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:

Genome-scale epigenetic reprogramming during epithelial-to-mesenchymal transition

Abstract

Epithelial-to-mesenchymal transition (EMT) is an extreme example of cell plasticity that is important for normal development, injury repair and malignant progression. Widespread epigenetic reprogramming occurs during stem cell differentiation and malignant transformation, but EMT-related epigenetic reprogramming is poorly understood. Here we investigated epigenetic modifications during EMT mediated by transforming growth factor beta. Although DNA methylation was unchanged during EMT, we found a global reduction in the heterochromatin mark H3 Lys9 dimethylation (H3K9Me2), an increase in the euchromatin mark H3 Lys4 trimethylation (H3K4Me3) and an increase in the transcriptional mark H3 Lys36 trimethylation (H3K36Me3). These changes depended largely on lysine-specific demethylase-1 (Lsd1), and loss of Lsd1 function had marked effects on EMT-driven cell migration and chemoresistance. Genome-scale mapping showed that chromatin changes were mainly specific to large organized heterochromatin K9 modifications (LOCKs), which suggests that EMT is characterized by reprogramming of specific chromatin domains across the genome.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Global changes in bulk chromatin modifications during EMT.
Figure 2: Lsd1 regulates bulk changes in chromatin modifications during EMT.
Figure 3: Lsd1 regulates certain EMT phenotypes.
Figure 4: ChIP-chip analysis of EMT reveals alterations of chromatin in LOCK domains.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Kalluri, R. & Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Invest. 119, 1420–1428 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Thiery, J.P., Acloque, H., Huang, R.Y.J. & Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 139, 871–890 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Mani, S.A. et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gupta, P.B. et al. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell 138, 645–659 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Feinberg, A.P. Phenotypic plasticity and the epigenetics of human disease. Nature 447, 433–440 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Jenuwein, T. & Allis, C.D. Translating the histone code. Science 293, 1074–1080 (2001).

    CAS  PubMed  Google Scholar 

  7. Hawkins, R.D. et al. Distinct epigenomic landscapes of pluripotent and lineage-committed human cells. Cell Stem Cell 6, 479–491 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jones, P.A. & Baylin, S.B. The epigenomics of cancer. Cell 128, 683–692 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Roussos, E.T. et al. AACR Special Conference on Epithelial-Mesenchymal Transition and Cancer Progression and Treatment. Cancer Res. 70, 7360–7364 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Rahimi, R.A. & Leof, E.B. TGF-β signaling: a tale of two responses. J. Cell. Biochem. 102, 593–608 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Xu, J., Lamouille, S. & Derynck, R. TGF-β-induced epithelial to mesenchymal transition. Cell Res. 19, 156–172 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Dooley, S. et al. Hepatocyte-specific Smad7 expression attenuates TGF-β-mediated fibrogenesis and protects against liver damage. Gastroenterology 135, 642–659 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Yang, Y., Pan, X., Lei, W., Wang, J. & Song, J. Transforming growth factor-beta1 induces epithelial-to-mesenchymal transition and apoptosis via a cell cycle-dependent mechanism. Oncogene 25, 7235–7244 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Kaimori, A. et al. Transforming growth factor-β induces an epithelial-to-mesenchymal transition state in mouse hepatocytes in vitro. J. Biol. Chem. 282, 22089–22101 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Pan, X. et al. Nitric oxide suppresses transforming growth factor-β1–induced epithelial-to-mesenchymal transition and apoptosis in mouse hepatocytes. Hepatology 50, 1577–1587 (2009).

    Article  CAS  PubMed  Google Scholar 

  16. Wu, J.C., Merlino, G. & Fausto, N. Establishment and characterization of differentiated, nontransformed hepatocyte cell lines derived from mice transgenic for transforming growth factor alpha. Proc. Natl. Acad. Sci. USA 91, 674–678 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mills, S.E. et al. Sternbergs Diagnostic Surgical Pathology (Lippincott Williams & Wilkins, Philadelphia, 2004).

  18. Dumont, N. et al. Sustained induction of epithelial to mesenchymal transition activates DNA methylation of genes silenced in basal-like breast cancers. Proc. Natl. Acad. Sci. USA 105, 14867–14872 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Irizarry, R.A. et al. Comprehensive high-throughput arrays for relative methylation (CHARM). Genome Res. 18, 780–790 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Irizarry, R.A. et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat. Genet. 41, 178–186 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tachibana, M. et al. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 16, 1779–1791 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rice, J.C. et al. Histone methyltransferases direct different degrees of methylation to define distinct chromatin domains. Mol. Cell 12, 1591–1598 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Peters, A.H.F.M. et al. Partitioning and plasticity of repressive histone methylation states in mammalian chromatin. Mol. Cell 12, 1577–1589 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Wen, B., Wu, H., Shinkai, Y., Irizarry, R.A. & Feinberg, A.P. Large histone H3 lysine 9 dimethylated chromatin blocks distinguish differentiated from embryonic stem cells. Nat. Genet. 41, 246–250 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chen, E.S. et al. Cell cycle control of centromeric repeat transcription and heterochromatin assembly. Nature 451, 734–737 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Kizer, K.O. et al. A novel domain in Set2 mediates RNA polymerase II interaction and couples histone H3 K36 methylation with transcript elongation. Mol. Cell. Biol. 25, 3305–3316 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mikkelsen, T.S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Onder, T.T. et al. Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 68, 3645–3654 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Zheng, G. et al. Disruption of E-cadherin by matrix metalloproteinase directly mediates epithelial-mesenchymal transition downstream of transforming growth factor-β in renal tubular epithelial cells. Am. J. Pathol. 175, 580–591 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shi, Y. et al. Histone demethylation mediated by the nuclear amine oxidase homolog Lsd1. Cell 119, 941–953 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Metzger, E. et al. Lsd1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 437, 436–439 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Wang, Y. et al. Lsd1 is a subunit of the NuRD complex and targets the metastasis programs in breast cancer. Cell 138, 660–672 (2009).

