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:

Baf60c is essential for function of BAF chromatin remodelling complexes in heart development

Abstract

Tissue-specific transcription factors regulate several important aspects of embryonic development. They must function in the context of DNA assembled into the higher-order structure of chromatin. Enzymatic complexes such as the Swi/Snf-like BAF complexes remodel chromatin to allow the transcriptional machinery access to gene regulatory elements1,2. Here we show that Smarcd3, encoding Baf60c, a subunit of the BAF complexes, is expressed specifically in the heart and somites in the early mouse embryo. Smarcd3 silencing by RNA interference in mouse embryos derived from embryonic stem cells causes defects in heart morphogenesis that reflect impaired expansion of the anterior/secondary heart field, and also results in abnormal cardiac and skeletal muscle differentiation. An intermediate reduction in Smarcd3 expression leads to defects in outflow tract remodelling reminiscent of human congenital heart defects. Baf60c overexpressed in cell culture can mediate interactions between cardiac transcription factors and the BAF complex ATPase Brg1, thereby potentiating the activation of target genes. These results reveal tissue-specific and dose-dependent roles for Baf60c in recruiting BAF chromatin remodelling complexes to heart-specific enhancers, providing a novel mechanism to ensure transcriptional regulation during organogenesis.

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: Baf60c is required for heart formation.
Figure 2: Cardiac gene expression in wild-type (WT) and Smarcd3 knockdown (siSmarcd3) embryos.
Figure 3: Transgenic rescue of heart defects in siSmarcd3 embryos.
Figure 4: Baf60c potentiates transcription by promoting interactions between transcription factors and the BAF complex.
Figure 5: Dosage-sensitive role of Baf60c in outflow tract septation.

Similar content being viewed by others

References

  1. Narlikar, G. J., Fan, H. Y. & Kingston, R. E. Cooperation between complexes that regulate chromatin structure and transcription. Cell 108, 475–487 (2002)

    Article  CAS  PubMed  Google Scholar 

  2. Martens, J. A. & Winston, F. Recent advances in understanding chromatin remodeling by Swi/Snf complexes. Curr. Opin. Genet. Dev. 13, 136–142 (2003)

    Article  CAS  PubMed  Google Scholar 

  3. Wang, W. et al. Diversity and specialization of mammalian SWI/SNF complexes. Genes Dev. 10, 2117–2130 (1996)

    Article  CAS  PubMed  Google Scholar 

  4. Debril, M. B. et al. Transcription factors and nuclear receptors interact with the SWI/SNF complex through the BAF60c subunit. J. Biol. Chem. 279, 16677–16686 (2004)

    Article  CAS  PubMed  Google Scholar 

  5. Kunath, T. et al. Transgenic RNA interference in ES cell-derived embryos recapitulates a genetic null phenotype. Nature Biotechnol. 21, 559–561 (2003)

    Article  CAS  Google Scholar 

  6. Cai, C. L. et al. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev. Cell 5, 877–889 (2003)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. von Both, I. et al. FoxH1 is essential for development of the anterior heart field. Dev. Cell 7, 331–345 (2004)

    Article  CAS  PubMed  Google Scholar 

  8. Srivastava, D. et al. Regulation of cardiac mesodermal and neural crest development by the bHLH transcription factor, dHAND. Nature Genet. 16, 154–160 (1997)

    Article  CAS  PubMed  Google Scholar 

  9. Meilhac, S. M. et al. A retrospective clonal analysis of the myocardium reveals two phases of clonal growth in the developing mouse heart. Development 130, 3877–3889 (2003)

    Article  CAS  PubMed  Google Scholar 

  10. Chen, H. et al. BMP10 is essential for maintaining cardiac growth during murine cardiogenesis. Development 131, 2219–2231 (2004)

    Article  CAS  PubMed  Google Scholar 

  11. Meilhac, S. M., Esner, M., Kelly, R. G., Nicolas, J. F. & Buckingham, M. E. The clonal origin of myocardial cells in different regions of the embryonic mouse heart. Dev. Cell 6, 685–698 (2004)

    Article  CAS  PubMed  Google Scholar 

  12. Buckingham, M. Skeletal muscle formation in vertebrates. Curr. Opin. Genet. Dev. 11, 440–448 (2001)

    Article  CAS  PubMed  Google Scholar 

  13. de la Serna, I. L., Carlson, K. A. & Imbalzano, A. N. Mammalian SWI/SNF complexes promote MyoD-mediated muscle differentiation. Nature Genet. 27, 187–190 (2001)

    Article  CAS  PubMed  Google Scholar 

  14. Roy, K., de la Serna, I. L. & Imbalzano, A. N. The myogenic basic helix-loop-helix family of transcription factors shows similar requirements for SWI/SNF chromatin remodeling enzymes during muscle differentiation in culture. J. Biol. Chem. 277, 33818–33824 (2002)

    Article  CAS  PubMed  Google Scholar 

  15. Biben, C., Hadchouel, J., Tajbakhsh, S. & Buckingham, M. Developmental and tissue-specific regulation of the murine cardiac actin gene in vivo depends on distinct skeletal and cardiac muscle- specific enhancer elements in addition to the proximal promoter. Dev. Biol. 173, 200–212 (1996)

    Article  CAS  PubMed  Google Scholar 

  16. Toyoda, M. et al. jumonji downregulates cardiac cell proliferation by repressing cyclin D1 expression. Dev. Cell 5, 85–97 (2003)

    Article  CAS  PubMed  Google Scholar 

  17. Durocher, D., Charron, F., Warren, R., Schwartz, R. J. & Nemer, M. The cardiac transcription factors Nkx2-5 and GATA-4 are mutual cofactors. EMBO J. 16, 5687–5696 (1997)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bruneau, B. G. et al. A murine model of Holt–Oram syndrome defines roles of the T-box transcription factor Tbx5 in cardiogenesis and disease. Cell 106, 709–721 (2001)

