33:5868–77.

    CAS  PubMed  Google Scholar 

  50. Morgan HD, Santos F, Green K et al. Epigenetic reprogramming in mammals. Hum Mol Genet 2005; 14 Spec No 1:R47–58.

    CAS  PubMed  Google Scholar 

  51. Klose RJ, Bird AP. Genomic DNA methylation: the mark and its mediators. Trends Biochem Sci 2006; 31:89–97.

    CAS  PubMed  Google Scholar 

  52. Bestor TH. The DNA methyltransferases of mammals. Hum Mol Genet 2000; 9:2395–402.

    CAS  PubMed  Google Scholar 

  53. Goll MG, Bestor TH. Eukaryotic cytosine methyltransferases. Annu Rev Biochem 2005; 74:481–514.

    CAS  PubMed  Google Scholar 

  54. Okano M, Bell DW, Haber DA et al. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 1999; 99:247–57.

    CAS  PubMed  Google Scholar 

  55. Aapola U, Kawasaki K, Scott HS et al. Isolation and initial characterization of a novel zinc finger gene, DNMT3L, on 21q22.3, related to the cytosine-5-methyltransferase 3 gene family. Genomics 2000; 65:293–8.

    CAS  PubMed  Google Scholar 

  56. Ooi SK, Qiu C, Bernstein E et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 2007; 448:714–7.

    CAS  PubMed  Google Scholar 

  57. Meshorer E, Misteli T. Chromatin in pluripotent embryonic stem cells and differentiation. Nat Rev Mol Cell Biol 2006; 7:540–6.

    CAS  PubMed  Google Scholar 

  58. Francastel C, Schubeler D, Martin DI et al. Nuclear compartmentalization and gene activity. Nat Rev Mol Cell Biol 2000; 1:137–43.

    CAS  PubMed  Google Scholar 

  59. Arney KL, Fisher AG. Epigenetic aspects of differentiation. J Cell Sci 2004; 117:4355–63.

    CAS  PubMed  Google Scholar 

  60. Meshorer E, Yellajoshula D, George E et al. Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells. Dev Cell 2006; 10:105–16.

    CAS  PubMed  Google Scholar 

  61. Efroni S, Duttagupta R, Cheng J et al. Global transcription in pluripotent embryonic stem cells. Cell Stem Cell 2008; 2:437–47.

    CAS  PubMed  Google Scholar 

  62. Liang J, Wan M, Zhang Y, et al. Nanog and Oct4 associate with unique transcriptional repression complexes in embryonic stem cells. Nat Cell Biol 2008; 10:731–9.

    CAS  PubMed  Google Scholar 

  63. Kaji K, Caballero IM, MacLeod R et al. The NuRD component Mbd3 is required for pluripotency of embryonic stem cells. Nat Cell Biol 2006; 8:285–92.

    CAS  PubMed  Google Scholar 

  64. Gaspar-Maia A, Alajem A, Polesso F et al. Chd1 regulates open chromatin and pluripotency of embryonic stem cells. Nature 2009; 460:863–8.

    CAS  PubMed  Google Scholar 

  65. Guenther MG, Levine SS, Boyer LA et al. A chromatin landmark and transcription initiation at most promoters in human cells. Cell 2007; 130:77–88.

    CAS  PubMed  Google Scholar 

  66. Pasini D, Hansen KH, Christensen J et al. Coordinated regulation of transcriptional repression by the RBP2 H3K4 demethylase and Polycomb-Repressive Complex 2. Genes Dev 2008; 22:1345–55.

    CAS  PubMed  Google Scholar 

  67. Chen X, Xu H, Yuan P et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 2008; 133:1106–17.

    CAS  PubMed  Google Scholar 

  68. Fazzio TG, Huff JT, Panning B. Chromatin regulation Tip(60)s the balance in embryonic stem cell self-renewal. Cell Cycle 2008; 7:3302–6.

    CAS  PubMed  Google Scholar 

  69. Torres-Padilla ME, Parfitt DE, Kouzarides T et al. Histone arginine methylation regulates pluripotency in the early mouse embryo. Nature 2007; 445:214–8.

    CAS  PubMed  Google Scholar 

  70. Wu Q, Bruce AW, Jedrusik A et al. CARM1 is required in embryonic stem cells to maintain pluripotency and resist differentiation. Stem Cells 2009; 27:2637–45.

