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:

IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype

This article has been updated

Abstract

Both genome-wide genetic and epigenetic alterations are fundamentally important for the development of cancers, but the interdependence of these aberrations is poorly understood. Glioblastomas and other cancers with the CpG island methylator phenotype (CIMP) constitute a subset of tumours with extensive epigenomic aberrations and a distinct biology1,2,3. Glioma CIMP (G-CIMP) is a powerful determinant of tumour pathogenicity, but the molecular basis of G-CIMP remains unresolved. Here we show that mutation of a single gene, isocitrate dehydrogenase 1 (IDH1), establishes G-CIMP by remodelling the methylome. This remodelling results in reorganization of the methylome and transcriptome. Examination of the epigenome of a large set of intermediate-grade gliomas demonstrates a distinct G-CIMP phenotype that is highly dependent on the presence of IDH mutation. Introduction of mutant IDH1 into primary human astrocytes alters specific histone marks, induces extensive DNA hypermethylation, and reshapes the methylome in a fashion that mirrors the changes observed in G-CIMP-positive lower-grade gliomas. Furthermore, the epigenomic alterations resulting from mutant IDH1 activate key gene expression programs, characterize G-CIMP-positive proneural glioblastomas but not other glioblastomas, and are predictive of improved survival. Our findings demonstrate that IDH mutation is the molecular basis of CIMP in gliomas, provide a framework for understanding oncogenesis in these gliomas, and highlight the interplay between genomic and epigenomic changes in human cancers.

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: Introduction of mutant IDH1 into human astrocytes remodels the methylome.
Figure 2: Global epigenetic analysis of LGGs reveals dependence of G-CIMP on IDH mutation.
Figure 3: IDH1 mutation directly generates the methylation patterns present in G-CIMP tumours.
Figure 4: Functional implications of IDH1-mutation-induced alterations in the glioma epigenome.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Data sets have been deposited in the Gene Expression Omnibus under accession number GSE30339.

Change history

  • 27 February 2012

    The original supplementary figures PDF was corrupted and has been replaced.

References

  1. Noushmehr, H. et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17, 510–522 (2010)

    Article  CAS  Google Scholar 

  2. Fang, F. et al. Breast cancer methylomes establish an epigenomic foundation for metastasis. Sci. Transl. Med. 3, 75ra25 (2011)

    Article  Google Scholar 

  3. Toyota, M. et al. CpG island methylator phenotype in colorectal cancer. Proc. Natl Acad. Sci. USA 96, 8681–8686 (1999)

    Article  ADS  CAS  Google Scholar 

  4. Yan, H. et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 360, 765–773 (2009)

    Article  CAS  Google Scholar 

  5. Parsons, D. W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008)

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  Google Scholar 

  7. Mardis, E. R. et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 361, 1058–1066 (2009)

    Article  CAS  Google Scholar 

  8. Dang, L. et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462, 739–744 (2009)

    Article  ADS  CAS  Google Scholar 

  9. Figueroa, M. E. et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 18, 553–567 (2010)

    Article  CAS  Google Scholar 

  10. Ward, P. S. et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting α-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 17, 225–234 (2010)

    Article  CAS  Google Scholar 

  11. Jones, P. A. & Baylin, S. B. The epigenomics of cancer. Cell 128, 683–692 (2007)

    Article  CAS  Google Scholar 

  12. Ohm, J. E. et al. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nature Genet. 39, 237–242 (2007)

    Article  CAS  Google Scholar 

  13. Cheng, Y. W. et al. CpG island methylator phenotype associates with low-degree chromosomal abnormalities in colorectal cancer. Clin. Cancer Res. 14, 6005–6013 (2008)

    Article  CAS  Google Scholar 

  14. Sonoda, Y. et al. Formation of intracranial tumors by genetically modified human astrocytes defines four pathways critical in the development of human anaplastic astrocytoma. Cancer Res. 61, 4956–4960 (2001)

    CAS  PubMed  Google Scholar 

  15. Vivanco, I. et al. Identification of the JNK signaling pathway as a functional target of the tumor suppressor PTEN. Cancer Cell 11, 555–569 (2007)

    Article  CAS  Google Scholar 

  16. Sonoda, Y. et al. Akt pathway activation converts anaplastic astrocytoma to glioblastoma multiforme in a human astrocyte model of glioma. Cancer Res. 61, 6674–6678 (2001)

