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:

CHD7 cooperates with PBAF to control multipotent neural crest formation

Abstract

Heterozygous mutations in the gene encoding the CHD (chromodomain helicase DNA-binding domain) member CHD7, an ATP-dependent chromatin remodeller homologous to the Drosophila trithorax-group protein Kismet1,2, result in a complex constellation of congenital anomalies called CHARGE syndrome, which is a sporadic, autosomal dominant disorder characterized by malformations of the craniofacial structures, peripheral nervous system, ears, eyes and heart3,4. Although it was postulated 25 years ago that CHARGE syndrome results from the abnormal development of the neural crest, this hypothesis remained untested5. Here we show that, in both humans and Xenopus, CHD7 is essential for the formation of multipotent migratory neural crest (NC), a transient cell population that is ectodermal in origin but undergoes a major transcriptional reprogramming event to acquire a remarkably broad differentiation potential and ability to migrate throughout the body, giving rise to craniofacial bones and cartilages, the peripheral nervous system, pigmentation and cardiac structures6,7. We demonstrate that CHD7 is essential for activation of the NC transcriptional circuitry, including Sox9, Twist and Slug. In Xenopus embryos, knockdown of Chd7 or overexpression of its catalytically inactive form recapitulates all major features of CHARGE syndrome. In human NC cells CHD7 associates with PBAF (polybromo- and BRG1-associated factor-containing complex)8 and both remodellers occupy a NC-specific distal SOX9 enhancer9 and a conserved genomic element located upstream of the TWIST1 gene. Consistently, during embryogenesis CHD7 and PBAF cooperate to promote NC gene expression and cell migration. Our work identifies an evolutionarily conserved role for CHD7 in orchestrating NC gene expression programs, provides insights into the synergistic control of distal elements by chromatin remodellers, illuminates the patho-embryology of CHARGE syndrome, and suggests a broader function for CHD7 in the regulation of cell motility.

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: CHD7 knockdown disrupts formation of the migratory, multipotent hNCLCs.
Figure 2: Chd7 is required for expression of transcription factors critical for neural crest specification and migration.
Figure 3: CHD7 interacts with the PBAF complex.
Figure 4: CHD7 and PBAF bind distal regulatory elements upstream of NC transcription factors and synergistically regulate NC gene expression and migration in vivo.

Similar content being viewed by others

References

  1. Srinivasan, S., Dorighi, K. M. & Tamkun, J. W. Drosophila Kismet regulates histone H3 lysine 27 methylation and early elongation by RNA polymerase II. PLoS Genet. 4, e1000217 (2008)

    Article  Google Scholar 

  2. Kennison, J. A. & Tamkun, J. W. Dosage-dependent modifiers of polycomb and antennapedia mutations in Drosophila . Proc. Natl Acad. Sci. USA 85, 8136–8140 (1988)

    Article  ADS  CAS  Google Scholar 

  3. Vissers, L. E. et al. Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nature Genet. 36, 955–957 (2004)

    Article  CAS  Google Scholar 

  4. Sanlaville, D. & Verloes, A. CHARGE syndrome: an update. Eur. J. Hum. Genet. 15, 389–399 (2007)

    Article  CAS  Google Scholar 

  5. Siebert, J. R., Graham, J. M. & MacDonald, C. Pathologic features of the CHARGE association: support for involvement of the neural crest. Teratology 31, 331–336 (1985)

    Article  CAS  Google Scholar 

  6. Sauka-Spengler, T. & Bronner-Fraser, M. A gene regulatory network orchestrates neural crest formation. Nature Rev. Mol. Cell Biol. 9, 557–568 (2008)

    Article  CAS  Google Scholar 

  7. Dupin, E., Creuzet, S. & Le Douarin, N. M. The contribution of the neural crest to the vertebrate body. Adv. Exp. Med. Biol. 589, 96–119 (2006)

    Article  CAS  Google Scholar 

  8. Lemon, B., Inouye, C., King, D. S. & Tjian, R. Selectivity of chromatin-remodelling cofactors for ligand-activated transcription. Nature 414, 924–928 (2001)