    CAS  PubMed  Google Scholar 

  71. Heintzman ND, Hon GC, Hawkins RD et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 2009; 459:108–12.

    CAS  PubMed  Google Scholar 

  72. Heintzman ND, Ren B. Finding distal regulatory elements in the human genome. Curr Opin Genet Dev 2009; 19:541–9.

    CAS  PubMed  Google Scholar 

  73. Boiani M, Scholer HR. Regulatory networks in embryo-derived pluripotent stem cells. Nat Rev Mol Cell Biol 2005; 6:872–84.

    CAS  PubMed  Google Scholar 

  74. Boyer LA, Plath K, Zeitlinger J et al. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature 2006; 441:349–53.

    CAS  PubMed  Google Scholar 

  75. Lee TI, Jenner RG, Boyer LA et al. Control of developmental regulators by Polycomb in human embryonic stem cells. Cell 2006; 125:301–13.

    CAS  PubMed  Google Scholar 

  76. Shen X, Liu Y, Hsu YJ et al. EZH1 mediates methylation on histone H3 lysine 27 and complements EZH2 in maintaining stem cell identity and executing pluripotency. Mol Cell 2008; 32:491–502.

    CAS  PubMed  Google Scholar 

  77. Chamberlain SJ, Yee D, Magnuson T. Polycomb repressive complex 2 is dispensable for maintenance of embryonic stem cell pluripotency. Stem Cells 2008; 26:1496–505.

    CAS  PubMed  Google Scholar 

  78. Pasini D, Bracken AP, Hansen JB et al. The polycomb group protein Suz12 is required for embryonic stem cell differentiation. Mol Cell Biol 2007; 27:3769–79.

    CAS  PubMed  Google Scholar 

  79. Peng JC, Valouev A, Swigut T et al. Jarid2/Jumonji coordinates control of PRC2 enzymatic activity and target gene occupancy in pluripotent cells. Cell 2009; 139:1290–302.

    PubMed  Google Scholar 

  80. Shen X, Kim W, Fujiwara Y et al. Jumonji modulates polycomb activity and self-renewal versus differentiation of stem cells. Cell 2009; 139:1303–14.

    PubMed  Google Scholar 

  81. Li G, Margueron R, Ku M et al. Jarid2 and PRC2, partners in regulating gene expression. Genes Dev. 2010 Feb 15;24(4):368–80.

    PubMed  Google Scholar 

  82. Pasini D, Cloos PA, Walfridsson J et al. JARID2 regulates binding of the Polycomb repressive complex 2 to target genes in ES cells. Nature. 2010 Jan 14. [Epub ahead of print].

    Google Scholar 

  83. Mikkelsen TS, Hanna J, Zhang X et al. Dissecting direct reprogramming through integrative genomic analysis. Nature 2008; 454:49–55.

    CAS  PubMed  Google Scholar 

  84. Bilodeau S, Kagey MH, Frampton GM et al. SetDB1 contributes to repression of genes encoding developmental regulators and maintenance of ES cell state. Genes Dev 2009; 23:2484–9.

    CAS  PubMed  Google Scholar 

  85. Squazzo SL, O’Geen H, Komashko VM et al. Suz12 binds to silenced regions of the genome in a cell-type-specific manner. Genome Res 2006; 16:890–900.

    CAS  PubMed  Google Scholar 

  86. Creyghton MP, Markoulaki S, Levine SS et al. H2AZ is enriched at polycomb complex target genes in ES cells and is necessary for lineage commitment. Cell 2008; 135:649–61.

    CAS  PubMed  Google Scholar 

  87. Hardy S, Jacques PE, Gévry N et al. The euchromatic and heterochromatic landscapes are shaped by antagonizing effects of transcription on H2A.Z deposition. PLoS Genet 2009; 5:e1000687.

    PubMed  Google Scholar 

  88. Xie W, Song C, Young NL 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.

    CAS  PubMed  Google Scholar 

  89. Martens JH, O’Sullivan RJ, Braunschweig U et al. The profile of repeat-associated histone lysine methylation states in the mouse epigenome. EMBO J 2005; 24:800–12.

    CAS  PubMed  Google Scholar 

  90. Daujat S, Weiss T, Mohn F et al. H3K64 trimethylation marks heterochromatin and is dynamically remodeled during developmental reprogramming. Nat Struct Mol Biol 2009; 16:777–81.

    CAS  PubMed  Google Scholar 

  91. Loh YH, Zhang W, Chen X et al. Jmjd1a and Jmjd2c histone H3 Lys 9 demethylases regulate self-renewal in embryonic stem cells. Genes Dev 2007; 21:2545–57.

