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.

Insulator dysfunction and oncogene activation in IDH mutant gliomas

Abstract

Gain-of-function IDH mutations are initiating events that define major clinical and prognostic classes of gliomas1,2. Mutant IDH protein produces a new onco-metabolite, 2-hydroxyglutarate, which interferes with iron-dependent hydroxylases, including the TET family of 5′-methylcytosine hydroxylases3,4,5,6,7. TET enzymes catalyse a key step in the removal of DNA methylation8,9. IDH mutant gliomas thus manifest a CpG island methylator phenotype (G-CIMP)10,11, although the functional importance of this altered epigenetic state remains unclear. Here we show that human IDH mutant gliomas exhibit hypermethylation at cohesin and CCCTC-binding factor (CTCF)-binding sites, compromising binding of this methylation-sensitive insulator protein. Reduced CTCF binding is associated with loss of insulation between topological domains and aberrant gene activation. We specifically demonstrate that loss of CTCF at a domain boundary permits a constitutive enhancer to interact aberrantly with the receptor tyrosine kinase gene PDGFRA, a prominent glioma oncogene. Treatment of IDH mutant gliomaspheres with a demethylating agent partially restores insulator function and downregulates PDGFRA. Conversely, CRISPR-mediated disruption of the CTCF motif in IDH wild-type gliomaspheres upregulates PDGFRA and increases proliferation. Our study suggests that IDH mutations promote gliomagenesis by disrupting chromosomal topology and allowing aberrant regulatory interactions that induce oncogene expression.

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: CTCF binding and gene insulation compromised in IDH mutant gliomas.
Figure 2: Topological domain boundaries disrupted in IDH mutant gliomas.
Figure 3: Insulator loss allows PDGFRA to interact with a constitutive enhancer.
Figure 4: Boundary methylation and CTCF occupancy affect PDGFRA expression and proliferation.

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Data generated for this study are available through the Gene Expression Omnibus (GEO) under accession number GSE70991.

References

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

    Article  ADS  CAS  Google Scholar 

  2. The Cancer Genome Atlas Research Network. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N. Engl. J. Med. 372, 2481–2498 (2015)

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

    Article  ADS  CAS  Google Scholar 

  4. 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 

  5. 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 

  6. Lu, C. et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483, 474–478 (2012)

    Article  ADS  CAS  Google Scholar 

  7. Cairns, R. A. & Mak, T. W. Oncogenic isocitrate dehydrogenase mutations: mechanisms, models, and clinical opportunities. Cancer Dis. 3, 730–741 (2013)

    Article  CAS  Google Scholar 

  8. Pastor, W. A., Aravind, L. & Rao, A. TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nature Rev. Mol. Cell Biol. 14, 341–356 (2013)

    Article  CAS  Google Scholar 

  9. Kohli, R. M. & Zhang, Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 502, 472–479 (2013)

    Article  ADS  CAS  Google Scholar 

  10. 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 

  11. Turcan, S. et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–483 (2012)

    Article  ADS  CAS  Google Scholar 

  12. Bickmore, W. A. & van Steensel, B. Genome architecture: domain organization of interphase chromosomes. Cell 152, 1270–1284 (2013)

    Article  CAS  Google Scholar 

  13. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009)

    Article  ADS  CAS  Google Scholar 

  14. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012)

    Article  ADS  CAS  Google Scholar 

  15. Rao, S. S. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014)

    Article  CAS  Google Scholar 

  16. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012)

    Article  ADS  CAS  Google Scholar 

  17. Lupiáñez, D. G. et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 161, 1012–1025 (2015)

    Article  Google Scholar 

  18. Bell, A. C. & Felsenfeld, G. Methylation of a CTCF-dependent boundary controls imprinted expression of the Igf2 gene. Nature 405, 482–485 (2000)

    Article  ADS  CAS  Google Scholar 

  19. Hark, A. T. et al. CTCF mediates methylation-sensitive enhancer-blocking activity at the H19/Igf2 locus. Nature 405, 486–489 (2000)

    Article  ADS  CAS  Google Scholar 

  20. The GTEx Consortium. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015)

  21. Zuin, J. et al. Cohesin and CTCF differentially affect chromatin architecture and gene expression in human cells. Proc. Natl Acad. Sci. USA 111, 996–1001 (2014)

    Article  ADS  CAS  Google Scholar 

  22. Sturm, D. et al. Paediatric and adult glioblastoma: multiform (epi)genomic culprits emerge. Nature Rev. Cancer 14, 92–107 (2014)

    Article  CAS  Google Scholar 

  23. Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013)

    Article  CAS  Google Scholar 

  24. 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 

  25. Wang, H. et al. Widespread plasticity in CTCF occupancy linked to DNA methylation. Genome Res. 22, 1680–1688 (2012)

    Article  CAS  Google Scholar 

  26. Hsu, P. D., Lander, E. S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014)

    Article  CAS  Google Scholar 

  27. Sander, J. D. & Joung, J. K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nature Biotechnol. 32, 347–355 (2014)

