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.

Integrated molecular analysis of clear-cell renal cell carcinoma

Subjects

Abstract

Clear-cell renal cell carcinoma (ccRCC) is the most prevalent kidney cancer and its molecular pathogenesis is incompletely understood. Here we report an integrated molecular study of ccRCC in which ≥100 ccRCC cases were fully analyzed by whole-genome and/or whole-exome and RNA sequencing as well as by array-based gene expression, copy number and/or methylation analyses. We identified a full spectrum of genetic lesions and analyzed gene expression and DNA methylation signatures and determined their impact on tumor behavior. Defective VHL-mediated proteolysis was a common feature of ccRCC, which was caused not only by VHL inactivation but also by new hotspot TCEB1 mutations, which abolished Elongin C–VHL binding, leading to HIF accumulation. Other newly identified pathways and components recurrently mutated in ccRCC included PI3K-AKT-mTOR signaling, the KEAP1-NRF2-CUL3 apparatus, DNA methylation, p53-related pathways and mRNA processing. This integrated molecular analysis unmasked new correlations between DNA methylation, gene mutation and/or gene expression and copy number profiles, enabling the stratification of clinical risks for patients with ccRCC.

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: Mutations in 3p target genes and their impact on survival.
Figure 2: New TCEB1 mutations and HIF accumulation.
Figure 3: Significantly mutated genes and pathways for 106 ccRCC specimens.
Figure 4: Significant copy number alterations in 240 ccRCC specimens.
Figure 5: Significantly mutated pathways for 106 ccRCC specimens.
Figure 6: Correlations between DNA methylation and other genetic lesions.

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. Rini, B.I., Campbell, S.C. & Escudier, B. Renal cell carcinoma. Lancet 373, 1119–1132 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Ljungberg, B. et al. EAU guidelines on renal cell carcinoma: the 2010 update. Eur. Urol. 58, 398–406 (2010).

    Article  PubMed  Google Scholar 

  4. Gnarra, J.R. et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat. Genet. 7, 85–90 (1994).

    Article  CAS  PubMed  Google Scholar 

  5. Gallou, C. et al. Mutations of the VHL gene in sporadic renal cell carcinoma: definition of a risk factor for VHL patients to develop an RCC. Hum. Mutat. 13, 464–475 (1999).

    Article  CAS  PubMed  Google Scholar 

  6. Schraml, P. et al. VHL mutations and their correlation with tumour cell proliferation, microvessel density, and patient prognosis in clear cell renal cell carcinoma. J. Pathol. 196, 186–193 (2002).

    Article  CAS  PubMed  Google Scholar 

  7. Herman, J.G. et al. Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc. Natl. Acad. Sci. USA 91, 9700–9704 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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 

  9. 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 

  10. Peña-Llopis, S. et al. BAP1 loss defines a new class of renal cell carcinoma. Nat. Genet. 44, 751–759 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Guo, G. et al. Frequent mutations of genes encoding ubiquitin-mediated proteolysis pathway components in clear cell renal cell carcinoma. Nat. Genet. 44, 17–19 (2012).

    Article  CAS  Google Scholar 

  12. Greenman, C. et al. Patterns of somatic mutation in human cancer genomes. Nature 446, 153–158 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. Huang, J. et al. Exome sequencing of hepatitis B virus–associated hepatocellular carcinoma. Nat. Genet. 44, 1117–1121 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Sung, W.K. et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat. Genet. 44, 765–769 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. 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).

    Article  CAS  PubMed  Google Scholar 

  17. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hakimi, A.A. et al. Adverse outcomes in clear cell renal cell carcinoma with mutations of 3p21 epigenetic regulators BAP1 and SETD2: a report by MSKCC and the KIRC TCGA Research Network. Clin. Cancer Res. 19, 3259–3267 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hakimi, A.A. et al. Clinical and pathologic impact of select chromatin-modulating tumor suppressors in clear cell renal cell carcinoma. Eur. Urol. 63, 848–854 (2013).

    Article  PubMed  Google Scholar 

  20. Kapur, P. et al. Effects on survival of BAP1 and PBRM1 mutations in sporadic clear-cell renal-cell carcinoma: a retrospective analysis with independent validation. Lancet Oncol. 14, 159–167 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Aso, T., Lane, W.S., Conaway, J.W. & Conaway, R.C. Elongin (SIII): a multisubunit regulator of elongation by RNA polymerase II. Science 269, 1439–1443 (1995).

