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.

  • Article
  • Published:

Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs

Abstract

Epigenetic aberrations are widespread in cancer, yet the underlying mechanisms and causality remain poorly understood1,2,3. A subset of gastrointestinal stromal tumours (GISTs) lack canonical kinase mutations but instead have succinate dehydrogenase (SDH) deficiency and global DNA hyper-methylation4,5. Here, we associate this hyper-methylation with changes in genome topology that activate oncogenic programs. To investigate epigenetic alterations systematically, we mapped DNA methylation, CTCF insulators, enhancers, and chromosome topology in KIT-mutant, PDGFRA-mutant and SDH-deficient GISTs. Although these respective subtypes shared similar enhancer landscapes, we identified hundreds of putative insulators where DNA methylation replaced CTCF binding in SDH-deficient GISTs. We focused on a disrupted insulator that normally partitions a core GIST super-enhancer from the FGF4 oncogene. Recurrent loss of this insulator alters locus topology in SDH-deficient GISTs, allowing aberrant physical interaction between enhancer and oncogene. CRISPR-mediated excision of the corresponding CTCF motifs in an SDH-intact GIST model disrupted the boundary between enhancer and oncogene, and strongly upregulated FGF4 expression. We also identified a second recurrent insulator loss event near the KIT oncogene, which is also highly expressed across SDH-deficient GISTs. Finally, we established a patient-derived xenograft (PDX) from an SDH-deficient GIST that faithfully maintains the epigenetics of the parental tumour, including hypermethylation and insulator defects. This PDX model is highly sensitive to FGF receptor (FGFR) inhibition, and more so to combined FGFR and KIT inhibition, validating the functional significance of the underlying epigenetic lesions. Our study reveals how epigenetic alterations can drive oncogenic programs in the absence of canonical kinase mutations, with implications for mechanistic targeting of aberrant pathways in 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

Fig. 1: Insulator dysfunction in SDH-deficient GISTs.
Fig. 2: FGF3FGF4 locus topology reorganized in SDH-deficient GISTs.
Fig. 3: KITPDGFRA locus topology reorganized in SDH-deficient GISTs.
Fig. 4: SDH-deficient GIST PDX trial confirms dependence on FGF signalling.

Similar content being viewed by others

Data availability

Sequencing data that support the findings of this study have been deposited in GEO with the accession code GSE107447.

References

  1. Kaelin, W. G. Jr & McKnight, S. L. Influence of metabolism on epigenetics and disease. Cell 153, 56–69 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    PubMed  PubMed Central  Google Scholar 

  4. Janeway, K. A. et al. Defects in succinate dehydrogenase in gastrointestinal stromal tumors lacking KIT and PDGFRA mutations. Proc. Natl Acad. Sci. USA 108, 314–318 (2011).

    ADS  CAS  PubMed  Google Scholar 

  5. Killian, J. K. et al. Succinate dehydrogenase mutation underlies global epigenomic divergence in gastrointestinal stromal tumor. Cancer Discov. 3, 648–657 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Dekker, J. & Mirny, L. The 3D genome as moderator of chromosomal communication. Cell 164, 1110–1121 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Hnisz, D. et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science 351, 1454–1458 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Xiao, M. et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 26, 1326–1338 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  Google Scholar 

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

    ADS  CAS  PubMed  Google Scholar 

  15. Liu, X. S. et al. Editing DNA methylation in the mammalian genome. Cell 167, 233–247.e17 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Flavahan, W. A. et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 529, 110–114 (2016).

    ADS  CAS  PubMed  Google Scholar 

  17. Hirota, S. et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 279, 577–580 (1998).

    ADS  CAS  PubMed  Google Scholar 

  18. Boikos, S. A. & Stratakis, C. A. The genetic landscape of gastrointestinal stromal tumor lacking KIT and PDGFRA mutations. Endocrine 47, 401–408 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Taguchi, T. et al. Conventional and molecular cytogenetic characterization of a new human cell line, GIST-T1, established from gastrointestinal stromal tumor. Lab. Invest. 82, 663–665 (2002).

