The H3K27 demethylase Utx regulates somatic and germ cell epigenetic reprogramming (original) (raw)

Nature volume 488, pages 409–413 (2012)Cite this article

Subjects

This article has been updated

Abstract

Induced pluripotent stem cells (iPSCs) can be derived from somatic cells by ectopic expression of different transcription factors, classically Oct4 (also known as Pou5f1), Sox2, Klf4 and Myc (abbreviated as OSKM)1. This process is accompanied by genome-wide epigenetic changes2,3,4,5, but how these chromatin modifications are biochemically determined requires further investigation. Here we show in mice and humans that the histone H3 methylated Lys 27 (H3K27) demethylase Utx6,7,8,9 (also known as Kdm6a) regulates the efficient induction, rather than maintenance, of pluripotency. Murine embryonic stem cells lacking Utx can execute lineage commitment and contribute to adult chimaeric animals; however, somatic cells lacking Utx fail to robustly reprogram back to the ground state of pluripotency. Utx directly partners with OSK reprogramming factors and uses its histone demethylase catalytic activity to facilitate iPSC formation. Genomic analysis indicates that Utx depletion results in aberrant dynamics of H3K27me3 repressive chromatin demethylation in somatic cells undergoing reprogramming. The latter directly hampers the derepression of potent pluripotency promoting gene modules (including Sall1, Sall4 and Utf1), which can cooperatively substitute for exogenous OSK supplementation in iPSC formation. Remarkably, Utx safeguards the timely execution of H3K27me3 demethylation observed in embryonic day 10.5–11 primordial germ cells (PGCs)10, and Utx-deficient PGCs show cell-autonomous aberrant epigenetic reprogramming dynamics during their embryonic maturation in vivo. Subsequently, this disrupts PGC development by embryonic day 12.5, and leads to diminished germline transmission in mouse chimaeras generated from Utx-knockout pluripotent cells. Thus, we identify Utx as a novel mediator with distinct functions during the re-establishment of pluripotency and germ cell development. Furthermore, our findings highlight the principle that molecular regulators mediating loss of repressive chromatin during in vivo germ cell reprogramming can be co-opted during in vitro reprogramming towards ground state pluripotency.

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

Access options

Subscribe to this journal

Receive 51 print issues and online access

$199.00 per year

only $3.90 per issue

Buy this article

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

Additional access options:

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Microarray data are available at the National Center for Biotechnology Information Gene Expression Omnibus database under the series accession number GSE37822.

Change history

The second supplementary information file (Supplementary Tables 1-5) was incorrect and has been updated.