    Article  CAS  Google Scholar 

  28. Baylin, S. B. & Jones, P. A. A decade of exploring the cancer epigenome – biological and translational implications. Nature Rev. Cancer 11, 726–734 (2011)

    Article  CAS  Google Scholar 

  29. Costello, J. F., Berger, M. S., Huang, H. S. & Cavenee, W. K. Silencing of p16/CDKN2 expression in human gliomas by methylation and chromatin condensation. Cancer Res. 56, 2405–2410 (1996)

    CAS  PubMed  Google Scholar 

  30. Koivunen, P. et al. Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation. Nature 483, 484–488 (2012)

    Article  ADS  CAS  Google Scholar 

  31. Chi, A. S. et al. Prospective, high-throughput molecular profiling of human gliomas. J. Neurooncol. 110, 89–98 (2012)

    Article  CAS  Google Scholar 

  32. Rheinbay, E. et al. An aberrant transcription factor network essential for Wnt signaling and stem cell maintenance in glioblastoma. Cell Rep. 3, 1567–1579 (2013)

    Article  CAS  Google Scholar 

  33. Suvà, M. L. et al. Reconstructing and reprogramming the tumor-propagating potential of glioblastoma stem-like cells. Cell 157, 580–594 (2014)

    Article  Google Scholar 

  34. Wakimoto, H. et al. Maintenance of primary tumor phenotype and genotype in glioblastoma stem cells. Neuro Oncol. 14, 132–144 (2012)

    Article  CAS  Google Scholar 

  35. Luchman, H. A. et al. An in vivo patient-derived model of endogenous IDH1-mutant glioma. Neuro Oncol. 14, 184–191 (2012)

    Article  CAS  Google Scholar 

  36. Lai, R. K. et al. Genome-wide methylation analyses in glioblastoma multiforme. PLoS ONE 9, e89376 (2014)

    Article  ADS  Google Scholar 

  37. Abyzov, A., Urban, A. E., Snyder, M. & Gerstein, M. CNVnator: an approach to discover, genotype, and characterize typical and atypical CNVs from family and population genome sequencing. Genome Res. 21, 974–984 (2011)

    Article  CAS  Google Scholar 

  38. Chudnovsky, Y. et al. ZFHX4 interacts with the NuRD core member CHD4 and regulates the glioblastoma tumor-initiating cell state. Cell Rep. 6, 313–324 (2014)

    Article  CAS  Google Scholar 

  39. Bao, S. et al. Targeting cancer stem cells through L1CAM suppresses glioma growth. Cancer Res. 68, 6043–6048 (2008)

    Article  CAS  Google Scholar 

  40. Sandelin, A., Alkema, W., Engstrom, P., Wasserman, W. W. & Lenhard, B. JASPAR: an open-access database for eukaryotic transcription factor binding profiles. Nucleic Acids Res. 32, D91–D94 (2004)

    Article  CAS  Google Scholar 

  41. de Laat, W. & Dekker, J. 3C-based technologies to study the shape of the genome. Methods 58, 189–191 (2012)

    Article  CAS  Google Scholar 

  42. Hagège, H. et al. Quantitative analysis of chromosome conformation capture assays (3C-qPCR). Nature Protocols 2, 1722–1733 (2007)

    Article  Google Scholar 

  43. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013)

    Article  ADS  CAS  Google Scholar 

  44. Cahoy, J. D. et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J. Neurosci. 28, 264–278 (2008)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Kim, the MGH Neuro Oncology Tissue Repository, and the MGH Pathology Flow Cytometry Core for assistance with clinical samples and analysis, and E. Lander and W. Kaelin for discussions. W.A.F. is supported by a basic research fellowship from the American Brain Tumor Association. B.B.L. is supported by a Jane Coffin Childs fellowship. B.E.B. is an American Cancer Society Research Professor. This research was supported by funds from Howard Hughes Medical Institute, the National Brain Tumor Society and the National Human Genome Research Institute.

Author information

Authors and Affiliations

Authors

Contributions

Conception and experimental design: W.A.F., Y.D., B.B.L., S.M.G., M.L.S. and B.E.B. Methodology and data acquisition: W.A.F., Y.D., B.B.L., S.M.G., A.S.V., A.O.S.-R., M.L.S. and B.E.B. Analysis and interpretation of data: W.A.F., Y.D. and B.E.B. Manuscript writing: W.A.F., Y.D. and B.E.B. W.A.F. and Y.D. contributed equally to this work.

Corresponding author

Correspondence to Bradley E. Bernstein.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 DNA methylation and CTCF binding at deregulated boundaries.

a, Box plots show DNA methylation levels over CTCF sites (200-bp window centred on the peak) within boundaries predicted by gene pair correlation analysis to be disrupted. All CTCF sites located within a 1-kb window centred on a disrupted boundary were considered. Methylation levels were determined from whole-genome bisulfite data for three IDH mutant (red labels) and three IDH wild-type (black labels) tumours. b, Bars show average normalized ChIP-seq signal over all CTCF sites located inside a 1-kb window centred on a disrupted boundary.