    Article  CAS  PubMed  Google Scholar 

  19. Garg, V. et al. GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5. Nature 424, 443–447 (2003)

    Article  ADS  CAS  PubMed  Google Scholar 

  20. Kioussi, C. et al. Identification of a Wnt/Dvl/β-catenin → Pitx2 pathway mediating cell-type-specific proliferation during development. Cell 111, 673–685 (2002)

    Article  CAS  PubMed  Google Scholar 

  21. Brown, C. O. et al. The cardiac determination factor, Nkx2-5, is activated by mutual cofactors GATA-4 and Smad1/4 via a novel upstream enhancer. J. Biol. Chem. 279, 10659–10669 (2004)

    Article  CAS  PubMed  Google Scholar 

  22. Kadam, S. & Emerson, B. M. Transcriptional specificity of human SWI/SNF BRG1 and BRM chromatin remodeling complexes. Mol. Cell 11, 377–389 (2003)

    Article  CAS  PubMed  Google Scholar 

  23. Barker, N. et al. The chromatin remodelling factor Brg-1 interacts with β-catenin to promote target gene activation. EMBO J. 20, 4935–4943 (2001)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Derynck, R. & Zhang, Y. E. Smad-dependent and Smad-independent pathways in TGF-β family signalling. Nature 425, 577–584 (2003)

    Article  ADS  CAS  PubMed  Google Scholar 

  25. Lyons, I. et al. Myogenic and morphogenetic defects in the heart tubes of murine embryos lacking the homeo box gene Nkx2-5. Genes Dev. 9, 1654–1666 (1995)

    Article  CAS  PubMed  Google Scholar 

  26. Tanaka, M., Chen, Z., Bartunkova, M., Yamazaki, N. & Izumo, S. The cardiac homeobox gene Csx/Nkx2.5 lies genetically upstream of multiple genes essential for heart development. Development 126, 1269–1280 (1999)

    CAS  PubMed  Google Scholar 

  27. Hemann, M. T. et al. An epi-allelic series of p53 hypomorphs created by stable RNAi produces distinct tumor phenotypes in vivo. Nature Genet. 33, 396–400 (2003)

    Article  CAS  PubMed  Google Scholar 

  28. Liu, W. et al. Bmp4 signaling is required for outflow-tract septation and branchial-arch artery remodeling. Proc. Natl Acad. Sci. USA 101, 4489–4494 (2004)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sharpe, J. et al. Optical projection tomography as a tool for 3D microscopy and gene expression studies. Science 296, 541–545 (2002)

    Article  ADS  CAS  PubMed  Google Scholar 

  30. Zhou, Y. Q. et al. Applications for multifrequency ultrasound biomicroscopy in mice from implantation to adulthood. Physiol. Genomics 10, 113–126 (2002)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Sharpe for providing a prototype OPT system, S. McMaster for tetraploid aggregations, D. Holmyard for scanning electron microscopy, K. Harpal and K. Koshiba-Takeuchi for histology, H. Hamada and C. Meno for the unpublished Pitx2 reporter, and A. Nagafuchi, M. Nemer, R. Schwartz, D. Srivastava and T. Takeuchi for reporter and expression constructs. This research was funded by the Canadian Institutes of Health Research (CIHR; to B.G.B., J.R. and J.L.W.), the Heart and Stroke Foundation of Ontario (to B.G.B.), the March of Dimes Birth Defects Foundation (to B.G.B.), NCIC (to J.L.W.), the Canada Foundation for Innovation (to R.M.H.), an Emmy–Noether fellowship of the Deutsche Forschungsgemeinschaft (to H.L.), a Human Frontiers Science Program long-term fellowship (to J.K.T.), an OGSST fellowship (to J.R.W.), and the Koeln Fortune Program, University of Cologne (to I.v.B.).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Janet Rossant or Benoit G. Bruneau.

Ethics declarations

Competing interests

The authors declare that they have no competing financial interests.

Supplementary information

Supplementary Figure 1

Smarcd3 gene expression at E8.5 and E10.5, showing expression in myocardium, high levels of expression in the outflow and inflow tracts, and expression in the myotome and limb bud mesenchyme. (JPG 142 kb)

Supplementary Figure 2

Expression of Smarcd1-3 in mouse development at E7.5, E8.75, and E9.25, showing lack of expression of Smarcd1 or Smarcd2 in the heart. (JPG 140 kb)

Supplementary Figure 3

Smarcd3 siRNA sequences , quantitation of Smarcd3 levels in transgenic ES cells, and protein levels of Baf60c and other Baf proteins. (JPG 176 kb)

Supplementary Figure 4

Normal Tbx5, Isl1, Fgf10, Gata4, and Nkx2-5 gene expression and abnormal Tbx20 and Irx3 gene expression in Smarcd3 knockdown embryos. (JPG 254 kb)

Supplementary Figure 5

Defective skeletal muscle differentiation in Smarcd3 knockdown embryos. (JPG 116 kb)

Supplementary Table 1

Cardiac function of Smarcd3 intermediate knockdown (siSMarcd3-A#2) and wild-type mouse embryos at E11.5. (DOC 22 kb)

Supplementary Movie

Rendered OPT of a wild-type (left) and Smarcd3 knockdown (right) embryos at E9.5. The heart is labeled red, and the rest of the embryo is translucent white. (MOV 5228 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lickert, H., Takeuchi, J., von Both, I. et al. Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature 432, 107–112 (2004). https://doi.org/10.1038/nature03071

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature03071

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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