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. Koche, R. P. et al. Reprogramming factor expression initiates widespread targeted chromatin remodeling. Cell Stem Cell 8, 96–105 (2011)
    Article CAS Google Scholar
  3. Hemberger, M., Dean, W. & Reik, W. Epigenetic dynamics of stem cells and cell lineage commitment: digging Waddington’s canal. Nature Rev. Mol. Cell Biol. 10, 526–537 (2009)
    Article CAS Google Scholar
  4. Singhal, N. et al. Chromatin-remodeling components of the BAF complex facilitate reprogramming. Cell 141, 943–955 (2010)
    Article CAS Google Scholar
  5. Onder, T. T. et al. Chromatin-modifying enzymes as modulators of reprogramming. Nature 483, 598–602 (2012)
    Article ADS CAS Google Scholar
  6. Agger, K. et al. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature 449, 731–734 (2007)
    Article ADS CAS Google Scholar
  7. Hong, S. et al. Identification of JmjC domain-containing UTX and JMJD3 as histone H3 lysine 27 demethylases. Proc. Natl Acad. Sci. USA 104, 18439–18444 (2007)
    Article ADS CAS Google Scholar
  8. Sen, G. L., Webster, D. E., Barragan, D. I., Chang, H. Y. & Khavari, P. A. Control of differentiation in a self-renewing mammalian tissue by the histone demethylase JMJD3. Genes Dev. 22, 1865–1870 (2008)
    Article CAS Google Scholar
  9. Lan, F. et al. A histone H3 lysine 27 demethylase regulates animal posterior development. Nature 449, 689–694 (2007)
    Article ADS CAS Google Scholar
  10. Hajkova, P. et al. Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 452, 877–881 (2008)
    Article ADS CAS Google Scholar
  11. Tesar, P. J. et al. New cell lines from mouse epiblast share defining features with human embryonic stem cells. Nature 448, 196–199 (2007)
    Article ADS CAS Google Scholar
  12. Brons, I. G. et al. Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature 448, 191–195 (2007)
    Article ADS CAS Google Scholar
  13. Guo, G., Huang, Y., Humphreys, P., Wang, X. & Smith, A. A PiggyBac-based recessive screening method to identify pluripotency regulators. PLoS ONE 6, e18189 (2011)
    Article ADS CAS Google Scholar
  14. Lee, S., Lee, J. W. & Lee, S. K. UTX, a histone H3-lysine 27 demethylase, acts as a critical switch to activate the cardiac developmental program. Dev. Cell 22, 25–37 (2011)
    Article Google Scholar
  15. Hanna, J. et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature 462, 595–601 (2009)
    Article ADS CAS Google Scholar
  16. Montgomery, N. D. et al. The murine polycomb group protein Eed is required for global histone H3 lysine-27 methylation. Curr. Biol. 15, 942–947 (2005)
    Article CAS Google Scholar
  17. 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
  18. Silva, J. et al. Nanog is the gateway to the pluripotent ground state. Cell 138, 722–737 (2009)
    Article CAS Google Scholar
  19. Hansen, K. H. et al. A model for transmission of the H3K27me3 epigenetic mark. Nature Cell Biol. 10, 1291–1300 (2008)
    Article CAS Google Scholar
  20. Sridharan, R. et al. Role of the murine reprogramming factors in the induction of pluripotency. Cell 136, 364–377 (2009)
    Article CAS Google Scholar
  21. Hayashi, K., Ohta, H., Kurimoto, K., Aramaki, S. & Saitou, M. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 146, 519–532 (2011)
    Article CAS Google Scholar
  22. Leitch, H. G. et al. Embryonic germ cells from mice and rats exhibit properties consistent with a generic pluripotent ground state. Development 137, 2279–2287 (2010)
    Article CAS Google Scholar
  23. Irizarry, R. A. et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249–264 (2003)
    Article Google Scholar
  24. Boyer, L. A. et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 122, 947–956 (2005)
    Article CAS Google Scholar
  25. Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007)
    Article ADS CAS Google Scholar
  26. Loh, Y. H. et al. The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nature Genet. 38, 431–440 (2006)
    Article CAS Google Scholar
  27. Kim, J., Chu, J., Shen, X., Wang, J. & Orkin, S. H. An extended transcriptional network for pluripotency of embryonic stem cells. Cell 132, 1049–1061 (2008)
    Article CAS Google Scholar
  28. Bruce, A. W. et al. Genome-wide analysis of repressor element 1 silencing transcription factor/neuron-restrictive silencing factor (REST/NRSF) target genes. Proc. Natl Acad. Sci. USA 101, 10458–10463 (2004)
    Article ADS CAS Google Scholar
  29. Zhao, X. D. et al. Whole-genome mapping of histone H3 Lys4 and 27 trimethylations reveals distinct genomic compartments in human embryonic stem cells. Cell Stem Cell 1, 286–298 (2007)
    Article CAS Google Scholar
  30. Mikkelsen, T. S. et al. Dissecting direct reprogramming through integrative genomic analysis. Nature 454, 49–55 (2008)
    Article ADS CAS Google Scholar
  31. De Santa, F. et al. The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell 130, 1083–1094 (2007)
    Article CAS Google Scholar
  32. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009)
    Article Google Scholar
  33. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008)
    Article Google Scholar
  34. Robinson, J. T. et al. Integrative genomics viewer. Nature Biotechnol. 29, 24–26 (2011)
    Article CAS Google Scholar
  35. He, H. H. et al. Nucleosome dynamics define transcriptional enhancers. Nature Genet. 42, 343–347 (2010)
    Article CAS Google Scholar

Download references

Acknowledgements

J.H.H. is supported by a generous gift from Ilana and Pascal Mantoux, The Sir Charles Clore Research Prize, an ERC starting investigator grant (StG-2011-281906), the Israel Science Foundation Regular and Bikura research grants, the ICRF Foundation, Fritz Thyssen Stiftung, the Alon Foundation scholar award, a grant from E. A. and R. Drake, and the Leona M. and Harry B. Helmsley Charitable Trust. N.N. and A.P. are supported by Weizmann Dean fellowship awards. We thank A. Smith, H. Niwa and M. Saitou for reagents. We thank the Weizmann Institute management for providing critical financial and infrastructural support.

