Abstract

CHARGE syndrome is a multiple anomaly disorder in which patients present with a variety of phenotypes, including ocular coloboma, heart defects, choanal atresia, retarded growth and development, genitourinary hypoplasia and ear abnormalities1. Despite 70–90% of CHARGE syndrome cases resulting from mutations in the gene CHD7, which encodes an ATP-dependent chromatin remodeller, the pathways underlying the diverse phenotypes remain poorly understood2. Surprisingly, our studies of a knock-in mutant mouse strain that expresses a stabilized and transcriptionally dead variant of the tumour-suppressor protein p53 (p5325,26,53,54)3, along with a wild-type allele of p53 (also known as Trp53), revealed late-gestational embryonic lethality associated with a host of phenotypes that are characteristic of CHARGE syndrome, including coloboma, inner and outer ear malformations, heart outflow tract defects and craniofacial defects. We found that the p5325,26,53,54 mutant protein stabilized and hyperactivated wild-type p53, which then inappropriately induced its target genes and triggered cell-cycle arrest or apoptosis during development. Importantly, these phenotypes were only observed with a wild-type p53 allele, as p5325,26,53,54/− embryos were fully viable. Furthermore, we found that CHD7 can bind to the p53 promoter, thereby negatively regulating p53 expression, and that CHD7 loss in mouse neural crest cells or samples from patients with CHARGE syndrome results in p53 activation. Strikingly, we found that p53 heterozygosity partially rescued the phenotypes in Chd7-null mouse embryos, demonstrating that p53 contributes to the phenotypes that result from CHD7 loss. Thus, inappropriate p53 activation during development can promote CHARGE phenotypes, supporting the idea that p53 has a critical role in developmental syndromes and providing important insight into the mechanisms underlying CHARGE syndrome.

Access optionsAccess options

Rent or Buy article

Get time limited or full article access on ReadCube.

from$8.99

All prices are NET prices.

References

  1. 1.

    , & The spectrum of clinical features in CHARGE syndrome. Clin. Genet. 29, 298–310 (1986)

  2. 2.

    et al. CHARGE syndrome: the phenotypic spectrum of mutations in the CHD7 gene. J. Med. Genet. 43, 306–314 (2006)

  3. 3.

    et al. Distinct p53 transcriptional programs dictate acute DNA-damage responses and tumor suppression. Cell 145, 571–583 (2011)

  4. 4.

    et al. Keeping p53 in check: essential and synergistic functions of Mdm2 and Mdm4. Cell Death Differ. 13, 927–934 (2006)

  5. 5.

    , , & The p53QS transactivation-deficient mutant shows stress-specific apoptotic activity and induces embryonic lethality. Nature Genet. 37, 145–152 (2005)

  6. 6.

    , , & Quantitative analysis of limb anomalies in CHARGE syndrome: correlation with diagnosis and characteristic CHARGE anomalies. Am. J. Med. Genet. A. 123A, 111–121 (2003)

  7. 7.

    , , & Molecular and phenotypic aspects of CHD7 mutation in CHARGE syndrome. Am. J. Med. Genet. A. 152A, 674–686 (2010)

  8. 8.

    et al. Spectrum of CHD7 mutations in 110 individuals with CHARGE syndrome and genotype–phenotype correlation. Am. J. Hum. Genet. 78, 303–314 (2006)

  9. 9.

    , , , & Genitourinary anomalies in the CHARGE association. J. Urol. 161, 622–625 (1999)

  10. 10.

    et al. Successful cord blood transplantation for a CHARGE syndrome with CHD7 mutation showing DiGeorge sequence including hypoparathyroidism. Eur. J. Pediatr. 169, 839–844 (2010)

  11. 11.

    et al. The cardiac phenotype in patients with a CHD7 mutation. Circ. Cardiovasc. Genet. 6, 248–254 (2013)

  12. 12.

    et al. p53 coordinates cranial neural crest cell growth and epithelial–mesenchymal transition/delamination processes. Development 138, 1827–1838 (2011)

  13. 13.

    , , & High-frequency developmental abnormalities in p53-deficient mice. Curr. Biol. 5, 931–936 (1995)

  14. 14.

    et al. Osteoblast differentiation and skeletal development are regulated by Mdm2–p53 signaling. J. Cell Biol. 172, 909–921 (2006)

  15. 15.

    et al. Phenotypic spectrum of CHARGE syndrome in fetuses with CHD7 truncating mutations correlates with expression during human development. J. Med. Genet. 43, 211–217 (2006)

  16. 16.

    et al. Antenatal spectrum of CHARGE syndrome in 40 fetuses with CHD7 mutations. J. Med. Genet. 49, 698–707 (2012)

  17. 17.