Extended Data Figure 2 Expression of Fip1l1 in mouse brain cells and survival effects of PDGFRA and FIP1L1.

a, Expression of Fip1l1 in a published data set for isolated mouse brain cell types44. b, Kaplan–Meier plot based on TCGA data23 indicates that combined FIP1L1 and PDGFRA expression is a negative prognostic factor in IDH1 mutant lower-grade gliomas. Multivariate analysis including the known prognostic factor 1p/19q deletion diminished this effect into non-significance, suggesting that other predictors of survival may also have a role in this model.

Extended Data Figure 3 CTCF-anchored loop in the PDGFRA region.

a, Schematic depiction of a HiC interaction signature of a CTCF-anchored loop domain, compared to an ordinary domain, as described previously15. CTCF-anchored loop domains are characterized by an increased interaction score at the apex of the domain, representing a CTCF–CTCF dimeric interaction. b, IMR90 HiC contact matrix for the PDGFRA/FIP1L1 locus, as presented in Fig. 3a. Solid circle indicates CTCF dimer interaction point; dashed circles indicate lack of CTCF dimeric anchor signature. c, IMR90 HiC contact matrix as in b, but with an expanded heatmap scale, more clearly conveys the CTCF-anchored loop that insulates PDGFRA. d, e, HiC contact matrix for GM12878 cells for the same region confirms a single CTCF-anchored loop (solid circle) between PDGFRA and FIP1L1. These data support the significance of this specific boundary in locus topology and PDGFRA insulation.

Extended Data Figure 4 Characterization of the FIP1L1 enhancer.

a, H3K27ac ChIP-seq track for GSC6 gliomaspheres reveals strong enrichment over the FIP1L1 enhancer. CTCF ChIP-seq track reveals location of the boundary element insulator (as in Fig. 3a). FIP1L1 enhancer (i) and promoter (ii) are indicated. b, H3K27ac ChIP-seq tracks for IDH mutant and wild-type gliomaspheres and glioma specimens reveal enrichment over the FIP1L1 enhancer. c, ChIP-seq tracks for glioma master transcription factors and other histone modifications support the enhancer identity of the element (H3K27ac, H3K4me1, SOX2, OLIG2; lacks H3K4me3, lacks H3K27me3). By contrast, the FIP1L1 promoter has a distinct ‘promoter-like’ chromatin state.

Extended Data Figure 5 Interaction of the FIP1L1 enhancer with nearby promoters and PDGFRA quantified by reciprocal 3C.

Top, the H3K27ac, CTCF and genetic architecture of the FIP1L1/PDGFRA locus is indicated, highlighting the 3C strategy. Bottom, plots indicate the interaction signal of the indicated sites (black lines) with the common enhancer primer. The FIP1L1 enhancer interacts with local promoters in wild-type and mutant tumours and models. In IDH wild-type gliomas, it shows essentially no interaction with the PDGFRA promoter. In IDH mutant gliomas, it interacts with the PDGFRA promoter with comparable strength to the local interactions, despite the much larger intervening distance (900 kb). Error bars reflect s.d.

Extended Data Figure 6 Crenolanib reverses the increased growth of PDGFRA insulator disrupted cells.

Insulator CRISPR-infected gliomaspheres exhibit a roughly twofold increase in proliferation rate, compared to control sgRNA-infected gliomaspheres. This proliferative advantage is eliminated by treatment with the PDGFRα inhibitor crenolanib. Crenolanib and dasatinib both inhibit PDGFRα, but their other targets are non-overlapping. Hence, this sensitivity provides further support that PDGFRA induction drives the increased proliferation of the insulator CRISPR gliomaspheres. Error bars reflect s.d.

Extended Data Figure 7 Signature of boundary deregulation in IDH mutant gliomas is robust.

Volcano plot depicts the significance (y axis) of gene pairs that are either more or less correlated in IDH mutant than IDH wild-type gliomas. This plot was generated by repeating the analysis in the main text and shown in Fig. 1f, except that here the statistics were performed using only the 14,055 genes expressed at >1 TPM in at least half of the samples. This indicates that the boundary deregulation signature in IDH mutant gliomas is not sensitive to noise from lowly expressed genes.

Extended Data Table 1 Clinical specimens and tumour models
Extended Data Table 2 Sequenced libraries characteristics

Supplementary information

Supplementary Table

This file contains a list of deregulated boundaries and genes. The table lists domains whose boundaries are predicted to be lost based on cross-boundary gene pairs that gain correlation and intra-domain gene pairs that lose correlation. All supporting gene pairs (FDR < 1%) are listed along with their significance. The overall significance of each domain is also shown. Genes that are up-regulated by at least 2-fold in IDH mutant gliomas, relative to the median of the wild-type, are indicated. For each domain, the coordinates of boundaries predicted to be lost are also listed. (XLSX 23 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Flavahan, W., Drier, Y., Liau, B. et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 529, 110–114 (2016). https://doi.org/10.1038/nature16490

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature16490

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