    Article  ADS  CAS  Google Scholar 

  9. Bagheri-Fam, S. et al. Long-range upstream and downstream enhancers control distinct subsets of the complex spatiotemporal Sox9 expression pattern. Dev. Biol. 291, 382–397 (2006)

    Article  CAS  Google Scholar 

  10. Bajpai, R. et al. Molecular stages of rapid and uniform neuralization of human embryonic stem cells. Cell Death Differ. 16, 807–825 (2009)

    Article  CAS  Google Scholar 

  11. Eccles, M. R. & Schimmenti, L. A. Renal-coloboma syndrome: a multi-system developmental disorder caused by PAX2 mutations. Clin. Genet. 56, 1–9 (1999)

    Article  CAS  Google Scholar 

  12. Wu, J. I., Lessard, J. & Crabtree, G. R. Understanding the words of chromatin regulation. Cell 136, 200–206 (2009)

    Article  CAS  Google Scholar 

  13. Mohrmann, L. & Verrijzer, C. P. Composition and functional specificity of SWI2/SNF2 class chromatin remodeling complexes. Biochim. Biophys. Acta 1681, 59–73 (2005)

    Article  CAS  Google Scholar 

  14. Kaeser, M. D., Aslanian, A., Dong, M. Q., Yates, J. R. & Emerson, B. M. BRD7, a novel PBAF-specific SWI/SNF subunit, is required for target gene activation and repression in embryonic stem cells. J. Biol. Chem. 283, 32254–32263 (2008)

    Article  CAS  Google Scholar 

  15. Kwon, C. S. & Wagner, D. Unwinding chromatin for development and growth: a few genes at a time. Trends Genet. 23, 403–412 (2007)

    Article  CAS  Google Scholar 

  16. Schnetz, M. P. et al. Genomic distribution of CHD7 on chromatin tracks H3K4 methylation patterns. Genome Res. 19, 590–601 (2009)

    Article  CAS  Google Scholar 

  17. Ho, L. et al. An embryonic stem cell chromatin remodeling complex, esBAF, is an essential component of the core pluripotency transcriptional network. Proc. Natl Acad. Sci. USA 106, 5187–5191 (2009)

    Article  ADS  CAS  Google Scholar 

  18. Heintzman, N. D. et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459, 108–112 (2009)

    Article  ADS  CAS  Google Scholar 

  19. Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007)

    Article  CAS  Google Scholar 

  20. Loring, J. F., Wesselschmidt, R. L. & Schwartz, P. H. Human Stem Cell Manual: A Laboratory Guide (Elsevier/Academic, 2007)

    Google Scholar 

  21. Minarcik, J. C. & Golden, J. A. AP-2 and HNK-1 define distinct populations of cranial neural crest cells. Orthod. Craniofac. Res. 6, 210–219 (2003)

    Article  CAS  Google Scholar 

  22. Del Barrio, M. G. & Nieto, M. A. Relative expression of Slug, RhoB, and HNK-1 in the cranial neural crest of the early chicken embryo. Dev. Dyn. 229, 136–139 (2004)

    Article  CAS  Google Scholar 

  23. Lee, G. et al. Isolation and directed differentiation of neural crest stem cells derived from human embryonic stem cells. Nature Biotechnol. 25, 1468–1475 (2007)

    Article  CAS  Google Scholar 

  24. Stegmeier, F., Hu, G., Rickles, R., Hannon, G. & Elledge, S. A lentiviral microRNA-based system for single-copy polymerase II-regulated RNA interference in mammalian cells. Proc. Natl Acad. Sci. USA 102, 13212–13217 (2005)

    Article  ADS  CAS  Google Scholar 

  25. Shimada, T. et al. Targeted and highly efficient gene transfer into CD4+ cells by a recombinant human immunodeficiency virus retroviral vector. J. Clin. Invest. 88, 1043–1047 (1991)

    Article  CAS  Google Scholar 

  26. Zufferey, R., Nagy, D., Mandel, R., Naldini, L. & Trono, D. Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo . Nature Biotechnol. 15, 871–875 (1997)

