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.

  • Letter
  • Published:

Epigenetic repression of cardiac progenitor gene expression by Ezh2 is required for postnatal cardiac homeostasis

Abstract

Adult-onset diseases can be associated with in utero events, but mechanisms for this remain unknown1,2. The Polycomb histone methyltransferase Ezh2 stabilizes transcription by depositing repressive marks during development that persist into adulthood3,4,5,6,7,8,9, but its function in postnatal organ homeostasis is unknown. We show that Ezh2 stabilizes cardiac gene expression and prevents cardiac pathology by repressing the homeodomain transcription factor gene Six1, which functions in cardiac progenitor cells but is stably silenced upon cardiac differentiation10. Deletion of Ezh2 in cardiac progenitors caused postnatal myocardial pathology and destabilized cardiac gene expression with activation of Six1-dependent skeletal muscle genes. Six1 induced cardiomyocyte hypertrophy and skeletal muscle gene expression. Furthermore, genetically reducing Six1 levels rescued the pathology of Ezh2-deficient hearts. Thus, Ezh2-mediated repression of Six1 in differentiating cardiac progenitors is essential for stable gene expression and homeostasis in the postnatal heart. Our results suggest that epigenetic dysregulation in embryonic progenitor cells is a predisposing factor for adult disease and dysregulated stress responses.

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: Ezh2 limits cardiac growth and fibrosis.
Figure 2: Ezh2 represses expression of fetal genes, profibrosis factors and Six1.
Figure 3: Six1 is epigenetically repressed by PRC2.
Figure 4: Six1 has differentiated myocardium expression potential.
Figure 5: Six1 induces cardiac hypertrophy and skeletal muscle gene expression in cardiomyocytes.
Figure 6: Ezh2 limits cardiac hypertrophy and fibrosis by repressing Six1.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Gluckman, P.D., Hanson, M.A., Buklijas, T., Low, F.M. & Beedle, A.S. Epigenetic mechanisms that underpin metabolic and cardiovascular diseases. Nat. Rev. Endocrinol. 5, 401–408 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Petronis, A. Epigenetics as a unifying principle in the aetiology of complex traits and diseases. Nature 465, 721–727 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Surface, L.E., Thornton, S.R. & Boyer, L.A. Polycomb group proteins set the stage for early lineage commitment. Cell Stem Cell 7, 288–298 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Margueron, R. & Reinberg, D. Chromatin structure and the inheritance of epigenetic information. Nat. Rev. Genet. 11, 285–296 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Su, I.H. et al. Ezh2 controls B cell development through histone H3 methylation and Igh rearrangement. Nat. Immunol. 4, 124–131 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Pereira, J.D. et al. Ezh2, the histone methyltransferase of PRC2, regulates the balance between self-renewal and differentiation in the cerebral cortex. Proc. Natl. Acad. Sci. USA 107, 15957–15962 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Xu, C.R. et al. Chromatin “prepattern” and histone modifiers in a fate choice for liver and pancreas. Science 332, 963–966 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Juan, A.H. et al. Polycomb EZH2 controls self-renewal and safeguards the transcriptional identity of skeletal muscle stem cells. Genes Dev. 25, 789–794 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Guo, C. et al. A Tbx1-Six1/Eya1-Fgf8 genetic pathway controls mammalian cardiovascular and craniofacial morphogenesis. J. Clin. Invest. 121, 1585–1595 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Heineke, J. & Molkentin, J.D. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat. Rev. Mol. Cell Biol. 7, 589–600 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Ahmad, F., Seidman, J.G. & Seidman, C.E. The genetic basis for cardiac remodeling. Annu. Rev. Genomics Hum. Genet. 6, 185–216 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Bergmann, O. et al. Evidence for cardiomyocyte renewal in humans. Science 324, 98–102 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Margueron, R. et al. Ezh1 and Ezh2 maintain repressive chromatin through different mechanisms. Mol. Cell 32, 503–518 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shen, X. et al. EZH1 mediates methylation on histone H3 lysine 27 and complements EZH2 in maintaining stem cell identity and executing pluripotency. Mol. Cell 32, 491–502 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Verzi, M.P., McCulley, D.J., De Val, S., Dodou, E. & Black, B.L. The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field. Dev. Biol. 287, 134–145 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Weiford, B.C., Subbarao, V.D. & Mulhern, K.M. Noncompaction of the ventricular myocardium. Circulation 109, 2965–2971 (2004).