    Article  CAS  PubMed  Google Scholar 

  22. Kamura, T. et al. Activation of HIF1a ubiquitination by a reconstituted von Hippel-Lindau (VHL) tumor suppressor complex. Proc. Natl. Acad. Sci. USA 97, 10430–10435 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Stebbins, C.E., Kaelin, W.G. Jr. & Pavletich, N.P. Structure of the VHL-ElonginC-ElonginB complex: implications for VHL tumor suppressor function. Science 284, 455–461 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Takagi, Y., Pause, A., Conaway, R.C. & Conaway, J.W. Identification of elongin C sequences required for interaction with the von Hippel-Lindau tumor suppressor protein. J. Biol. Chem. 272, 27444–27449 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Delhommeau, F. et al. Mutation in TET2 in myeloid cancers. N. Engl. J. Med. 360, 2289–2301 (2009).

    Article  PubMed  Google Scholar 

  26. Langemeijer, S.M. et al. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat. Genet. 41, 838–842 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Chen, Q., Chen, Y., Bian, C., Fujiki, R. & Yu, X. TET2 promotes histone O-GlcNAcylation during gene transcription. Nature 493, 561–564 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. The Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

  29. Zimmerman, E.S., Schulman, B.A. & Zheng, N. Structural assembly of cullin–RING ubiquitin ligase complexes. Curr. Opin. Struct. Biol. 20, 714–721 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Padmanabhan, B. et al. Structural basis for defects of Keap1 activity provoked by its point mutations in lung cancer. Mol. Cell 21, 689–700 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Shibata, T. et al. Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Proc. Natl. Acad. Sci. USA 105, 13568–13573 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kim, Y.R. et al. Oncogenic NRF2 mutations in squamous cell carcinomas of oesophagus and skin. J. Pathol. 220, 446–451 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Adam, J. et al. Renal cyst formation in Fh1-deficient mice is independent of the Hif/Phd pathway: roles for fumarate in KEAP1 succination and Nrf2 signaling. Cancer Cell 20, 524–537 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kinch, L., Grishin, N.V. & Brugarolas, J. Succination of Keap1 and activation of Nrf2-dependent antioxidant pathways in FH-deficient papillary renal cell carcinoma type 2. Cancer Cell 20, 418–420 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ooi, A. et al. CUL3 and NRF2 mutations confer an NRF2 activation phenotype in a sporadic form of papillary renal cell carcinoma. Cancer Res. 73, 2044–2051 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Kucejova, B. et al. Interplay between pVHL and mTORC1 pathways in clear-cell renal cell carcinoma. Mol. Cancer Res. 9, 1255–1265 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yoshida, K. et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 478, 64–69 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Kadoch, C. et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat. Genet. 45, 592–601 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Clark, J. et al. Fusion of splicing factor genes PSF and NonO (p54nrb) to the TFE3 gene in papillary renal cell carcinoma. Oncogene 15, 2233–2239 (1997).

    Article  CAS  PubMed  Google Scholar 

  40. Ross, H. & Argani, P. Xp11 translocation renal cell carcinoma. Pathology 42, 369–373 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Kuroda, N. et al. Review of renal carcinoma associated with Xp11.2 translocations/TFE3 gene fusions with focus on pathobiological aspect. Histol. Histopathol. 27, 133–140 (2012).

    PubMed  Google Scholar 

  42. Brannon, A.R. et al. Molecular stratification of clear cell renal cell carcinoma by consensus clustering reveals distinct subtypes and survival patterns. Genes Cancer 1, 152–163 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Huang, 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kent, W.J. BLAT—the BLAST-like alignment tool. Genome Res. 12, 656–664 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Benson, G. Tandem repeats finder: a program to analyze DNA sequences. Nucleic Acids Res. 27, 573–580 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shiraishi, Y. et al. An empirical Bayesian framework for somatic mutation detection from cancer genome sequencing data. Nucleic Acids Res. 41, e89 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hellmann, I. et al. Why do human diversity levels vary at a megabase scale? Genome Res. 15, 1222–1231 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Stamatoyannopoulos, J.A. et al. Human mutation rate associated with DNA replication timing. Nat. Genet. 41, 393–395 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wendl, M.C. et al. PathScan: a tool for discerning mutational significance in groups of putative cancer genes. Bioinformatics 27, 1595–1602 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Nannya, Y. et al. A robust algorithm for copy number detection using high-density oligonucleotide single nucleotide polymorphism genotyping arrays. Cancer Res. 65, 6071–6079 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Yamamoto, G. et al. Highly sensitive method for genomewide detection of allelic composition in nonpaired, primary tumor specimens by use of Affymetrix single-nucleotide-polymorphism genotyping microarrays. Am. J. Hum. Genet. 81, 114–126 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tsuji, K., Ishikawa, Y. & Imamura, T. Technique for differentiating alveolar soft part sarcoma from other tumors in paraffin-embedded tissue: comparison of immunohistochemistry for TFE3 and CD147 and of reverse transcription polymerase chain reaction for ASPSCR1-TFE3 fusion transcript. Hum. Pathol. 43, 356–363 (2012).