Author information

Author notes

  1. Abed AlFatah Mansour, Ohad Gafni and Noa Novershtern: These authors contributed equally to this work.

Authors and Affiliations

  1. The Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel,
    Abed AlFatah Mansour, Ohad Gafni, Leehee Weinberger, Asaf Zviran, Yoach Rais, Vladislav Krupalnik, Mirie Zerbib, Itay Maza, Shay Geula, Sergey Viukov, Liad Holtzman, Ariel Pribluda, Noa Novershtern & Jacob H. Hanna
  2. The Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel,
    Muneef Ayyash
  3. The Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel,
    Daniela Amann-Zalcenstein & Ido Amit
  4. The Department of Biological Services—Genomics Unit, Weizmann Institute of Science, Rehovot 76100, Israel,
    Daniela Amann-Zalcenstein & Shirley Horn-Saban
  5. The Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel,
    Eli Canaani

Authors

  1. Abed AlFatah Mansour
    You can also search for this author inPubMed Google Scholar
  2. Ohad Gafni
    You can also search for this author inPubMed Google Scholar
  3. Leehee Weinberger
    You can also search for this author inPubMed Google Scholar
  4. Asaf Zviran
    You can also search for this author inPubMed Google Scholar
  5. Muneef Ayyash
    You can also search for this author inPubMed Google Scholar
  6. Yoach Rais
    You can also search for this author inPubMed Google Scholar
  7. Vladislav Krupalnik
    You can also search for this author inPubMed Google Scholar
  8. Mirie Zerbib
    You can also search for this author inPubMed Google Scholar
  9. Daniela Amann-Zalcenstein
    You can also search for this author inPubMed Google Scholar
  10. Itay Maza
    You can also search for this author inPubMed Google Scholar
  11. Shay Geula
    You can also search for this author inPubMed Google Scholar
  12. Sergey Viukov
    You can also search for this author inPubMed Google Scholar
  13. Liad Holtzman
    You can also search for this author inPubMed Google Scholar
  14. Ariel Pribluda
    You can also search for this author inPubMed Google Scholar
  15. Eli Canaani
    You can also search for this author inPubMed Google Scholar
  16. Shirley Horn-Saban
    You can also search for this author inPubMed Google Scholar
  17. Ido Amit
    You can also search for this author inPubMed Google Scholar
  18. Noa Novershtern
    You can also search for this author inPubMed Google Scholar
  19. Jacob H. Hanna
    You can also search for this author inPubMed Google Scholar

Contributions

O.G., A.A.M., N.N. and J.H.H. concieved the idea for this project, designed and conducted experiments and wrote the manuscript. L.W. conducted PGC differentiation. A.Z. conducted computational simulations. N.N. conducted bioinformatics analysis. O.G., I.A., D.A.-Z., S.H.-S. and E.C. conducted genomic and chromatin immunoprecipiation experiments. A.A.M., A.P., L.H. and I.M. conducted teratoma and embryo analysis. O.G. and S.V. generated knockout cells. J.H.H., O.G., V.K. and Y.R. conducted reprogramming experiments. M.Z. conducted microinjections. S.G. conducted pre-implantation analysis. M.A. provided critical technical advice.

Corresponding authors

Correspondence toNoa Novershtern or Jacob H. Hanna.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This fie contains Supplementary Figures 1-21 and legends for Supplementary Tables 1-5 (see separate zipped file for tables). (PDF 4398 kb)

Supplementary Tables

This file contains Supplementary Tables 1-5 (see Supplementary Information file for legends). The original incorrect zipped file was replaced on 30 July 2012. (ZIP 2005 kb)

PowerPoint slides

Rights and permissions

About this article

Cite this article

Mansour, A., Gafni, O., Weinberger, L. et al. The H3K27 demethylase Utx regulates somatic and germ cell epigenetic reprogramming.Nature 488, 409–413 (2012). https://doi.org/10.1038/nature11272

Download citation

This article is cited by

Editorial Summary

Epigenetic stem-cell programming

Epigenetic changes are a key feature of cell reprogramming, but little is known about the factors responsible for chromatin or DNA modifications during this process. Jacob Hanna and colleagues report that histone H3K27 demethylase Utx is a critical regulator of the initiation of somatic-cell reprogramming. Cells lacking Utx cannot be reprogrammed by known transcription-factor cocktails, and Utx-deficient cells fail to contribute to germline transmission in mouse chimaeras. Therefore, Utx is necessary for both somatic and germ-cell reprogramming, in vitro and in vivo. This work highlights a connection between mechanisms of in vitro production of induced pluripotent stem cells and the regulation of early germline development.