    CAS  PubMed  Google Scholar 

  92. Jackson M, Krassowska A, Gilbert N et al. Severe global DNA hypomethylation blocks differentiation and induces histone hyperacetylation in embryonic stem cells. Mol Cell Biol 2004; 24:8862–71.

    CAS  PubMed  Google Scholar 

  93. Tsumura A, Hayakawa T, Kumaki Y et al. Maintenance of self-renewal ability of mouse embryonic stem cells in the absence of DNA methyltransferases Dnmt1, Dnmt3a and Dnmt3b. Genes Cells 2006; 11:805–14.

    CAS  PubMed  Google Scholar 

  94. Jackson-Grusby L, Beard C, Possemato R et al. Loss of genomic methylation causes p53-dependent apoptosis and epigenetic deregulation. Nat Genet 2001; 27:31–9.

    CAS  PubMed  Google Scholar 

  95. Gaudet F, Hodgson JG, Eden A et al. Induction of tumors in mice by genomic hypomethylation. Science 2003; 300:489–92.

    CAS  PubMed  Google Scholar 

  96. Bourc’his D, Bestor TH. Meiotic catastrophe and retrotransposon reactivation in male germ cells lacking Dnmt3L. Nature 2004; 431:96–9.

    CAS  Google Scholar 

  97. Dodge JE, Okano M, Dick F et al. Inactivation of Dnmt3b in mouse embryonic fibroblasts results in DNA hypomethylation, chromosomal instability and spontaneous immortalization. J Biol Chem 2005; 280:17986–91.

    CAS  PubMed  Google Scholar 

  98. Nimura K, Ishida C, Koriyama H et al. Dnmt3a2 targets endogenous Dnmt3L to ES cell chromatin and induces regional DNA methylation. Genes Cells 2006; 11:1225–37.

    CAS  PubMed  Google Scholar 

  99. Fouse SD, Shen Y, Pellegrini M et al. Promoter CpG methylation contributes to ES cell gene regulation in parallel with Oct4/Nanog, PcG complex and histone H3 K4/K27 trimethylation. Cell Stem Cell 2008; 2:160–9.

    CAS  PubMed  Google Scholar 

  100. Meissner A, Mikkelsen TS, Gu H et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 2008; 454:766–70.

    CAS  PubMed  Google Scholar 

  101. Weber M, Hellmann I, Stadler MB et al. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet 2007; 39:457–66.

    CAS  PubMed  Google Scholar 

  102. Mohn F, Weber M, Rebhan M et al. Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol Cell 2008; 30:755–66.

    CAS  PubMed  Google Scholar 

  103. Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet 2009; 10:295–304.

    CAS  PubMed  Google Scholar 

  104. Haines TR, Rodenhiser DI, Ainsworth PJ. Allele-specific nonCpG methylation of the Nf1 gene during early mouse development. Dev Biol 2001; 240:585–98.

    CAS  PubMed  Google Scholar 

  105. Tahiliani M, Koh KP, Shen Y et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009; 324:930–5.

    CAS  PubMed  Google Scholar 

  106. Niwa H. How is pluripotency determined and maintained? Development 2007; 134:635–46.

    CAS  PubMed  Google Scholar 

  107. Silva J, Smith A. Capturing pluripotency. Cell 2008; 132:532–6.

    CAS  PubMed  Google Scholar 

  108. Chi AS, Bernstein BE. Developmental biology. Pluripotent chromatin state. Science 2009; 323:220–1.

    CAS  PubMed  Google Scholar 

  109. Gu H, Bock C, Mikkelsen TS et al. Genome-scale DNA methylation mapping of clinical samples at single-nucleotide resolution. Nat Methods. 2010 Feb;7(2):133–6.

    CAS  PubMed  Google Scholar 

  110. Goren A, Ozsolak F, Shoresh N et al. Chromatin profiling by directly sequencing small quantities of immunoprecipitated DNA. Nat Methods 7:47–9.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alexander Meissner .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2010 Landes Bioscience and Springer Science+Business Media

About this chapter

Cite this chapter

Tomazou, E.M., Meissner, A. (2010). Epigenetic Regulation of Pluripotency. In: Meshorer, E., Plath, K. (eds) The Cell Biology of Stem Cells. Advances in Experimental Medicine and Biology, vol 695. Springer, Boston, MA. https://doi.org/10.1007/978-1-4419-7037-4_3

Download citation

Publish with us

Policies and ethics