    Article  PubMed  Google Scholar 

  18. Swynghedauw, B. Phenotypic plasticity of adult myocardium: molecular mechanisms. J. Exp. Biol. 209, 2320–2327 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Houweling, A.C., van Borren, M.M., Moorman, A.F. & Christoffels, V.M. Expression and regulation of the atrial natriuretic factor encoding gene Nppa during development and disease. Cardiovasc. Res. 67, 583–593 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. McFadden, D.G. et al. A GATA-dependent right ventricular enhancer controls dHAND transcription in the developing heart. Development 127, 5331–5341 (2000).

    CAS  PubMed  Google Scholar 

  21. Takeuchi, J.K. et al. Chromatin remodeling complex dosage modulates transcription factor function in heart development. Nat. Commun. 2, 187 (2011).

    Article  PubMed  Google Scholar 

  22. Kumar, J.P. The sine oculis homeobox (SIX) family of transcription factors as regulators of development and disease. Cell. Mol. Life Sci. 66, 565–583 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, X. et al. Eya protein phosphatase activity regulates Six1-Dach-Eya transcriptional effects in mammalian organogenesis. Nature 426, 247–254 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Schönberger, J. et al. Mutation in the transcriptional coactivator EYA4 causes dilated cardiomyopathy and sensorineural hearing loss. Nat. Genet. 37, 418–422 (2005).

    Article  PubMed  Google Scholar 

  25. Liu, Y., Chu, A., Chakroun, I., Islam, U. & Blais, A. Cooperation between myogenic regulatory factors and SIX family transcription factors is important for myoblast differentiation. Nucleic Acids Res. 38, 6857–6871 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Christodoulou, D.C., Gorham, J.M., Herman, D.S. & Seidman, J.G. Construction of normalized RNA-seq libraries for next-generation sequencing using the crab duplex–specific nuclease. Curr. Protoc. Mol. Biol. 94, 4.12.1–4.12.11 (2011).

    Google Scholar 

Download references

Acknowledgements

We thank A. Blais for Six1 antiserum, T. Sukonnik for in situ hybridization, J. Wythe for mT/mG embryos, J. Wylie for technical assistance, D. Miguel-Perez for animal husbandry, A. Holloway and A. Williams (Gladstone Bioinformatics Core) for RNA-seq and microarray data analysis, L. Ta (Gladstone Genomics Core) for microarray experiments, J. Zhang (Gladstone Transgenic Core) for oocyte injection, C. Miller (Gladstone Histology Core) for histology, Bruneau lab members for helpful discussions and G. Howard for editorial assistance. This work was supported by a fellowship from the California Institute for Regenerative Medicine (P.D.-O.), US National Institutes of Health grants U01HL098179 (B.G.B.) and U01HL098166 (C.E.S. and J.G.S.), by the Lawrence J. and Florence A. DeGeorge Charitable Trust/American Heart Association Established Investigator Award (B.G.B.) and by William H. Younger, Jr. (B.G.B.). Animal work was supported by a US National Institutes of Health/National Center for Research Resources grant (C06 RR018928) to the J. David Gladstone Institutes.

Author information

Authors and Affiliations

Authors

Contributions

P.D.-O. designed the study with B.G.B. and performed most of the experiments. Y.H. performed echocardiography. X.L. provided essential information before publication. X.L. and A.T. provided mouse lines. D.C. generated and analyzed RNA-eq data with C.E.S. and J.G.S. P.D.-O. and B.G.B. wrote the paper with input from all the authors.

Corresponding author

Correspondence to Benoit G Bruneau.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1 and 4 and Supplementary Figures 1–14 (PDF 14815 kb)

Supplementary Table 2

RNA-seq data obtained from right ventricular myocardium from Ezh2-deficient hearts (XLSX 1825 kb)

Supplementary Table 3

Signaling pathways in which Six1 target genes missregulated in Ezh2-deficient hearts participate (XLSX 13 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Delgado-Olguín, P., Huang, Y., Li, X. et al. Epigenetic repression of cardiac progenitor gene expression by Ezh2 is required for postnatal cardiac homeostasis. Nat Genet 44, 343–347 (2012). https://doi.org/10.1038/ng.1068

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.1068

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