    PubMed  Google Scholar 

  20. Mumbach, M. R. et al. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nat. Methods 13, 919–922 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Arao, T. et al. FGF3/FGF4 amplification and multiple lung metastases in responders to sorafenib in hepatocellular carcinoma. Hepatology 57, 1407–1415 (2013).

    CAS  PubMed  Google Scholar 

  22. Javidi-Sharifi, N. et al. Crosstalk between KIT and FGFR3 promotes gastrointestinal stromal tumor cell growth and drug resistance. Cancer Res. 75, 880–891 (2015).

    CAS  PubMed  Google Scholar 

  23. Li, F. et al. FGFR-mediated reactivation of MAPK signaling attenuates antitumor effects of imatinib in gastrointestinal stromal tumors. Cancer Discov. 5, 438–451 (2015).

    CAS  PubMed  Google Scholar 

  24. West, R. B. et al. The novel marker, DOG1, is expressed ubiquitously in gastrointestinal stromal tumors irrespective of KIT or PDGFRA mutation status. Am. J. Pathol. 165, 107–113 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Pal, S. K. et al. Efficacy of BGJ398, a fibroblast growth factor receptor 1-3 inhibitor, in patients with previously treated advanced urothelial carcinoma with FGFR3 alterations. Cancer Discov. 8, 812–821 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Janeway, K. A. et al. Sunitinib treatment in pediatric patients with advanced GIST following failure of imatinib. Pediatr. Blood Cancer 52, 767–771 (2009).

    PubMed  Google Scholar 

  27. Sircar, K. et al. Interstitial cells of Cajal as precursors of gastrointestinal stromal tumors. Am. J. Surg. Pathol. 23, 377–389 (1999).

    CAS  PubMed  Google Scholar 

  28. Gomez-Pinilla, P. J. et al. Ano1 is a selective marker of interstitial cells of Cajal in the human and mouse gastrointestinal tract. Am. J. Physiol. Gastrointest. Liver Physiol. 296, G1370–G1381 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Singh, R. D. et al. Ano1, a Ca2+-activated Cl- channel, coordinates contractility in mouse intestine by Ca2+ transient coordination between interstitial cells of Cajal. J. Physiol. 592, 4051–4068 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Urbini, M. et al. Gain of FGF4 is a frequent event in KIT/PDGFRA/SDH/RAS-P WT GIST. Genes Chromosom. Cancer 58, 636–642 (2019).

    CAS  PubMed  Google Scholar 

  31. 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 

  32. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    CAS  PubMed  Google Scholar 

  35. Ooi, W. F. et al. Epigenomic profiling of primary gastric adenocarcinoma reveals super-enhancer heterogeneity. Nat. Commun. 7, 12983 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cohen, A. J. et al. Hotspots of aberrant enhancer activity punctuate the colorectal cancer epigenome. Nat. Commun. 8, 14400 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. McDonald, O. G. et al. Epigenomic reprogramming during pancreatic cancer progression links anabolic glucose metabolism to distant metastasis. Nat. Genet. 49, 367–376 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Hovestadt, V. et al. Decoding the regulatory landscape of medulloblastoma using DNA methylation sequencing. Nature 510, 537–541 (2014).

    ADS  CAS  PubMed  Google Scholar 

  39. Servant, N. et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16, 259 (2015).

    PubMed  PubMed Central  Google Scholar 

  40. Zhou, X. et al. Exploring long-range genome interactions using the WashU Epigenome Browser. Nat. Methods 10, 375–376 (2013).

    CAS  PubMed  Google Scholar 

  41. Phanstiel, D. H. Sushi: Tools for visualizing genomics data. R package version 1.16.0. https://www.bioconductor.org/packages/release/bioc/html/Sushi.html (2019).