    CAS  PubMed  Google Scholar 

  17. Pieper, R. O. Defined human cellular systems in the study of glioma development. Front. Biosci. 8, s19–s27 (2003)

    Article  CAS  Google Scholar 

  18. Bibikova, M. et al. High density DNA methylation array with single CpG site resolution. Genomics 98, 288–295 (2011)

    Article  CAS  Google Scholar 

  19. Sandoval, J. et al. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics 6, 692–702 (2011)

    Article  CAS  Google Scholar 

  20. Mutskov, V. & Felsenfeld, G. Silencing of transgene transcription precedes methylation of promoter DNA and histone H3 lysine 9. EMBO J. 23, 138–149 (2004)

    Article  CAS  Google Scholar 

  21. Bracken, A. P. et al. Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Genes Dev. 20, 1123–1136 (2006)

    Article  CAS  Google Scholar 

  22. Furnari, F. B. et al. Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 21, 2683–2710 (2007)

    Article  CAS  Google Scholar 

  23. Docherty, S. J. et al. Bisulfite-based epityping on pooled genomic DNA provides an accurate estimate of average group DNA methylation. Epigenetics Chromatin 2, 3 (2009)

    Article  Google Scholar 

  24. Hegi, M. E. et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 352, 997–1003 (2005)

    Article  CAS  Google Scholar 

  25. Solomon, D. A. et al. Identification of p18INK4c as a tumor suppressor gene in glioblastoma multiforme. Cancer Res. 68, 2564–2569 (2008)

    Article  CAS  Google Scholar 

  26. Etcheverry, A. et al. DNA methylation in glioblastoma: impact on gene expression and clinical outcome. BMC Genomics 11, 701 (2010)

    Article  CAS  Google Scholar 

  27. The Cancer Genome Atlas Network Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008)

    Article  Google Scholar 

  28. Lendahl, U., Zimmerman, L. B. & McKay, R. D. CNS stem cells express a new class of intermediate filament protein. Cell 60, 585–595 (1990)

    Article  CAS  Google Scholar 

  29. Xu, W. et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 19, 17–30 (2011)

    Article  CAS  Google Scholar 

  30. He, Y. F. et al. Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science 333, 1303–1307 (2011)

    Article  ADS  CAS  Google Scholar 

  31. Reich, M. et al. GenePattern 2.0. Nature Genet. 38, 500–501 (2006)

    Article  CAS  Google Scholar 

  32. Verhaak, R. G. et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17, 98–110 (2010)

    Article  CAS  Google Scholar 

  33. Thomas, P. D. et al. PANTHER: a library of protein families and subfamilies indexed by function. Genome Res. 13, 2129–2141 (2003)

    Article  CAS  Google Scholar 

  34. Rhodes, D. R. et al. Molecular concepts analysis links tumors, pathways, mechanisms, and drugs. Neoplasia 9, 443–454 (2007)

    Article  CAS  Google Scholar 

  35. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005)

    Article  ADS  CAS  Google Scholar 

  36. Vlassenbroeck, I. et al. Validation of real-time methylation-specific PCR to determine O6-methylguanine-DNA methyltransferase gene promoter methylation in glioma. J. Mol. Diagn. 10, 332–337 (2008)

    Article  CAS  Google Scholar 

  37. Gordon, D., Abajian, C. & Green, P. Consed: a graphical tool for sequence finishing. Genome Res. 8, 195–202 (1998)

    Article  CAS  Google Scholar 

  38. Nickerson, D. A., Tobe, V. O. & Taylor, S. L. PolyPhred: automating the detection and genotyping of single nucleotide substitutions using fluorescence-based resequencing. Nucleic Acids Res. 25, 2745–2751 (1997)

    Article  CAS  Google Scholar 

  39. Chen, K. et al. PolyScan: an automatic indel and SNP detection approach to the analysis of human resequencing data. Genome Res. 17, 659–666 (2007)