    Article  CAS  PubMed  Google Scholar 

  34. Shi, Y.-J. Regulation of Lsd1 histone demethylase activity by its associated factors. Mol. Cell 19, 857–864 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Wang, J. et al. Opposing Lsd1 complexes function in developmental gene activation and repression programmes. Nature 446, 882–887 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Liu, Y., Subrahmanyam, R., Chakraborty, T., Sen, R. & Desiderio, S. A plant homeodomain in Rag-2 that binds hypermethylated lysine 4 of histone H3 is necessary for efficient antigen-receptor-gene rearrangement. Immunity 27, 561–571 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Matthews, A.G.W. et al. RAG2 PHD finger couples histone H3 lysine 4 trimethylation with V(D)J recombination. Nature 450, 1106–1110 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Borde, V. et al. Histone H3 lysine 4 trimethylation marks meiotic recombination initiation sites. EMBO J. 28, 99–111 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Faucher, D. & Wellinger, R.J. Methylated H3K4, a transcription-associated histone modification, is involved in the DNA damage response pathway. PLoS Genet. 6, e1001082 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Guelen, L. et al. Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions. Nature 453, 948–951 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Huang, D.W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2008).

    Article  Google Scholar 

  42. Bernstein, B.E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Misteli, T. & Soutoglou, E. The emerging role of nuclear architecture in DNA repair and genome maintenance. Nat. Rev. Mol. Cell Biol. 10, 243–254 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chosed, R. & Dent, S.Y.R.A. Two-way street: Lsd1 regulates chromatin boundary formation in S. pombe and Drosophila. Mol. Cell 26, 160–162 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Sierra, J., Yoshida, T., Joazeiro, C.A. & Jones, K.A. The APC tumor suppressor counteracts β-catenin activation and H3K4 methylation at Wnt target genes. Genes Dev. 20, 586–600 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lin, T., Ponn, A., Hu, X., Law, B.K. & Lu, J. Requirement of the histone demethylase Lsd1 in Snai1-mediated transcriptional repression during epithelial-mesenchymal transition. Oncogene 29, 4896–4904 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Efroni, S. et al. Global transcription in pluripotent embryonic stem cells. Cell Stem Cell 2, 437–447 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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  PubMed  Google Scholar 

  49. 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  PubMed  Google Scholar 

  50. Lin, C. et al. Nuclear receptor-induced chromosomal proximity and DNA breaks underlie specific translocations in cancer. Cell 139, 1069–1083 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Haffner, M.C. et al. Androgen-induced TOP2B-mediated double-strand breaks and prostate cancer gene rearrangements. Nat. Genet. 42, 668–675 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ju, B.-G. et al. A topoisomerase IIβ-mediated dsDNA break required for regulated transcription. Science 312, 1798–1802 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Feng, S., Jacobsen, S.E. & Reik, W. Epigenetic reprogramming in plant and animal development. Science 330, 622–627 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Shechter, D., Dormann, H.L., Allis, C.D. & Hake, S.B. Extraction, purification and analysis of histones. Nat. Protoc. 2, 1445–1457 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Litt, M.D., Simpson, M., Recillas-Targa, F., Prioleau, M.-N. & Felsenfeld, G. Transitions in histone acetylation reveal boundaries of three separately regulated neighboring loci. EMBO J. 20, 2224–2235 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lee, M.G., Wynder, C., Norman, J. & Shiekhattar, R. Isolation and characterization of histone H3 lysine 4 demethylase-containing complexes. Methods 40, 327–330 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by US National Institutes of Health grant 5R37CA054358 to A.P.F. We thank A. Tackett for assistance with mass spectrometry experiments and data analysis; C. McCall for advice on large-scale immunoprecipitation experiments; the microscopy facility at Johns Hopkins University for electron microscopy; and K. Reddy and S. Taverna for advice.

Author information

Authors and Affiliations

Authors

Contributions

O.G.M. and A.P.F. conceived this work, designed the experiments and wrote the manuscript. H.W. performed the statistical analyses. W.T. performed the immunofluorescence. A.D. performed the CHARM.

Corresponding author

Correspondence to Andrew P Feinberg.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Tables 1–5 (PDF 2802 kb)

Supplementary Data 1

Locations of K9Me2 LOCKs identified in AML12 cells (XLSX 183 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

McDonald, O., Wu, H., Timp, W. et al. Genome-scale epigenetic reprogramming during epithelial-to-mesenchymal transition. Nat Struct Mol Biol 18, 867–874 (2011). https://doi.org/10.1038/nsmb.2084

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2084

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