    Article  CAS  Google Scholar 

  27. Bajpai, R., Lesperance, J., Kim, M. & Terskikh, A. V. Efficient propagation of single cells Accutase-dissociated human embryonic stem cells. Mol. Reprod. Dev. 75, 818–827 (2008)

    Article  CAS  Google Scholar 

  28. Dignam, J., Lebovitz, R. & Roeder, R. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 11, 1475–1489 (1983)

    Article  CAS  Google Scholar 

  29. Shimizu, K. & Gurdon, J. B. A quantitative analysis of signal transduction from activin receptor to nucleus and its relevance to morphogen gradient interpretation. Proc. Natl Acad. Sci. USA 96, 6791–6796 (1999)

    Article  ADS  CAS  Google Scholar 

  30. Boyer, L. A. et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 122, 947–956 (2005)

    Article  CAS  Google Scholar 

  31. Fu, Y., Sinha, M., Peterson, C. L. & Weng, Z. The insulator binding protein CTCF positions 20 nucleosomes around its binding sites across the human genome. PLoS Genet. 4, e1000138 (2008)

    Article  Google Scholar 

  32. Seo, S., Richardson, G. A. & Kroll, K. L. The SWI/SNF chromatin remodeling protein Brg1 is required for vertebrate neurogenesis and mediates transactivation of Ngn and NeuroD. Development 132, 105–115 (2005)

    Article  CAS  Google Scholar 

  33. Higuchi, R., Krummel, B. & Saiki, R. A general method of in vitro preparation and specific mutagenesis of DNA fragments: study of protein and DNA interactions. Nucleic Acids Res. 16, 7351–7367 (1988)

    Article  CAS  Google Scholar 

  34. Nieuwkoop, P. D. & Faber, J. Normal Table of Xenopus laevis (Daudin) (Garland, 1994)

    Google Scholar 

  35. Sive, H. L., Grainger, R. M. & Harland, R. M. Early Development of Xenopus laevis: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 2000)

    Google Scholar 

Download references

Acknowledgements

We thank laboratory members for input during the course of this work; S. Brugmann and A. C. Foley for sharing expertise in Xenopus and chick embryology, B. Bedogni, L. Ho and I. Shestopalov for reagents; A. Sanchez-Alvarado for the cartilage staining protocol; G. Crump and S. Cox for sharing unpublished information; and S. Brugmann, E. Duncan, Z. Ma, J. Peng, A. M. Ring and R. A. Roth for comments on the manuscript. This work was supported by CIRM SEED grant RS1-00323, a W. M. Keck Foundation Distinguished Young Scholar Award, a Searle Scholar Award to J.W., an EMBO long-term fellowship to A.R-I., National Institutes of Health (NIH) grant R01DK082664 to Y.Z., March of Dimes grant 6-FY06-335 to J.H., NIH grant R01HL085345 to C-P.C., and an Oak Foundation Fellowship to Y.X.

Author Contributions R.B. developed the in vitro model of human NC formation and designed, performed and interpreted most experiments. D.A.C. did a preliminary characterization of NC migration defects in Xenopus, performed the in situ hybridization analyses of wild-type and CHD7 morphant embryos, and analysed the craniofacial defects in hCHD7 ATPaseK998R tadpoles. A.R-I. performed the genomic analyses of CHD7-Brg1 co-occupancy and contributed ideas. J.Z. and Y.Z. performed the mass spectrometric analysis of CHD7 immunoprecipitates. Y.X. and C-P.C. characterized the heart defects in hCHD7 ATPaseK998R tadpoles. J.H. provided expertise and guidance on in ovo transplantation experiments. T.S. advised on the design and interpretation of Xenopus experiments and contributed reagents. J.W. conceived the project, contributed ideas, interpreted results and wrote the manuscript. All authors edited the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joanna Wysocka.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-19 with Legends and Supplementary Tables 1-4. (PDF 8933 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bajpai, R., Chen, D., Rada-Iglesias, A. et al. CHD7 cooperates with PBAF to control multipotent neural crest formation. Nature 463, 958–962 (2010). https://doi.org/10.1038/nature08733

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature08733

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