    Article  CAS  PubMed  Google Scholar 

  54. Morita, S., Kojima, T. & Kitamura, T. Plat-E: an efficient and stable system for transient packaging of retroviruses. Gene Ther. 7, 1063–1066 (2000).

    CAS  PubMed  Google Scholar 

  55. Nabekura, T., Otsu, M., Nagasawa, T., Nakauchi, H. & Onodera, M. Potent vaccine therapy with dendritic cells genetically modified by the gene-silencing-resistant retroviral vector GCDNsap. Mol. Ther. 13, 301–309 (2006).

    Article  CAS  PubMed  Google Scholar 

  56. Kamura, T. et al. VHL-box and SOCS-box domains determine binding specificity for Cul2-Rbx1 and Cul5-Rbx2 modules of ubiquitin ligases. Genes Dev. 18, 3055–3065 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Garrett, K.P. et al. Positive regulation of general transcription factor SIII by a tailed ubiquitin homolog. Proc. Natl. Acad. Sci. USA 92, 7172–7176 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Y. Mori, M. Nakamura, N. Mizota and S. Ichimura for their technical assistance. We also thank M. Nangaku and N. Takeda for fruitful discussion and comments. We thank T. Kitamura (University of Tokyo) for providing pMXs-puro, M. Onodera (National Center for Child Health and Development, Japan) for providing pGCDNsamIRESEGFP and R.C. Mulligan (Boston Children's Hospital) for providing 293gp cells. This work was supported by KAKENHI (22134006), the Industrial Technology Research Grant Program from the New Energy and Industrial Technology Development Organization (NEDO) (08C46598a) and the Japan Society for the Promotion of Science through the Funding Program for World-Leading Innovative R&D on Science and Technology, initiated by the Council for Science and Technology Policy.

Author information

Authors and Affiliations

Authors

Contributions

Y. Sato, S. Maekawa, Y.N., H.S., Y. Suzuki, S.S., K.Y. and A.K. performed DNA sequencing. Y. Shiraishi, Y.O., K.C., H.T., A.F., T.T. and S. Miyano performed bioinformatics analyses of the sequencing data. T.Y., M.S. and T.K. performed the functional analyses of Elongin C mutants. Y. Sato, A.S.-O., A.N. and M.S. performed SNP array and expression array analyses. T.S., G.N. and H.A. performed methylation analysis. H.K. and Y.H. provided specimens and were also involved in planning the project. T.M., D.M. and M.F. confirmed histological diagnosis and performed immunostaining for HIF proteins. Y. Sato, T.Y., Y.O., A.S.-O. and S.O. generated figures and tables and wrote the manuscript. S.O. led the entire project. All authors participated in the discussion and interpretation of data and results.

Corresponding author

Correspondence to Seishi Ogawa.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–22, Supplementary Tables 5–9 and 15, and Supplementary Note (PDF 29982 kb)

Supplementary Table 1

Characteristics of the patients (XLSX 36 kb)

Supplementary Table 2

List of mutations in whole-genome sequencing (XLSX 5326 kb)

Supplementary Table 3

List of mutations in exome sequencing (XLSX 525 kb)

Supplementary Table 4

Recurrently mutated genes found in whole-exome sequencing (XLSX 64 kb)

Supplementary Table 10

Summary of RNA sequencing data (XLSX 15 kb)

Supplementary Table 11

Fusion transcripts detected in RNA sequencing (XLSX 15 kb)

Supplementary Table 12

Overexpressed annotation terms among differentially methylated genes (XLSX 23 kb)

Supplementary Table 13

List of PCR primers for frequently mutated genes (XLSX 13 kb)

Supplementary Table 14

List of PCR primers used for deep sequencing (XLSX 63 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sato, Y., Yoshizato, T., Shiraishi, Y. et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet 45, 860–867 (2013). https://doi.org/10.1038/ng.2699

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2699

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