    & Algorithm for prediction of tumour suppressor p53 affinity for binding sites in DNA. Nucleic Acids Res. 36, 1589–1598 (2008)

  18. 18.

    et al. CHD7 cooperates with PBAF to control multipotent neural crest formation. Nature 463, 958–962 (2010)

  19. 19.

    et al. CHD7 targets active gene enhancer elements to modulate ES cell-specific gene expression. PLoS Genet. 6, e1001023 (2010)

  20. 20.

    et al. Multiple mutations in mouse Chd7 provide models for CHARGE syndrome. Hum. Mol. Genet. 14, 3463–3476 (2005)

  21. 21.

    et al. Loss of Chd7 function in gene-trapped reporter mice is embryonic lethal and associated with severe defects in multiple developing tissues. Mamm. Genome 18, 94–104 (2007)

  22. 22.

    et al. Defects in vestibular sensory epithelia and innervation in mice with loss of Chd7 function: implications for human CHARGE syndrome. J. Comp. Neurol. 504, 519–532 (2007)

  23. 23.

    et al. Absent semicircular canals in CHARGE syndrome: radiologic spectrum of findings. AJNR Am. J. Neuroradiol. 27, 1663–1671 (2006)

  24. 24.

    , & Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature 378, 203–206 (1995)

  25. 25.

    et al. mdm2 is critical for inhibition of p53 during lymphopoiesis and the response to ionizing irradiation. Mol. Cell. Biol. 23, 462–472 (2003)

  26. 26.

    et al. Tumor suppression and normal aging in mice with constitutively high p53 activity. Genes Dev. 20, 16–21 (2006)

  27. 27.

    et al. p53 mutant mice that display early ageing-associated phenotypes. Nature 415, 45–53 (2002)

  28. 28.

    et al. Knockdown of fbxl10/kdm2bb rescues chd7 morphant phenotype in a zebrafish model of CHARGE syndrome. Dev. Biol. 382, 57–69 (2013)

  29. 29.

    et al. More clinical overlap between 22q11.2 deletion syndrome and CHARGE syndrome than often anticipated. Mol. Syndromol. 4, 235–245 (2013)

  30. 30.

    et al. Prevention of the neurocristopathy Treacher Collins syndrome through inhibition of p53 function. Nature Med.14. 125–133 (2008)

  31. 31.

    , & A cre-transgenic mouse strain for the ubiquitous deletion of loxP-flanked gene segments including deletion in germ cells. Nucleic Acids Res. 23, 5080–5081 (1995)

  32. 32.

    & Analysis of embryonic vascular morphogenesis. Methods Mol. Biol. 137, 223–233 (2000)

  33. 33.

    Alcian blue/alizarin red staining of cartilage and bone in mouse. Cold Spring Harb. Protoc. (March 2009)

  34. 34.

    , , , & The ATP-dependent chromatin remodeling enzyme CHD7 regulates pro-neural gene expression and neurogenesis in the inner ear. Development 137, 3139–3150 (2010)

  35. 35.

    et al. Epitope analysis of the murine p53 tumour suppressor protein. Oncogene 12, 2461–2466 (1996)

  36. 36.

    et al. Epigenomic annotation of enhancers predicts transcriptional regulators of human neural crest. Cell Stem Cell 11, 633–648 (2012)

  37. 37.

    et al. Global genomic profiling reveals an extensive p53-regulated autophagy program contributing to key p53 responses. Genes Dev. 27, 1016–1031 (2013)

Download references

Acknowledgements

We thank S. Spano-Mello, K. T. Bieging, N. Raj and M. Monje-Deisseroth for reading the manuscript and S. E. Artandi and T. Williams for discussion. We thank H. Chou for immunohistochemistry assistance; E. L. Van Nostrand, P. Lavori, and A. McMillian for statistical analysis; K. Weinberg and D. Min for thymus analysis assistance; M. Shkreli for kidney analysis assistance; B. Liu and J. A. Helms for craniofacial analysis assistance; and M. Bowen for Chd7 mouse experiment assistance. We thank S. E. Artandi for plasmids; S. E. Artandi and P. Khavari for control human fibroblast cell lines; D. Lane and B. Vojtesek for wild-type p53-specific antibody (pAB242); P. Scacheri for wild-type and Chd7-null mouse embryonic stem cells; and T. Denecker and G. Goudefroye for TP53 sequencing in patients. This work was supported by funding from the NSF and NCI (grant number 1F31CA167917-01) to J.L.V.N.; from the NIH (RO1 GM095555) to J.W.; from the American Heart Association (12EIA8960018), March of Dimes Foundation (#6-FY11-260) and NIH (R01 HL118087 and RO1 HL121197) to C.-P.C.; from the NIH (R01 DC009410) to D.M.M.; and from the ACS, LLS and NIH (RO1 CA140875) to L.D.A.

Author information

Author notes

    • Colleen A. Brady
    • , Margaret M. Kozak
    •  & Thomas M. Johnson

    Present addresses: Cardiovascular Research Center and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA (C.A.B.); Department of Medicine, University of Central Florida, Orlando, Florida 32827, USA (M.M.K.); Department of Emergency Medicine, Oregon Health and Science University, Portland, Oregon 97239, USA (T.M.J.).

Affiliations

  1. Department of Radiation Oncology, Division of Radiation and Cancer Biology, Stanford University School of Medicine, Stanford, California 94305, USA

    • Jeanine L. Van Nostrand
    • , Colleen A. Brady
    • , Heiyoun Jung
    • , Margaret M. Kozak
    • , Thomas M. Johnson
    •  & Laura D. Attardi
  2. Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California 94305, USA

    • Daniel R. Fuentes
    •  & Joanna Wysocka
  3. Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA

    • Chieh-Yu Lin
    •  & Hannes Vogel
  4. Department of Developmental Biology, Stanford University School of Medicine, Stanford, California 94305, USA

    • Chien-Jung Lin
    •  & Joanna Wysocka
  5. Department of Otolaryngology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA

    • Donald L. Swiderski
  6. Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA

    • Jonathan A. Bernstein
  7. Département de Génétique, Hôpital Necker-Enfants Malades, APHP, 75015 Paris, France

    • Tania Attié-Bitach
  8. Unite INSERM U1163, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, 75015 Paris, France

    • Tania Attié-Bitach
  9. Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA

    • Ching-Pin Chang
  10. Department of Pediatrics, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA

    • Donna M. Martin
  11. Department of Human Genetics, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA

    • Donna M. Martin
  12. Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA

    • Laura D. Attardi

Authors

  1. Search for Jeanine L. Van Nostrand in:

  2. Search for Colleen A. Brady in:

  3. Search for Heiyoun Jung in:

  4. Search for Daniel R. Fuentes in:

  5. Search for Margaret M. Kozak in:

  6. Search for Thomas M. Johnson in:

  7. Search for Chieh-Yu Lin in:

  8. Search for Chien-Jung Lin in:

  9. Search for Donald L. Swiderski in:

  10. Search for Hannes Vogel in:

  11. Search for Jonathan A. Bernstein in:

  12. Search for Tania Attié-Bitach in:

  13. Search for Ching-Pin Chang in:

  14. Search for Joanna Wysocka in:

  15. Search for Donna M. Martin in:

  16. Search for Laura D. Attardi in:

Contributions

J.L.V.N. designed and carried out experiments, interpreted data and wrote the manuscript. C.A.B. generated the p5325,26,53,54 mice, designed and carried out experiments, and interpreted data. H.J. performed p53 ChIP analyses. M.M.K. and T.M.J. performed certain mouse analyses. D.R.F. and J.W. performed NCC differentiation and CHD7 ChIP analyses. C-Y.L., C-J.L. and C-P.C. assisted with heart analyses. D.L.S. and D.M.M. performed inner ear analyses, interpreted data and provided human fibroblasts. H.V. assisted with histological analyses. J.A.B. generated CHARGE and control human fibroblast lines. T.A.-B. performed TP53 sequencing analysis in patients and supplied CHARGE thymus samples. L.D.A. designed experiments, interpreted data and wrote the manuscript.

Competing interests

The authors declare no competing financial interests.

Corresponding author

Correspondence to Laura D. Attardi.

Extended data

Supplementary information

PDF files

  1. 1.

    Supplementary Information

    This file contains Supplementary Tables 1-2.

About this article

Publication history

Received

Accepted

Published

DOI

https://doi.org/10.1038/nature13585

Further reading

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.