    Article  CAS  Google Scholar 

  40. Major, J. E. Genomic mutation consequence calculator. Bioinformatics 23, 3091–3092 (2007)

    Article  CAS  Google Scholar 

  41. Galli, R. et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 64, 7011–7021 (2004)

    Article  CAS  Google Scholar 

  42. De Filippis, L. et al. A novel, immortal, and multipotent human neural stem cell line generating functional neurons and oligodendrocytes. Stem Cells 25, 2312–2321 (2007)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Kayserian, K. Huberman, I. Dolgalev and S. Thomas for technical expertise. We thank C. Sawyers and E. Holland for helpful discussions. This grant was supported in part by the National Institutes of Health (R01CA154767-01) (T.A.C.), the National Cancer Institute (U54-CA143798) (I.K.M.), an Advanced Clinical Research Award in Glioma from the American Society of Clinical Oncology (I.K.M.), the Doris Duke Charitable Fund (I.K.M., T.A.C.), a National Brain Tumor Society Systems Biology Research Grant (I.K.M.), the MSKCC Brain Tumor Center (T.A.C.), the Louis Gerstner Foundation (T.A.C.), the STARR Cancer Consortium (T.A.C.) and the Sontag Foundation (T.A.C., I.K.M.).

Author information

Authors and Affiliations

Authors

Contributions

T.A.C., S.T., A.G. and I.K.M. designed the experiments. S.T., A.G., F.F., D.R., A.H., L.A.W., C.C., E.Y., C.L., P.S.W., A.V., J.T.H., A.W.M.F. and L.G.T.M. performed the experiments. S.T., J.T.H., A.G., F.F., A.K., A.H., E.Y., A.V., P.S.W., C.B.T., T.A.C. and I.K.M. analysed the data. D.R., O.G., R.L. and I.K.M. contributed new reagents. T.A.C., S.T., I.K.M. and A.G. wrote the paper.

Corresponding authors

Correspondence to Ingo K. Mellinghoff or Timothy A. Chan.

Ethics declarations

Competing interests

C.B.T. is a consultant of Agios Pharmaceuticals and has a financial interest in Agios.

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-14 with legends. These figures show methylation profiles of parental, IDH1 wild-type and IDH1 mutant astrocytes; validation of differentially methylated regions using EpiTYPER; association of CIMP identified from lower grade gliomas with various clinical covariates; predictive power of the 17-gene signature on the MSKCC cohort and the Rembrandt validation dataset; promotion of a neurosphere phenotype in human astrocytes upon mutant IDH1 expression. The original file posted on line was corrupted and was replaced on 27 February 2012. (PDF 3831 kb)

Supplementary Tables 1-17

Table 1 shows the differentially methylated genes at passage 40 in IDH1 R132H expressing human astrocytes. Table 2 shows the differentially expressed genes at passage 40 in IDH1 R132H expressing human astrocytes. Table 3 shows the enrichment of PANTHER pathways and biological processes in differentially expressed genes at passage 40 in mutant IDH1 expressing human astrocytes. Table 4 includes the enriched literature defined Oncomine concepts in mutant IDH1 expressing human astrocytes. Table 5 includes the patient characteristics for the MSKCC Cohort used for methylation and expression screens (MS Excel spreadsheet; 49 KB). Table 6 shows the differentially methylated genes between CIMP groups in MSKCC cohort of lower grade glioma samples (MS Excel spreadsheet; 9.9 MB). Table 7 shows the enriched gene sets and PRC2 targets as identified by GSEA in CIMP+ tumors. Table 8 shows the differentially expressed genes in CIMP tumors. Table 9 shows the PANTHER ontology terms enriched in the differentially expressed genes in CIMP tumors. Table 10 shows the multivariate analysis of predictors of CIMP in MSKCC Cohort. Table 11 shows the multivariate analysis of survival in lower grade gliomas (MS Word document 156 KB). Table 12 shows the hypermethylated and downregulated genes in CIMP positive tumors. Table 13 shows the hypermethylated probes used for GSEA. Table 14 shows the differentially methylated probes from IDH1 expressing human astrocytes used to classify CIMP tumors from MSKCC cohort. Table 15 shows the 17 gene signature derived from clinical and cell line data used to classify TCGA samples.  Table 16 shows the multivariate analysis of predictors of survival in Rembrandt validation data set. Table 17 shows the EpiTYPER primers used for validation of methylated genes in IDH1 expressing astrocytes and LGG tumors. (ZIP 8126 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Turcan, S., Rohle, D., Goenka, A. et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–483 (2012). https://doi.org/10.1038/nature10866

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature10866

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