  42. Lareau, C. A. & Aryee, M. J. hichipper: a preprocessing pipeline for calling DNA loops from HiChIP data. Nat. Methods 15, 155–156 (2018).

    CAS  PubMed  Google Scholar 

  43. Splinter, E., de Wit, E., van de Werken, H. J., Klous, P. & de Laat, W. Determining long-range chromatin interactions for selected genomic sites using 4C-seq technology: from fixation to computation. Methods 58, 221–230 (2012).

    CAS  PubMed  Google Scholar 

  44. van de Werken, H. J. et al. Robust 4C-seq data analysis to screen for regulatory DNA interactions. Nat. Methods 9, 969–972 (2012).

    PubMed  Google Scholar 

  45. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  46. Hemming, M. L. et al. Gastrointestinal stromal tumor enhancers support a transcription factor network predictive of clinical outcome. Proc. Natl Acad. Sci. USA 115, E5746–E5755 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Wagner, G. P., Kin, K. & Lynch, V. J. Measurement of mRNA abundance using RNA-seq data: RPKM measure is inconsistent among samples. Theory Biosci. 131, 281–285 (2012).

    CAS  PubMed  Google Scholar 

  48. GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

    Google Scholar 

  49. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Grant, C. E., Bailey, T. L. & Noble, W. S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017–1018 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Mathelier, A. et al. JASPAR 2014: an extensively expanded and updated open-access database of transcription factor binding profiles. Nucleic Acids Res. 42, D142–D147 (2014).

    CAS  PubMed  Google Scholar 

  52. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  53. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  55. Wagle, M. C. et al. A transcriptional MAPK pathway activity score (MPAS) is a clinically relevant biomarker in multiple cancer types. NPJ Precis. Oncol. 7, 2 (2018).Correct publication information for this reference?

  56. Chi, P. et al. ETV1 is a lineage survival factor that cooperates with KIT in gastrointestinal stromal tumours. Nature 467, 849–853 (2010).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  58. Manning, G., Whyte, D. B., Martinez, R., Hunter, T. & Sudarsanam, S. The protein kinase complement of the human genome. Science 298, 1912–1934 (2002).

    ADS  CAS  PubMed  Google Scholar 

  59. Yu, G. et al. ggtree: an R package for visualization and annotation of phylogenetic trees with their covariates and other associated data. Methods Ecol. Evol. 8, 28–36 (2017).

    Google Scholar 

  60. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    ADS  Google Scholar 

  61. Lee, M. Y. et al. Transcriptome of interstitial cells of Cajal reveals unique and selective gene signatures. PLoS One 12, e0176031 (2017).

    PubMed  PubMed Central  Google Scholar 

  62. Killian, J. K. et al. Recurrent epimutation of SDHC in gastrointestinal stromal tumors. Sci. Transl. Med. 6, 268ra177 (2014).

    PubMed  PubMed Central  Google Scholar 

  63. Tang, C. M. et al. Hedgehog pathway dysregulation contributes to the pathogenesis of human gastrointestinal stromal tumors via GLI-mediated activation of KIT expression. Oncotarget 7, 78226–78241 (2016).

    PubMed  PubMed Central  Google Scholar 

  64. Ostrowski, J. et al. Functional features of gene expression profiles differentiating gastrointestinal stromal tumours according to KIT mutations and expression. BMC Cancer 9, 413 (2009).

    PubMed  PubMed Central  Google Scholar 

  65. Astolfi, A. et al. A molecular portrait of gastrointestinal stromal tumors: an integrative analysis of gene expression profiling and high-resolution genomic copy number. Lab. Invest. 90, 1285–1294 (2010).

    CAS  PubMed  Google Scholar 

  66. Gautier, L., Cope, L., Bolstad, B. M. & Irizarry, R. A. affy—analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20, 307–315 (2004).

