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:

Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer

Abstract

Improved sequencing technologies offer unprecedented opportunities for investigating the role of rare genetic variation in common disease. However, there are considerable challenges with respect to study design, data analysis and replication1. Using pooled next-generation sequencing of 507 genes implicated in the repair of DNA in 1,150 samples, an analytical strategy focused on protein-truncating variants (PTVs) and a large-scale sequencing case–control replication experiment in 13,642 individuals, here we show that rare PTVs in the p53-inducible protein phosphatase PPM1D are associated with predisposition to breast cancer and ovarian cancer. PPM1D PTV mutations were present in 25 out of 7,781 cases versus 1 out of 5,861 controls (P = 1.12 × 10−5), including 18 mutations in 6,912 individuals with breast cancer (P = 2.42 × 10−4) and 12 mutations in 1,121 individuals with ovarian cancer (P = 3.10 × 10−9). Notably, all of the identified PPM1D PTVs were mosaic in lymphocyte DNA and clustered within a 370-base-pair region in the final exon of the gene, carboxy-terminal to the phosphatase catalytic domain. Functional studies demonstrate that the mutations result in enhanced suppression of p53 in response to ionizing radiation exposure, suggesting that the mutant alleles encode hyperactive PPM1D isoforms. Thus, although the mutations cause premature protein truncation, they do not result in the simple loss-of-function effect typically associated with this class of variant, but instead probably have a gain-of-function effect. Our results have implications for the detection and management of breast and ovarian cancer risk. More generally, these data provide new insights into the role of rare and of mosaic genetic variants in common conditions, and the use of sequencing in their identification.

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: Clustering of cancer predisposing mutations in PPM1D.
Figure 2: PPM1D mutations are mosaic in lymphocyte DNA.
Figure 3: The effect of mutant PPM1D isoforms on p53 activation.

Similar content being viewed by others

References

  1. Cirulli, E. T. & Goldstein, D. B. Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nature Rev. Genet. 11, 415–425 (2010)

    Article  CAS  PubMed  Google Scholar 

  2. Turnbull, C. & Rahman, N. Genetic predisposition to breast cancer: past, present, and future. Annu. Rev. Genomics Hum. Genet. 9, 321–345 (2008)

    Article  CAS  PubMed  Google Scholar 

  3. Gayther, S. A. & Pharoah, P. D. The inherited genetics of ovarian and endometrial cancer. Curr. Opin. Genet. Dev. 20, 231–238 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Rahman, N. et al. PALB2, which encodes a BRCA2-interacting protein, is a breast cancer susceptibility gene. Nature Genet. 39, 165–167 (2007)

    Article  CAS  PubMed  Google Scholar 

  5. Renwick, A. et al. ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nature Genet. 38, 873–875 (2006)

    Article  CAS  PubMed  Google Scholar 

  6. Seal, S. et al. Truncating mutations in the Fanconi anemia J gene BRIP1 are low-penetrance breast cancer susceptibility alleles. Nature Genet. 38, 1239–1241 (2006)

    Article  CAS  PubMed  Google Scholar 

  7. Meijers-Heijboer, H. et al. Low-penetrance susceptibility to breast cancer due to CHEK2*1100delC in noncarriers of BRCA1 or BRCA2 mutations. Nature Genet. 31, 55–59 (2002)

    Article  CAS  PubMed  Google Scholar 

  8. Meindl, A. et al. Germline mutations in breast and ovarian cancer pedigrees establish RAD51C as a human cancer susceptibility gene. Nature Genet. 42, 410–414 (2010)

    Article  CAS  PubMed  Google Scholar 

  9. Loveday, C. et al. Germline RAD51C mutations confer susceptibility to ovarian cancer. Nature Genet. 44, 475–476 (2012)

    Article  CAS  PubMed  Google Scholar 

  10. Loveday, C. et al. Germline mutations in RAD51D confer susceptibility to ovarian cancer. Nature Genet. 43, 879–882 (2011)

    Article  CAS  PubMed  Google Scholar 

  11. Rivas, M. A. et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nature Genet. 43, 1066–1073 (2011)

    Article  CAS  PubMed  Google Scholar 

  12. Snape, K. et al. Predisposition gene identification in common cancers by exome sequencing: insights from familial breast cancer. Breast Cancer Res. Treat. 134, 429–433 (2012)

    Article  PubMed  PubMed Central  Google Scholar 

  13. Caruccio, N. Preparation of next-generation sequencing libraries using Nextera technology: simultaneous DNA fragmentation and adaptor tagging by in vitro transposition. Methods Mol. Biol. 733, 241–255 (2011)

    Article  CAS  PubMed  Google Scholar 

  14. Schouten, J. P. et al. Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification. Nucleic Acids Res. 30, e57 (2002)

    Article  PubMed  PubMed Central  Google Scholar 

  15. Fiscella, M. et al. Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc. Natl Acad. Sci. USA 94, 6048–6053 (1997)

    Article  ADS  CAS  PubMed  Google Scholar 

  16. Lu, X. et al. The type 2C phosphatase Wip1: an oncogenic regulator of tumor suppressor and DNA damage response pathways. Cancer Metastasis Rev. 27, 123–135 (2008)

    Article  PubMed  PubMed Central  Google Scholar 

  17. Lu, X., Nguyen, T. A. & Donehower, L. A. Reversal of the ATM/ATR-mediated DNA damage response by the oncogenic phosphatase PPM1D. Cell Cycle 4, 4060–4064 (2005)

    Article  Google Scholar 

  18. Shreeram, S. et al. Regulation of ATM/p53-dependent suppression of myc-induced lymphomas by Wip1 phosphatase. J. Exp. Med. 203, 2793–2799 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Fujimoto, H. et al. Regulation of the antioncogenic Chk2 kinase by the oncogenic Wip1 phosphatase. Cell Death Differ. 13, 1170–1180 (2006)

    Article  CAS  PubMed  Google Scholar 

  20. Bulavin, D. V. et al. Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nature Genet. 31, 210–215 (2002)

    Article  CAS  PubMed  Google Scholar 

  21. Natrajan, R. et al. Tiling path genomic profiling of grade 3 invasive ductal breast cancers. Clin. Cancer Res. 15, 2711–2722 (2009)

    Article  CAS  PubMed  Google Scholar 

  22. Tan, D. S. et al. PPM1D is a potential therapeutic target in ovarian clear cell carcinomas. Clin. Cancer Res. 15, 2269–2280 (2009)

    Article  CAS  PubMed  Google Scholar 

  23. Rayter, S. et al. A chemical inhibitor of PPM1D that selectively kills cells overexpressing PPM1D. Oncogene 27, 1036–1044 (2008)

    Article  CAS  PubMed  Google Scholar 

  24. Hayashi, R. et al. Optimization of a cyclic peptide inhibitor of Ser/Thr phosphatase PPM1D (Wip1). Biochemistry 50, 4537–4549 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chuman, Y. et al. PPM1D430, a novel alternative splicing variant of the human PPM1D, can dephosphorylate p53 and exhibits specific tissue expression. J. Biochem. 145, 1–12 (2009)

    Article  CAS  PubMed  Google Scholar 

  26. Silva, A. L., Ribeiro, P., Inacio, A., Liebhaber, S. A. & Romao, L. Proximity of the poly(A)-binding protein to a premature termination codon inhibits mammalian nonsense-mediated mRNA decay. RNA 14, 563–576 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Jacobs, K. B. et al. Detectable clonal mosaicism and its relationship to aging and cancer. Nature Genet. 44, 651–658 (2012)

    Article  CAS  PubMed  Google Scholar 

  28. Laurie, C. C. et al. Detectable clonal mosaicism from birth to old age and its relationship to cancer. Nature Genet. 44, 642–650 (2012)

    Article  CAS  PubMed  Google Scholar 

  29. Barnes, D. R. et al. Evaluation of assosciation methods for analysing modifiers of disease risk in carriers of high risk mutations. Genet. Epidemiol. 36, 274–291 (2012)

    Article  PubMed  Google Scholar 

  30. Antoniou, A. C. & Easton, D. F. Polygenic inheritance of breast cancer: implications for design of association studies. Genet. Epidemiol. 25, 190–202 (2003)

    Article  PubMed  Google Scholar 

  31. Tavassoli, F. A. & Devilee, P. in Pathology and Genetics of Tumours of the Breast and Female Genital Organs (IARC, 2003)

    Google Scholar 

  32. Hernandez, L. et al. Genomic and mutational profiling of ductal carcinomas in situ and matched adjacent invasive breast cancers reveals intra-tumour genetic heterogeneity and clonal selection. J. Pathol. 227, 42–52 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gnirke, A. et al. Solution hybrid selection with ultra-long oligonucleotides for massively parallel targeted sequencing. Nature Biotechnol. 27, 182–189 (2009)

    Article  CAS  Google Scholar 

  34. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Turnbull, C. et al. Genome-wide association study identifies five new breast cancer susceptibility loci. Nature Genet. 42, 504–507 (2010)

    Article  CAS  PubMed  Google Scholar 

  36. Lunter, G. & Goodson, M. Stampy: a statistical algorithm for sensitive and fast mapping of Illumina sequence reads. Genome Res. 21, 936–939 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Rimmer, A., Mathieson, I., Lunter, G. & McVean, G. Platypus: An Integrated Variant Caller (http://www.well.ox.ac.uk/platypus) (2012)

  38. Curado, M. P. et al. Cancer incidence in five continents, Volume VIII. IARC Sci. Publ. No 160, 1–781 (2002)

    Google Scholar 

  39. Antoniou, A. C. et al. Evidence for further breast cancer susceptibility genes in addition to BRCA1 and BRCA2 in a population-based study. Genet. Epidemiol. 21, 1–18 (2001)

    Article  CAS  PubMed  Google Scholar 

  40. Lange, K., Weeks, D. & Boehnke, M. Programs for Pedigree Analysis: MENDEL, FISHER, and dGENE. Genet. Epidemiol. 5, 471–472 (1988)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all the subjects and families that participated in the research and D. Dudakia, J. Bull and R. Linger for their assistance in recruitment. We are indebted to M. Stratton for discussions of the data and to A. Strydom for editorial assistance. We thank the High-Throughput Genomics Group at the Wellcome Trust Centre for Human Genetics, Oxford (funded by Wellcome Trust grant reference 090532/Z/09/Z and Medical Research Council (MRC) Hub grant G0900747 91070) for the generation of the phase 1 sequencing data. This work was funded by the Institute of Cancer Research, The Wellcome Trust, Cancer Research UK and Breakthrough Breast Cancer. We acknowledge support by the RMH-ICR National Institute for Health Research (NIHR) Specialist Biomedical Research Centre for Cancer. We acknowledge the use of DNA from the British 1958 Birth Cohort collection funded by the MRC grant G0000934 and the Wellcome Trust grant 068545/Z/02. A.C.A. is a Cancer Research UK Senior Cancer Research Fellow (C12292/A11174). P.Do. is supported by a Wolfson-Royal Society Merit Award. K.S. is supported by the Michael and Betty Kadoorie Cancer Genetics Research Programme.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

E.R., K.S., P.H., N.M.W., T.-P.Y., M.A.B., M.J.C., C.T., J.T., M.I.M., P.De., P.Do. and N.R. (chair) are the Wellcome Trust Case Control Consortium (WTCCC) exon-resequencing group who devised and funded phase 1. J.W.A., J.Ba., J.Be., A.F.B., C.B., G.B., C.C., J.C., R.D., A.D., F.D., D.E., D.G.E., L.G., A.H., L.I., A.K., F.L., Z.M., P.J.M., J.P., M.P., M.T.R., S.Sh., L.W. and N.R. are centre leads of the Breast and Ovarian Cancer Susceptibility Collaboration (BOCS), which is coordinated by M.W.-P. and A.Z. A full list of the WTCCC and BOCS consortia is in the Supplementary Information. R.H. and M.G. assembled the unselected ovarian cancer series. L.G. coordinated phase 1 sequencing. P.H., M.R. and P.Do. undertook analysis of the pooled DNA repair panel. J.D., A.M., S.Se., S.H. and E.R. undertook NGS sequencing and analysis. S.Se. E.R., S.D.V.S., N.A.P., A.Re., K.S., C.L. and J.D. undertook Sanger sequencing, MLPA and tumour microsatellite analysis. E.R. undertook sample selection and data analyses with C.T. A.C.A. wrote the risk analysis software and oversaw the penetrance analysis that was performed by E.R. A.Ri. provided and optimized Platypus. D.N.R., A.C.-F. and J.S.R.-F. undertook histopathological analyses and performed microdissections. I.B., R.B., C.J.L. and A.A. undertook functional analyses. E.R., K.S. and N.R. managed and oversaw all aspects of the study and wrote the manuscript.

Corresponding author

Correspondence to Nazneen Rahman.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Lists of participants and their affiliations appear in the Supplementary Information.

Lists of participants and their affiliations appear in the Supplementary Information.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-8 and details of the WTCCC and BOCS consortia. (PDF 2900 kb)

Supplementary Tables

This file contains Supplementary Tables 1-8. (XLS 168 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ruark, E., Snape, K., Humburg, P. et al. Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer. Nature 493, 406–410 (2013). https://doi.org/10.1038/nature11725

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature11725

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: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer