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:

Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes

Abstract

Endometrial cancer is the sixth most commonly diagnosed cancer in women worldwide, causing 74,000 deaths annually1. Serous endometrial cancers are a clinically aggressive subtype with a poorly defined genetic etiology2,3,4. We used whole-exome sequencing to comprehensively search for somatic mutations within 22,000 protein-encoding genes in 13 primary serous endometrial tumors. We subsequently resequenced 18 genes, which were mutated in more than 1 tumor and/or were components of an enriched functional grouping, from 40 additional serous tumors. We identified high frequencies of somatic mutations in CHD4 (17%), EP300 (8%), ARID1A (6%), TSPYL2 (6%), FBXW7 (29%), SPOP (8%), MAP3K4 (6%) and ABCC9 (6%). Overall, 36.5% of serous tumors had a mutated chromatin-remodeling gene, and 35% had a mutated ubiquitin ligase complex gene, implicating frequent mutational disruption of these processes in the molecular pathogenesis of one of the deadliest forms of endometrial cancer.

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: Somatic mutations in CHD4, FBXW7 and SPOP cluster within important functional domains of the encoded proteins.
Figure 2: Distribution of nonsynonymous somatic mutations in endometrial cancers.
Figure 3: Somatic mutations in the consensus cancer genes EP300 and ARID1A relative to the functional domains of the encoded proteins.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

References

  1. Ferlay, J. et al. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer 127, 2893–2917 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Sherman, M.E. Theories of endometrial carcinogenesis: a multidisciplinary approach. Mod. Pathol. 13, 295–308 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. Hamilton, C.A. et al. Uterine papillary serous and clear cell carcinomas predict for poorer survival compared to grade 3 endometrioid corpus cancers. Br. J. Cancer 94, 642–646 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hendrickson, M., Ross, J., Eifel, P., Martinez, A. & Kempson, R. Uterine papillary serous carcinoma: a highly malignant form of endometrial adenocarcinoma. Am. J. Surg. Pathol. 6, 93–108 (1982).

    Article  CAS  PubMed  Google Scholar 

  5. McConechy, M.K. et al. Subtype-specific mutation of PPP2R1A in endometrial and ovarian carcinomas. J. Pathol. 223, 567–573 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Rudd, M.L. et al. A unique spectrum of somatic PIK3CA (p110α) mutations within primary endometrial carcinomas. Clin. Cancer Res. 17, 1331–1340 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Shih, I.M. et al. Somatic mutations of PPP2R1A in ovarian and uterine carcinomas. Am. J. Pathol. 178, 1442–1447 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hayes, M.P., Douglas, W. & Ellenson, L.H. Molecular alterations of EGFR and PIK3CA in uterine serous carcinoma. Gynecol. Oncol. 113, 370–373 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. National Cancer Institute, SEER Program. SEER Survival Monograph: Cancer Survival Among Adults: U.S. SEER Program, 1988–2001. Patient and Tumor Characteristics (eds. Ries, L.A.G., Young, J.L., Keel, G.E., Eisner, M.P., Lin, Y.D. & Horner, M.-J.) (National Cancer Institute, SEER Program, Bethesda, MD, 2007).

  10. Zhang, Y., LeRoy, G., Seelig, H.P., Lane, W.S. & Reinberg, D. The dermatomyositis-specific autoantigen Mi2 is a component of a complex containing histone deacetylase and nucleosome remodeling activities. Cell 95, 279–289 (1998).

    Article  CAS  PubMed  Google Scholar 

  11. Li, J., Lin, Q., Wang, W., Wade, P. & Wong, J. Specific targeting and constitutive association of histone deacetylase complexes during transcriptional repression. Genes Dev. 16, 687–692 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tong, J.K., Hassig, C.A., Schnitzler, G.R., Kingston, R.E. & Schreiber, S.L. Chromatin deacetylation by an ATP-dependent nucleosome remodelling complex. Nature 395, 917–921 (1998).

    Article  CAS  PubMed  Google Scholar 

  13. Xue, Y. et al. NURD, a novel complex with both ATP-dependent chromatin-remodeling and histone deacetylase activities. Mol. Cell 2, 851–861 (1998).

    Article  CAS  PubMed  Google Scholar 

  14. Hall, J.A. & Georgel, P.T. CHD proteins: a diverse family with strong ties. Biochem. Cell Biol. 85, 463–476 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Lai, A.Y. & Wade, P.A. Cancer biology and NuRD: a multifaceted chromatin remodelling complex. Nat. Rev. Cancer 11, 588–596 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Polo, S.E., Kaidi, A., Baskcomb, L., Galanty, Y. & Jackson, S.P. Regulation of DNA-damage responses and cell-cycle progression by the chromatin remodelling factor CHD4. EMBO J. 29, 3130–3139 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Smeenk, G. et al. The NuRD chromatin-remodeling complex regulates signaling and repair of DNA damage. J. Cell Biol. 190, 741–749 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chou, D.M. et al. A chromatin localization screen reveals poly (ADP ribose)–regulated recruitment of the repressive polycomb and NuRD complexes to sites of DNA damage. Proc. Natl. Acad. Sci. USA 107, 18475–18480 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Larsen, D.H. et al. The chromatin-remodeling factor CHD4 coordinates signaling and repair after DNA damage. J. Cell Biol. 190, 731–740 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Boerkoel, C.F. et al. Mutant chromatin remodeling protein SMARCAL1 causes Schimke immuno-osseous dysplasia. Nat. Genet. 30, 215–220 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Tsurusaki, Y. et al. Mutations affecting components of the SWI/SNF complex cause Coffin-Siris syndrome. Nat. Genet. 44, 376–378 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Van Houdt, J.K. et al. Heterozygous missense mutations in SMARCA2 cause Nicolaides-Baraitser syndrome. Nat. Genet. 44, 445–449 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Welcker, M. & Clurman, B.E. FBW7 ubiquitin ligase: a tumour suppressor at the crossroads of cell division, growth and differentiation. Nat. Rev. Cancer 8, 83–93 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Cassia, R. et al. Cyclin E gene (CCNE) amplification and hCDC4 mutations in endometrial carcinoma. J. Pathol. 201, 589–595 (2003).

    Article  CAS  PubMed  Google Scholar 

  25. Dutt, A. et al. Drug-sensitive FGFR2 mutations in endometrial carcinoma. Proc. Natl. Acad. Sci. USA 105, 8713–8717 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Spruck, C.H. et al. hCDC4 gene mutations in endometrial cancer. Cancer Res. 62, 4535–4539 (2002).

    CAS  PubMed  Google Scholar 

  27. Suehiro, Y. et al. Aneuploidy predicts outcome in patients with endometrial carcinoma and is related to lack of CDH13 hypermethylation. Clin. Cancer Res. 14, 3354–3361 (2008).

    Article  CAS  PubMed  Google Scholar 

  28. Forbes, S.A. et al. The Catalogue of Somatic Mutations in Cancer (COSMIC). Curr. Protoc. Hum. Genet. Ch. 10, Unit 10.11 (2008).

    Google Scholar 

  29. Reva, B., Antipin, Y. & Sander, C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 39, e118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wertz, I.E. et al. Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature 471, 110–114 (2011).

    Article  CAS  PubMed  Google Scholar 

  31. Garnett, M.J. et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 483, 570–575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Barbieri, C.E. et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat. Genet. 44, 685–689 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Li, C. et al. Tumor-suppressor role for the SPOP ubiquitin ligase in signal-dependent proteolysis of the oncogenic co-activator SRC-3/AIB1. Oncogene 30, 4350–4364 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Glaeser, M., Floetotto, T., Hanstein, B., Beckmann, M.W. & Niederacher, D. Gene amplification and expression of the steroid receptor coactivator SRC3 (AIB1) in sporadic breast and endometrial carcinomas. Horm. Metab. Res. 33, 121–126 (2001).

    Article  CAS  PubMed  Google Scholar 

  35. Huang, D.W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  36. Huang, D.W., Sherman, B.T. & Lempicki, R.A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  CAS  Google Scholar 

  37. Dalgliesh, G.L. et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463, 360–363 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Fujimoto, A. et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat. Genet. 44, 760–764 (2012).

    CAS  PubMed  Google Scholar 

  39. Grasso, C.S. et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 487, 239–243 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gui, Y. et al. Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat. Genet. 43, 875–878 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Guichard, C. et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat. Genet. 44, 694–698 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Jiao, Y. et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331, 1199–1203 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Jones, S. et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228–231 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ong, C.K. et al. Exome sequencing of liver fluke–associated cholangiocarcinoma. Nat. Genet. 44, 690–693 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Parsons, D.W. et al. The genetic landscape of the childhood cancer medulloblastoma. Science 331, 435–439 (2011).

    Article  CAS  PubMed  Google Scholar 

  46. Schwartzentruber, J. et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226–231 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Shain, A.H. et al. Convergent structural alterations define SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeler as a central tumor suppressive complex in pancreatic cancer. Proc. Natl. Acad. Sci. USA 109, E252–E259 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Varela, I. et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469, 539–542 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wiegand, K.C. et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N. Engl. J. Med. 363, 1532–1543 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang, K. et al. Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat. Genet. 43, 1219–1223 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Ogryzko, V.V., Schiltz, R.L., Russanova, V., Howard, B.H. & Nakatani, Y. The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87, 953–959 (1996).

    Article  CAS  PubMed  Google Scholar 

  52. Huang, J., Zhao, Y.L., Li, Y., Fletcher, J.A. & Xiao, S. Genomic and functional evidence for an ARID1A tumor suppressor role. Genes Chromosom. Cancer 46, 745–750 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Guan, B., Wang, T.L. & Shih, I.M. ARID1A, a factor that promotes formation of SWI/SNF-mediated chromatin remodeling, is a tumor suppressor in gynecologic cancers. Cancer Res. 71, 6718–6727 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hargreaves, D.C. & Crabtree, G.R. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res. 21, 396–420 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Guan, B. et al. Mutation and loss of expression of ARID1A in uterine low-grade endometrioid carcinoma. Am. J. Surg. Pathol. 35, 625–632 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  56. McConechy, M.K. et al. Use of mutation profiles to refine the classification of endometrial carcinomas. J. Pathol. 228, 20–30 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Wiegand, K.C. et al. Loss of BAF250a (ARID1A) is frequent in high-grade endometrial carcinomas. J. Pathol. 224, 328–333 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Urick, M.E. et al. PIK3R1 (p85α) is somatically mutated at high frequency in primary endometrial cancer. Cancer Res. 71, 4061–4067 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kuhn, E. et al. Identification of molecular pathway aberrations in uterine serous carcinoma by genome-wide analyses. J. Natl. Cancer Inst. 104, 1503–1513 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Teer, J.K. et al. Systematic comparison of three genomic enrichment methods for massively parallel DNA sequencing. Genome Res. 20, 1420–1431 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Teer, J.K., Green, E.D., Mullikin, J.C. & Biesecker, L.G. VarSifter: visualizing and analyzing exome-scale sequence variation data on a desktop computer. Bioinformatics 28, 599–600 (2012).

    Article  CAS  PubMed  Google Scholar 

  62. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J.R. Stat. Soc. 57, 289–300 (1995).

    Google Scholar 

  63. Sjöblom, T. et al. The consensus coding sequences of human breast and colorectal cancers. Science 314, 268–274 (2006).

    Article  CAS  PubMed  Google Scholar 

  64. Rubin, A.F. & Green, P. Comment on “The consensus coding sequences of human breast and colorectal cancers”. Science 317, 1500 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank our colleagues for critical reading of the manuscript; R.T. Moreland and N. Trivedi of the National Human Genome Research Institute Bioinformatics and Scientific Programming Core, respectively, for performing in silico PCR and giving advice on statistics; and J. Teer for sharing expertise on VarSifter. A. Santin (Yale School of Medicine) kindly provided the ARK1 and ARK2 cell lines. The study was funded in part by the Intramural Program of the National Human Genome Research Institute, US NIH (D.W.B., J.C.M. and M.J.M.); NIH grant R01CA112021 (D.C.S.); the Avon Foundation (D.C.S.); NIH grant R01CA140323 (A.K.G.); and the Ovarian Cancer Fund (A.K.G.). P.H. is supported by grants from the NIH (CA016519) and by the Canadian Institutes for Health Research (MOP-38096).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

D.W.B. designed and directed the study and wrote the manuscript. A.K.G. contributed clinical specimens. M.J.M. and D.C.S. conducted pathological review of clinical specimens. M.L.R. prepared DNA samples and performed identity testing and microsatellite instability analysis. NISC performed library construction and whole-exome sequencing. NISC and N.F.H. performed variant calling. M.L.G. and A.J.O. curated and orthogonally validated exome sequencing data. M.L.G., A.J.O. and D.W.B. interpreted the exome data and established filtering criteria. M.L.G., A.J.O., M.L.R., J.C.P., B.M.E., S.Z. and D.W.B. designed, performed, analyzed and interpreted the mutation prevalence screens. A.J.O. and M.L.G. analyzed MSH6. M.E.U. and M.L.G. generated sequence conservation alignments. M.E.U. performed cell culture and immunoblotting. N.F.H., M.L.G. and J.C.M. performed statistical analyses. N.F.H. performed the power calculation. D.W.B., M.E.U., M.L.G., M.L.R., A.J.O., N.F.H., J.C.M., A.K.G., P.H. and N.J.O. edited and commented on the manuscript.

Corresponding author

Correspondence to Daphne W Bell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A list of members appears in the Supplementary Note.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10, Supplementary Tables 2, 3, 6–10, 12, 14 and 15 and Supplementary Note (PDF 2801 kb)

Supplementary Table 1

Characteristics of endometrial tumors included in the discovery and prevalence screens (XLS 34 kb)

Supplementary Table 4

Filtered exonic and splice junction somatic mutations in a hypermutated tumor (T155) in the discovery screen (XLS 506 kb)

Supplementary Table 5

Filtered exonic and splice junction somatic mutations among 12 tumors in the discovery screen (XLS 272 kb)

Supplementary Table 11

Microsatellite instability (MSI) status and MSH6 status of 160 tumors included in the discovery and prevalence screens of CHD4, FBXW7, and SPOP (XLS 29 kb)

Supplementary Table 13

Enriched functional groupings identified by DAVID analysis (XLS 285 kb)

Supplementary Table 16

All exonic and splice junction somatic mutations among 12 tumors in the discovery screen (XLS 396 kb)

Supplementary Table 17

All exonic and splice junction somatic mutations in a hypermutated tumor (T155) in the discovery screen (XLS 518 kb)

Supplementary Table 18

Primers used for PCR amplification (XLS 219 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Le Gallo, M., O'Hara, A., Rudd, M. et al. Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes. Nat Genet 44, 1310–1315 (2012). https://doi.org/10.1038/ng.2455

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2455

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