    CAS  PubMed  Google Scholar 

  67. Li, Z. et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 15, 501–513 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Murakami, A. et al. Hypoxia increases gefitinib-resistant lung cancer stem cells through the activation of insulin-like growth factor 1 receptor. PLoS One 9, e86459 (2014).

    ADS  PubMed  PubMed Central  Google Scholar 

  69. Bosch-Marce, M. et al. Effects of aging and hypoxia-inducible factor-1 activity on angiogenic cell mobilization and recovery of perfusion after limb ischemia. Circ. Res. 101, 1310–1318 (2007).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. Gillespie, M. Miri, F. Najm, P. van Galen and E. Choy for assistance with clinical samples and analysis, H. Gu and A. Gnirke for sequencing assistance, and E. Gaskell for discussions. W.A.F. is supported by an F32 from the National Cancer Institute. Y.D. is supported by the Tosteson Postdoctoral Fellowship. B.E.B. is the Bernard and Mildred Kayden Endowed MGH Research Institute Chair and an American Cancer Society Research Professor. This research was supported by the National Cancer Institute, the NIH Common Fund, the Starr Cancer Consortium, and the Ludwig Center at Harvard.

Author information

Authors and Affiliations

Authors

Contributions

Conception and experimental design: W.A.F., Y.D., S.E.J., M.L.H., E.T.S., G.D.D. and B.E.B. Methodology and data acquisition: W.A.F., Y.D., S.E.J., M.L.H., D.R.T., E.H., S.J.S., N.M.J., C.P.R., B.K.E., P.C.G., J.L.H., E.T.S., G.D.D. and B.E.B. Analysis and interpretation of data: W.A.F., Y.D., M.L.H., S.J.S., N.M.J., G.D.D. and B.E.B. Manuscript writing and revision: W.A.F., Y.D. and B.E.B.

Corresponding authors

Correspondence to Yotam Drier, George D. Demetri or Bradley E. Bernstein.

Ethics declarations

Competing interests

B.E.B. is an advisor and equity holder for Fulcrum Therapeutics, 1CellBio, HiFiBio and Arsenal Biosciences, is an advisor for Cell Signaling Technologies, and has equity in Nohla Therapeutics. G.D.D. reports relationships with Novartis, Bayer, Pfizer, EMD-Serono, Sanofi, Ignyta, Roche, Loxo Oncology, AbbVie, Mirati Therapeutics, Epizyme, Daiichi-Sankyo, WIRB Copernicus Group, ZioPharm, Polaris Pharmaceuticals, M.J. Hennessey / OncLive, Adaptimmune, GlaxoSmithKline, Blueprint Medicines, Merrimack Pharmaceuticals, G1 Therapeutics, CARIS Life Sciences, Bessor Pharmaceuticals, ERASCA Pharmaceuticals, CHAMPIONS Oncology, Janssen, PharmaMar; in addition, G.D.D. has a Use patent on imatinib for GIST, licensed to Novartis, with royalties paid to the Dana-Farber Cancer Institute.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Peer review information Nature thanks Christian Frezza, Michael Heinrich, Michael Rehli and Peter Scacheri for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Fig. 1 Epigenomic characterization of GIST.

a, ChIP–seq profiles for H3K27ac were compared for GIST specimens and other gastrointestinal tract tumour specimens (GAC, gastric adenocarcinoma; CRC, colorectal cancer; PDAC, pancreatic ductal adenocarcinoma). Heat map depicts pairwise Pearson correlations between the top 10,000 most variable peaks (yellow indicates high correlation; blue indicates low correlation). The dendrogram (left) was derived by unweighted average distance linkage. Enhancer patterns are relatively consistent across GIST subtypes, compared to other tumour types. b, DNA methylation levels in the vicinity of CTCF sites were profiled genome-wide by hybrid-selection bisulfite sequencing. CTCF sites are binned according to the amount their methylation increased in SDH-deficient GISTs, relative to SDH-intact GISTs (methylation change computed over a 250 bp window centred on the motif). For each bin, bar graphs depict the percentage of sites that lose CTCF binding in SDH-deficient GISTs, per ChIP–seq. Separate plots are shown for CTCF sites for which motifs do or do not contain a CpG. Increased methylation over CTCF sites is associated with more frequent loss of CTCF binding, even when the CTCF motif lacks a CpG. c, Plot depicts correlation between CTCF occupancy and DNA methylation in SDH-deficient GISTs. Red points show Spearman correlations between CTCF ChIP–seq signal and methylation of CpGs at indicated positions relative to the centre of the CTCF motif. Red line reflects correlation to average methylation over 10 bp windows. Randomly permuted data (black) are shown for comparison. Anti-correlation between CTCF occupancy and methylation is evident over a ~250-bp binding footprint. d, Genomic views of a representative 10 Mb region on chromosome 21 depict chromosome topology (HiC, red), CTCF binding (ChIP–seq, orange) and CTCF–CTCF loop interactions (HiChIP, black) for the SDH-intact GIST model, GIST-T1. TADs are visible as triangles of enhanced interaction in HiC data, flanked by boundaries that correspond to loop interactions in HiChIP data. Genes (blue) are also indicated.

Extended Data Fig. 2 Super-enhancers and insulators in GIST.

a, Traces depict H3K27ac ChIP–seq signal for normal stomach muscle (NSM) and GISTs of the indicated subtype over the FGF–ANO1 locus. b, Traces depict H3K27ac ChIP–seq signal for NSM and GISTs of the indicated subtype over the PDGFRA–KIT locus. Genes are indicated in blue, and super-enhancer locations are indicated by green bars. For a, b, traces are representative of 11 KIT-mutant and 6 SDH-deficient tumours with similar results. c, Traces depict CTCF binding over the FGF insulator in normal stomach muscle (NSM) and GIST clinical specimens. d, Plot depicts CTCF ChIP–seq signal over the strongest CTCF peak in the FGF insulator in normal stomach muscle (NSM, n = 4), and KIT mutant (n = 11), PDGFRA mutant (n = 2) and SDH-deficient GISTs (n = 6). e, Traces depict CTCF binding over the KIT insulator in normal stomach muscle (NSM) and GIST clinical specimens. f, Plot depicts CTCF ChIP–seq signal over the strongest CTCF peak in the KIT insulator in normal stomach muscle (NSM, n = 4), and KIT mutant (n = 11), PDGFRA mutant (n = 2) and SDH-deficient GISTs (n = 6). For d and f, horizontal bars reflect mean values and P values indicate significance of CTCF loss in SDH-deficient GIST, as determined by the Walt test (via DEseq254). All n values represent the number of biologically independent clinical specimens.

Extended Data Fig. 3 FGF locus 4C-seq data and insulator deletion.

a, Traces depict 4C-seq data at FGF locus, as in Fig. 2b, except graphed on the same axis to allow for direct comparison. b, c, Bar plots quantify 4C-seq interactions between the super-enhancer viewpoint and FGF4 (b) or FGF3 (c). Expression of these genes in the corresponding SDH-deficient GIST specimens is also shown. d, Traces depict CTCF ChIP–seq signal in GIST-T1 cells infected with CRISPR–Cas9 and either a control sgRNA directed at GFP (black, top) or sgRNAs directed against the two indicated CTCF motifs in the FGF insulator (second row, red).

Extended Data Fig. 4 Allelic imbalance in FGF3 and FGF4 activation.

a, Two heterozygous SNPs in FGF4 (both 3′ UTR) enabled us to evaluate allelic expression in three SDH-deficient GISTs (tumours S6, S1 and S4). Both alleles for each SNP were detected in DNA sequencing data for these tumours, but only one allele was detected in RNA-seq data of tumours S1 and S6, indicative of mono-allelic FGF4 expression. Both alleles are detected in tumour S4, indicating bi-allelic expression of FGF4. b, Heterozygous SNPs in FGF3 exons (both synonymous base substitutions) enabled us to evaluate allelic expression in the SDH-deficient GISTs (tumours S2 and S5). In both cases, DNA sequencing confirmed heterozygosity at the genome level (C/A and T/C, respectively), but RNA-seq data demonstrated mono-allelic FGF3 expression. c, Both alleles of heterozygous SNPs in ANO1 exons were found in the RNA-seq data derived from SDH-deficient GIST samples, confirming bi-allelic expression of ANO1. Similarly, both alleles of heterozygous SNPs were found in the histone H3K27ac ChIP–seq data, confirming the bi-allelic nature of the super-enhancer (not shown). d, One SDH-deficient GIST sample was heterozygous for a SNP (rs386829467) located about 50 bp from the CTCF motif of Peak 2 in the FGF insulator. Allele-agnostic methylation data confirmed 43% methylation of the CTCF peak in this tumour, while essentially no methylation was detected in the SDH-intact tumours (left). Separation of the two alleles using the heterozygous SNP revealed strong allelic bias in the SDH-deficient tumour: one allele was largely unmethylated (~3% methylation), while the other was highly methylated (~75% methylation), consistent with mono-allelic methylation of the CTCF site (right). e, Schematic depicts 4C-seq experimental protocol and primer design for detecting SNPs. DNA elements in close physical proximity are crosslinked and restricted with an enzyme that leaves nucleotide overhangs. These overhangs are then proximity ligated to crosslinked fragments. A second restriction enzyme (with different restriction sites) is then used to circularize the ligated fragments, allowing for inverse PCR. Here we selected restriction enzymes and designed a custom read 2 primer to capture a heterozygous SNP within the super-enhancer. This second read is normally non-informative as contact frequencies are determined through the viewpoint primer (read 1), but in this case enabled us to detect the SNP and assign each ligated fragment to a specific allele. f, The left trace (grey) depicts standard 4C-seq data (allele agnostic), which demonstrates strong interaction between super-enhancer viewpoint and FGF4. However, the SNP covered in the non-viewpoint read enabled us to distinguish interactions involving the minor (top right) or major (bottom right) allele. This revealed that the major allele (purple) is responsible for ~97% of super-enhancer–FGF4 interactions.

Extended Data Fig. 5 KIT locus 4C-seq data and insulator deletion.

a, Traces depict KIT locus 4C-seq data, as in Fig. 3b, except graphed on the same axis to allow for direct comparison. bd, Bar plots quantify 4C-seq interactions (top, reproduced from Fig. 3b) between the super-enhancer viewpoint and positions within the super-enhancer TAD (b), sequences just beyond the KIT insulator (c), or the KIT gene itself (d). P values indicate significance of difference between SDH-intact and SDH-deficient, by two-sided t-test. e, Traces depict CTCF ChIP–seq signal in GIST-T1 cells infected with Cas9 and either a control sgRNA directed at GFP (black, top) or sgRNAs directed against the two bound CTCF motifs in the KIT insulator (second row, red).

Extended Data Fig. 6 Hypoxia marker induction in GIST-T1 cells.

a, Bar plot depicts levels of the TET product, 5-hydroxymethyl-cytosine (5-hmC), measured by ELISA in GIST-T1 cells infected with CRISPR–Cas9 and either a short guide RNA targeting GFP (sgGFP) or SDHB. Cells were cultured in either control media or media supplemented with 20 μM of dimethylsuccinate (DMS), a membrane-permeable ester of succinate, for 3 days, as indicated. b, c, Plots show relative expression of pseudo-hypoxia-associated genes EPAS1 (also known as HIF2A)67 and IGF1R68 (b), and KITLG (also known as SCF)69 (c) in control GIST-T1 cells (black), SDH-deficient GIST-T1 cells generated by CRISPR–Cas9 knockout of SDHB and cultured with exogenous succinate (red), or GIST-T1 cells treated with the iron chelator DFX to simulate hypoxia (blue). Upregulation of KIT ligand due to pseudo-hypoxia or tumour hypoxia may supplement FGF ligands in promoting RTK signalling in SDH-deficient GIST.

Extended Data Fig. 7 FGF and KIT insulator methylation and expression in GIST subtypes and non-malignant cells.

a, Bar plot depicts methylation of FGF insulator CTCF peak 2 (top) and KIT insulator CTCF peak 2 (bottom) in 34 tissues and primary cells available through ENCODE60. Values are average methylation of CpGs nearest the CTCF motifs, determined by whole genome bisulfite sequencing (WGBS) (KIT insulator, 3 CpGs; FGF insulator, 6 CpGs). Methylation of these sites is also shown for SDH-intact and SDH-deficient GISTs (see Fig. 3e, f), and for flow-sorted CD45ANO1+KIT+ (ICC enriched) cells from normal stomach muscle (NSM) tissue (n values represent biologically independent specimens). b, Left, table depicts FPKM (fragments per kilobase of transcript per million mapped reads) values of relevant genes in mouse ICCs isolated from jejunum or colon61. Right, dot plot depicts expression of FGF4 in either flow sorted ICCs (green) or GISTs of the indicated subtype: KIT mutant in black, PDGFRA mutant in blue, SDH-deficient in red, and KIT-/PDGFRA wild type in purple62,63,64,65,66. SDH status of the latter group is unknown, but SDH-deficient GIST represent a significant portion of KIT-/PDGFRA wild-type tumours. Data are drawn from the indicated GEO series and publications.

Extended Data Fig. 8 PDX trial of FGFR and KIT combination therapy in SDH-deficient GIST.

a, b, Genomic views depict RNA expression (black), H3K27ac (green) and CTCF occupancy (orange) over the FGF (a) and KIT (b) loci for the S1 primary tumour and PDX. Genes (blue), super-enhancers (green bar and shade) and lost CTCF insulators (orange shade) are indicated. c, Plot depicts tumour volume during treatment and observation periods of experiment, as in Fig. 4g, except with time axis extended until final group reached censor point (one tumour in the group >2,000 mm3). Points represent mean tumour size, error bars represent s.e.m., and shading represents range of tumour sizes for n = 8 biologically independent xenograft-bearing mice per group. For statistics, see Fig. 4g. d, Kaplan–Meier plot depicts survival until clinical endpoint (tumour size >2,000 mm3) for the same PDX trial. Median and range survival are indicated for each group. P values reflect difference in survival between groups as calculated by logrank test.

Supplementary information

Reporting Summary

Supplementary Information Table 1

| Clinical Specimens Analysed Table lists all of the clinical specimens analysed in the manuscript, noting tissue type, source (FFPE vs. fresh frozen), primary site and metastatic status (if known), driver mutations (if known), and experiments performed on the tissue. For FFPE tissue, no genetic testing was performed; instead SDH status was determined by IHC

Supplementary Information Table 2

| Candidate deregulated genes due to insulator loss Table lists results of analysis of genes potentially regulated by insulator loss (see Fig. 1e). Listed are methylation difference at lost insulators and median expression in GIST of various driver mutations

Supplementary Information Table 3

| PDX Tumor Growth Data Table lists measured volumes of Patient Derived Xenograft (PDX) trial (see Fig. 4f, g, Extended Data Fig. 8.) Volumes are listed as mm3 with each column representing serial measurements of a single biologically-independent PDX-bearing mouse; for further details of study, see Methods

Supplementary Information Table 4

| ChIP–seq information Table lists information from ChIPseq runs associated with the work, including sample, antibody, processed data file name(s), instrument, read count, and sequencing strategy (e.g. paired/single end)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Flavahan, W.A., Drier, Y., Johnstone, S.E. et al. Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs. Nature 575, 229–233 (2019). https://doi.org/10.1038/s41586-019-1668-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1668-3

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