Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Kdm2b promotes induced pluripotent stem cell generation by facilitating gene activation early in reprogramming

An Erratum to this article was published on 30 May 2012

This article has been updated

Abstract

Transcription-factor-directed reprogramming from somatic cells to induced pluripotent stem cells (iPSCs) is by nature an epigenetic process of cell fate change. Previous studies have demonstrated that this inefficient process can be facilitated by the inclusion of additional factors. To gain insight into the reprogramming mechanism, we aimed to identify epigenetic enzymes capable of promoting iPSC generation. Here we show that Kdm2b, a histone H3 Lys 36 dimethyl (H3K36me2)-specific demethylase, has the capacity to promote iPSC generation. This capacity depends on its demethylase and DNA-binding activities, but is largely independent of its role in antagonizing senescence. Kdm2b functions at the beginning of the reprogramming process and enhances activation of early responsive genes in reprogramming. Kdm2b contributes to gene activation by binding to and demethylating the gene promoters. Our studies not only identify an important epigenetic factor for iPSC generation, but also reveal the molecular mechanism underlying how Kdm2b contributes to reprogramming.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Kdm2b promotes iPSC generation.
Figure 2: iPSCs generated by enforced expression of OSK plus Kdm2b are pluripotent.
Figure 3: Kdm2b facilitates iPSC generation in a JmjC- and ZF-domain-dependent manner.
Figure 4: Kdm2b promotes iPSC generation independently of Ink4a/Arf.
Figure 5: Kdm2b exerts its effect from the beginning of the reprogramming process and facilitates gene activation early in reprogramming.
Figure 6: Kdm2b activates early responsive genes in reprogramming.
Figure 7: Kdm2b localizes on and demethylates the promoter region of early-activated genes.
Figure 8: Inhibition of the expression of early-activated genes compromises the capacity of Kdm2b to enhance iPSC generation.

Similar content being viewed by others

Accession codes

Accessions

ArrayExpress

Change history

  • 25 April 2012

    In the version of this Article originally published, in the legend for Fig. 4, the statement "Data in a and d represent the mean of two independent experiments" should have read: "Data in a and e represent the mean of two independent experiments". This error has been corrected in both the HTML and PDF versions of the Article.

References

  1. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  Google Scholar 

  2. Hanna, J. H., Saha, K. & Jaenisch, R. Pluripotency and cellular reprogramming: facts, hypotheses, unresolved issues. Cell 143, 508–525 (2010).

    Article  CAS  Google Scholar 

  3. Stadtfeld, M. & Hochedlinger, K. Induced pluripotency: history, mechanisms, and applications. Genes Dev 24, 2239–2263 (2010).

    Article  CAS  Google Scholar 

  4. Plath, K. & Lowry, W. M. Progress in understanding reprogramming to the induced pluripotent state. Nat. Rev. Genet. 12, 253–265 (2011).

    Article  CAS  Google Scholar 

  5. Li, H. et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature 460, 1136–1139 (2009).

    Article  CAS  Google Scholar 

  6. Hanna, J. et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature 462, 595–601 (2009).

    Article  CAS  Google Scholar 

  7. Utikal, J. et al. Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature 460, 1145–1148 (2009).

    Article  CAS  Google Scholar 

  8. Kawamura, T. et al. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 460, 1140–1144 (2009).

    Article  CAS  Google Scholar 

  9. Hong, H. et al. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature 460, 1132–1135 (2009).

    Article  CAS  Google Scholar 

  10. Banito, A. et al. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev 23, 2134–2139 (2009).

    Article  CAS  Google Scholar 

  11. Samavarchi-Tehrani, P. et al. Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming. Cell Stem Cell 7, 64–77 (2010).

    Article  CAS  Google Scholar 

  12. Maherali, N. & Hochedlinger, K. Tgfβ signal inhibition cooperates in the induction of iPSCs and replaces Sox2 and cMyc. Curr. Biol. 19, 1718–1723 (2009).

    Article  CAS  Google Scholar 

  13. Li, R. et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63 (2010).

    Article  CAS  Google Scholar 

  14. Ichida, J. K. et al. A small-molecule inhibitor of tgf-β signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell 5, 491–503 (2009).

    Article  CAS  Google Scholar 

  15. Goldberg, A. D., Allis, C. D. & Bernstein, E. Epigenetics: a landscape takes shape. Cell 128, 635–638 (2007).

    Article  CAS  Google Scholar 

  16. Mikkelsen, T. S. et al. Dissecting direct reprogramming through integrative genomic analysis. Nature 454, 49–55 (2008).

    Article  CAS  Google Scholar 

  17. Huangfu, D. et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol. 26, 795–797 (2008).

    Article  CAS  Google Scholar 

  18. Liang, G., Taranova, O., Xia, K. & Zhang, Y. Butyrate promotes induced pluripotent stem cell generation. J. Biol. Chem. 285, 25516–25521 (2010).

    Article  CAS  Google Scholar 

  19. Shi, Y. et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell 3, 568–574 (2008).

    Article  CAS  Google Scholar 

  20. Singhal, N. et al. Chromatin-remodeling components of the BAF complex facilitate reprogramming. Cell 141, 943–955 (2010).

    Article  CAS  Google Scholar 

  21. He, J., Kallin, E. M., Tsukada, Y. & Zhang, Y. The H3K36 demethylase Jhdm1b/Kdm2b regulates cell proliferation and senescence through p15(Ink4b). Nat. Struct. Mol. Biol. 15, 1169–1175 (2008).

    Article  CAS  Google Scholar 

  22. Tsukada, Y. et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature 439, 811–816 (2006).

    Article  CAS  Google Scholar 

  23. Blackledge, N. P. et al. CpG islands recruit a histone H3 lysine 36 demethylase. Mol. Cell 38, 179–190 (2010).

    Article  CAS  Google Scholar 

  24. Voo, K. S., Carlone, D. L., Jacobsen, B. M., Flodin, A. & Skalnik, D. G. Cloning of a mammalian transcriptional activator that binds unmethylated CpG motifs and shares a CXXC domain with DNA methyltransferase, human trithorax, and methyl-CpG binding domain protein 1. Mol. Cell Biol. 20, 2108–2121 (2000).

    Article  CAS  Google Scholar 

  25. Tzatsos, A., Pfau, R., Kampranis, S. C. & Tsichlis, P. N. Ndy1/KDM2B immortalizes mouse embryonic fibroblasts by repressing the Ink4a/Arf locus. Proc. Natl Acad. Sci. USA 106, 2641–2646 (2009).

    Article  CAS  Google Scholar 

  26. Pfau, R. et al. Members of a family of JmjC domain-containing oncoproteins immortalize embryonic fibroblasts via a JmjC domain-dependent process. Proc. Natl Acad. Sci. USA 105, 1907–1912 (2008).

    Article  CAS  Google Scholar 

  27. Brambrink, T. et al. Sequential expression of pluripotency markers during direct reprogramming of mouse somatic cells. Cell Stem Cell 2, 151–159 (2008).

    Article  CAS  Google Scholar 

  28. Stadtfeld, M., Maherali, N., Breault, D. T. & Hochedlinger, K. Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell 2, 230–240 (2008).

    Article  CAS  Google Scholar 

  29. Sridharan, R. et al. Role of the murine reprogramming factors in the induction of pluripotency. Cell 136, 364–377 (2009).

    Article  CAS  Google Scholar 

  30. Silva, J. et al. Nanog is the gateway to the pluripotent ground state. Cell 138, 722–737 (2009).

    Article  CAS  Google Scholar 

  31. Richardson, R. J., Dixon, J., Jiang, R. & Dixon, M. J. Integration of IRF6 and Jagged2 signalling is essential for controlling palatal adhesion and fusion competence. Hum. Mol. Genet. 18, 2632–2642 (2009).

    Article  CAS  Google Scholar 

  32. Thomason, H. A. et al. Cooperation between the transcription factors p63 and IRF6 is essential to prevent cleft palate in mice. J. Clin. Invest. 120, 1561–1569 (2010).

    Article  CAS  Google Scholar 

  33. Gierl, M. S., Karoulias, N., Wende, H., Strehle, M. & Birchmeier, C. The zinc-finger factor Insm1 (IA-1) is essential for the development of pancreatic beta cells and intestinal endocrine cells. Genes Dev 20, 2465–2478 (2006).

    Article  CAS  Google Scholar 

  34. Mellitzer, G. et al. IA1 is NGN3-dependent and essential for differentiation of the endocrine pancreas. EMBO J. 25, 1344–1352 (2006).

    Article  CAS  Google Scholar 

  35. Farkas, L. M. et al. Insulinoma-associated 1 has a panneurogenic role and promotes the generation and expansion of basal progenitors in the developing mouse neocortex. Neuron 60, 40–55 (2008).

    Article  CAS  Google Scholar 

  36. Heldin, C. H., Landstrom, M. & Moustakas, A. Mechanism of TGF-beta signaling to growth arrest, apoptosis, and epithelial-mesenchymal transition. Curr. Opin. Cell Biol. 21, 166–176 (2009).

    Article  CAS  Google Scholar 

  37. Chen, T. et al. E-cadherin-mediated cell-cell contact is critical for induced pluripotent stem cell generation. Stem Cells 28, 1315–1325 (2010).

    Article  CAS  Google Scholar 

  38. Redmer, T. et al. E-cadherin is crucial for embryonic stem cell pluripotency and can replace OCT4 during somatic cell reprogramming. EMBO Rep. 12, 720–726 (2011).

    Article  CAS  Google Scholar 

  39. Wang, T. et al. The histone demethylases Jhdm1a/1b enhance somatic cell reprogramming in a vitamin-C-dependent manner. Cell Stem Cell 9, 575–587 (2011).

    Article  CAS  Google Scholar 

  40. Esteban, M. A. et al. Vitamin C enhances the generation of mouse and human induced pluripotent stem cells. Cell Stem Cell 6, 71–79 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank UNC Animal Models Core for chimaera generation and UNC Functional Genomic Core for microarray analysis. We thank S. Yamaguchi and K-H. Hong for helpful discussions. This work is supported by U01DK089565 from the NIH. Y.Z. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

G.L. and Y.Z. designed all of the experiments and wrote the manuscript. G.L. performed most of the experiments. J.H. constructed the Kdm2b plasmids and lentiviral doxycyclin inducible system.

Corresponding author

Correspondence to Yi Zhang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

Supplementary Information (PDF 542 kb)

Supplementary Table 1

Supplementary Information (XLSX 284 kb)

Supplementary Table 2

Supplementary Information (XLSX 10 kb)

Supplementary Table 3

Supplementary Information (XLSX 10 kb)

Supplementary Table 4

Supplementary Information (XLSX 9 kb)

Supplementary Table 5

Supplementary Information (XLSX 12 kb)

Supplementary Table 6

Supplementary Information (XLSX 10 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Liang, G., He, J. & Zhang, Y. Kdm2b promotes induced pluripotent stem cell generation by facilitating gene activation early in reprogramming. Nat Cell Biol 14, 457–466 (2012). https://doi.org/10.1038/ncb2483

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2483

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing