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:

ETV6 dependency in Ewing sarcoma by antagonism of EWS-FLI1-mediated enhancer activation

Abstract

The EWS-FLI1 fusion oncoprotein deregulates transcription to initiate the paediatric cancer Ewing sarcoma. Here we used a domain-focused CRISPR screen to implicate the transcriptional repressor ETV6 as a unique dependency in this tumour. Using biochemical assays and epigenomics, we show that ETV6 competes with EWS-FLI1 for binding to select DNA elements enriched for short GGAA repeat sequences. Upon inactivating ETV6, EWS-FLI1 overtakes and hyper-activates these cis-elements to promote mesenchymal differentiation, with SOX11 being a key downstream target. We show that squelching of ETV6 with a dominant-interfering peptide phenocopies these effects and suppresses Ewing sarcoma growth in vivo. These findings reveal targeting of ETV6 as a strategy for neutralizing the EWS-FLI1 oncoprotein by reprogramming of genomic occupancy.

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

Fig. 1: ETV6 is a dependency in Ewing sarcoma.
Fig. 2: ETV6 knockout in Ewing sarcoma drives mesenchymal differentiation.
Fig. 3: ETV6-mediated transcriptional repression is essential in Ewing sarcoma.
Fig. 4: ETV6 antagonizes EWS-FLI1-mediated enhancer activation at select DNA elements.
Fig. 5: ETV6 competes with EWS-FLI1 at short, interspersed GGAA repeats.
Fig. 6: Squelching of ETV6 with a SAM domain reprogrammes EWS-FLI1 occupancy and blocks Ewing sarcoma growth in vivo.

Similar content being viewed by others

Data availability

All the raw sequencing and related processed data are deposited in and publicly available from the GEO under the accession number GSE189418. These data include processed data such as peak region sets, ChIP signal and RNA-seq quantification. The cancer dependency dataset was obtained online (https://depmap.org/portal/download/, DepMap Public 21Q1). The Ewing sarcoma patient survival time and ETV6 expression level in the primary tumour were obtained from ICGC BOCA-FR dataset (https://dcc.icgc.org/projects/BOCA-FR, EXP-S). Numerical source data for Figs. 16 and Extended Data Figs. 17 and 9 are provided along with the unprocessed images of immunoblots for Figs. 1, 5 and 6 and Extended Data Figs. 1 and 59. Source data are provided with this paper.

Code availability

Customized Python script used to analyse GGAA motif in each peak is available on GitHub (https://github.com/ygaoyg/ETV6-dependency-in-Ewing-sarcoma.git).

References

  1. Ewing, J. Diffuse endothelioma of bone. CA Cancer J. Clin. 22, 95–98 (1972).

    Article  CAS  PubMed  Google Scholar 

  2. Grünewald, T. G. P. et al. Ewing sarcoma. Nat. Rev. Dis. Prim. 4, 5 (2018).

    Article  PubMed  Google Scholar 

  3. Stahl, M. et al. Risk of recurrence and survival after relapse in patients with Ewing sarcoma. Pediatr. Blood Cancer 57, 549–553 (2011).

    Article  PubMed  Google Scholar 

  4. Gaspar, N. et al. Ewing sarcoma: current management and future approaches through collaboration. J. Clin. Oncol. 33, 3036–3046 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Zucman, J. et al. Cloning and characterization of the Ewing’s sarcoma and peripheral neuroepithelioma t(11;22) translocation breakpoints. Genes Chromosomes Cancer 5, 271–277 (1992).

    Article  CAS  PubMed  Google Scholar 

  6. Delattre, O. et al. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature 359, 162–165 (1992).

    Article  CAS  PubMed  Google Scholar 

  7. Wasylyk, B., Hahn, S. L. & Giovane, A. The Ets family of transcription factors. EJB Rev. https://doi.org/10.1007/978-3-642-78757-7_2 (1993).

  8. Wilson, N. K. et al. Combinatorial transcriptional control in blood stem/progenitor cells: genome-wide analysis of ten major transcriptional regulators. Cell Stem Cell 7, 532–544 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Wei, G. et al. Genome‐wide analysis of ETS‐family DNA‐binding in vitro and in vivo. EMBO J. 29, 2147–2160 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Riggi, N. et al. EWS-FLI1 utilizes divergent chromatin remodeling mechanisms to directly activate or repress enhancer elements in Ewing sarcoma. Cancer Cell 26, 668–681 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gangwal, K. & Lessnick, S. L. Microsatellites are EWS/FLI response elements: genomic “junk” is EWS/FLI’s treasure. Cell Cycle 7, 3127–3132 (2008).

    Article  CAS  PubMed  Google Scholar 

  12. Gangwal, K. et al. Microsatellites as EWS/FLI response elements in Ewing’s sarcoma. Proc. Natl Acad. Sci. USA 105, 10149–10154 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. García-Aragoncillo, E. et al. DAX1, a direct target of EWS/FLI1 oncoprotein, is a principal regulator of cell-cycle progression in Ewing’s tumor cells. Oncogene 27, 6034–6043 (2008).

    Article  PubMed  Google Scholar 

  14. Guillon, N. et al. The oncogenic EWS-FLI1 protein binds in vivo GGAA microsatellite sequences with potential transcriptional activation function. PLoS ONE 4, e4932 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Boulay, G. et al. Cancer-specific retargeting of baf complexes by a prion-like domain. Cell 171, 163–178.e19 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gangwal, K., Close, D., Enriquez, C. A., Hill, C. P. & Lessnick, S. L. Emergent properties of EWS/FLI regulation via GGAA microsatellites in Ewing’s sarcoma. Genes Cancer 1, 177–187 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Patel, M. et al. Tumor-specific retargeting of an oncogenic transcription factor chimera results in dysregulation of chromatin and transcription. Genome Res. 22, 259–270 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Johnson, K. M., Taslim, C., Saund, R. S. & Lessnick, S. L. Identification of two types of GGAA-microsatellites and their roles in EWS/FLI binding and gene regulation in Ewing sarcoma. PLoS ONE 12, e0186275 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Ramakrishnan, R. et al. Role of protein–protein interactions in the antiapoptotic function of EWS-Fli-1. Oncogene 23, 7087–7094 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Nakatani, F. et al. Identification of p21 WAF1/CIP1 as a direct target of EWS-Fli1 oncogenic fusion protein. J. Biol. Chem. 278, 15105–15115 (2003).

    Article  CAS  PubMed  Google Scholar 

  21. Torchia, E. C., Jaishankar, S. & Baker, S. J. Ewing tumor fusion proteins block the differentiation of pluripotent marrow stromal cells. Cancer Res. 63, 3464–3468 (2003).

    CAS  PubMed  Google Scholar 

  22. Tirode, F. et al. Mesenchymal stem cell features of Ewing tumors. Cancer Cell 11, 421–429 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Lopez, R. G. et al. TEL is a sequence-specific transcriptional repressor. J. Biol. Chem. 274, 30132–30138 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Poirel, H. et al. The TEL gene products: nuclear phosphoproteins with DNA binding properties. Oncogene 14, 349–357 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Kim, C. A. et al. Polymerization of the SAM domain of TEL in leukemogenesis and transcriptional repression. EMBO J. 20, 4173–4182 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Green, S. M., Coyne, H. J., McIntosh, L. P. & Graves, B. J. DNA binding by the ETS protein TEL (ETV6) is regulated by autoinhibition and self-association. J. Biol. Chem. 285, 18496–18504 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang, L. C. et al. Yolk sac angiogenic defect and intra‐embryonic apoptosis in mice lacking the Ets‐related factor TEL. EMBO J. 16, 4374–4383 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang, L. C. et al. The TEL/ETV6 gene is required specifically for hematopoiesis in the bone marrow. Gene Dev. 12, 2392–2402 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Feurstein, S. & Godley, L. A. Germline ETV6 mutations and predisposition to hematological malignancies. Int. J. Hematol. 106, 189–195 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Biswas, A., Rajesh, Y., Mitra, P. & Mandal, M. ETV6 gene aberrations in non-haematological malignancies: a review highlighting ETV6 associated fusion genes in solid tumors. Biochim. Biophys. Acta Rev. Cancer 1874, 188389 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Braekeleer, E. D. et al. ETV6 fusion genes in hematological malignancies: a review. Leuk. Res. 36, 945–961 (2012).

    Article  PubMed  Google Scholar 

  32. Lu, B. et al. A transcription factor addiction in leukemia imposed by the MLL promoter sequence. Cancer Cell 34, 970–981.e8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Huang, Y.-H. et al. POU2F3 is a master regulator of a tuft cell-like variant of small cell lung cancer. Gene Dev. 32, 915–928 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gryder, B. E. et al. Histone hyperacetylation disrupts core gene regulatory architecture in rhabdomyosarcoma. Nat. Genet. 51, 1714–1722 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dempster, J. M. et al. Extracting biological insights from the Project Achilles genome-scale CRISPR screens in cancer cell lines. Preprint at bioRxiv https://doi.org/10.1101/720243 (2019).

  36. Meyers, R. M. et al. Computational correction of copy number effect improves specificity of CRISPR–Cas9 essentiality screens in cancer cells. Nat. Genet. 49, 1779–1784 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Tamir, A. et al. Fli-1, an Ets-related transcription factor, regulates erythropoietin-induced erythroid proliferation and differentiation: evidence for direct transcriptional repression of the Rb gene during differentiation. Mol. Cell. Biol. 19, 4452–4464 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Pereira, R. et al. FLI-1 inhibits differentiation and induces proliferation of primary erythroblasts. Oncogene 18, 1597–1608 (1999).

    Article  CAS  PubMed  Google Scholar 

  39. Athanasiou, M., Mavrothalassitis, G., Sun-Hoffman, L. & Blair, D. FLI-1 is a suppressor of erythroid differentiation in human hematopoietic cells. Leukemia 14, 439–445 (2000).

    Article  CAS  PubMed  Google Scholar 

  40. Potikyan, G. et al. Genetically defined EWS/FLI1 model system suggests mesenchymal origin of Ewing’s family tumors. Lab Invest. 88, 1291–1302 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Eliazer, S., Spencer, J., Ye, D., Olson, E. & Ilaria, R. L. Alteration of mesodermal cell differentiation by EWS/FLI-1, the oncogene implicated in Ewing’s sarcoma. Mol. Cell. Biol. 23, 482–492 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chia, W., Liu, J., Huang, Y.-G. & Zhang, C. A circular RNA derived from DAB1 promotes cell proliferation and osteogenic differentiation of BMSCs via RBPJ/DAB1 axis. Cell Death Dis. 11, 372 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Franzetti, G.-A. et al. Cell-to-cell heterogeneity of EWSR1-FLI1 activity determines proliferation/migration choices in Ewing sarcoma cells. Oncogene 36, 3505–3514 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chaturvedi, A., Hoffman, L. M., Welm, A. L., Lessnick, S. L. & Beckerle, M. C. The EWS/FLI oncogene drives changes in cellular morphology, adhesion, and migration in ewing sarcoma. Genes Cancer 3, 102–116 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hu-Lieskovan, S. et al. EWS-FLI1 fusion protein up-regulates critical genes in neural crest development and is responsible for the observed phenotype of Ewing’s family of tumors. Cancer Res. 65, 4633–4644 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Kwiatkowski, B. A. et al. The ets family member Tel binds to the Fli-1 oncoprotein and inhibits its transcriptional activity. J. Biol. Chem. 273, 17525–17530 (1998).

    Article  CAS  PubMed  Google Scholar 

  47. Gill, G. & Ptashne, M. Negative effect of the transcriptional activator GAL4. Nature 334, 721–724 (1988).

    Article  CAS  PubMed  Google Scholar 

  48. Sizemore, G. M., Pitarresi, J. R., Balakrishnan, S. & Ostrowski, M. C. The ETS family of oncogenic transcription factors in solid tumours. Nat. Rev. Cancer 17, 337–351 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Karim, F. D. et al. The ETS-domain: a new DNA-binding motif that recognizes a purine-rich core DNA sequence. Gene Dev. 4, 1451–1453 (1990).

    Article  CAS  PubMed  Google Scholar 

  50. Nye, J. A., Petersen, J. M., Gunther, C. V., Jonsen, M. D. & Graves, B. J. Interaction of murine ets-1 with GGA-binding sites establishes the ETS domain as a new DNA-binding motif. Gene Dev. 6, 975–990 (1992).

    Article  CAS  PubMed  Google Scholar 

  51. Kruse, E. A. et al. Dual requirement for the ETS transcription factors Fli-1 and Erg in hematopoietic stem cells and the megakaryocyte lineage. Proc. Natl Acad. Sci. USA 106, 13814–13819 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kwiatkowski, B. A., Zielinska-Kwiatkowska, A. G., Bauer, T. R. & Hickstein, D. D. The ETS family member Tel antagonizes the Fli-1 phenotype in hematopoietic cells. Blood Cells Mol. Dis. 26, 84–90 (2000).

    Article  CAS  PubMed  Google Scholar 

  53. Bose, R. et al. ERF mutations reveal a balance of ETS factors controlling prostate oncogenesis. Nature 546, 671–675 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Adane, B. et al. STAG2 loss rewires oncogenic and developmental programs to promote metastasis in Ewing sarcoma. Cancer Cell 39, 827–844.e10 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Crompton, B. D. et al. The genomic landscape of pediatric Ewing sarcoma. Cancer Discov. 4, 1326–1341 (2014).

    Article  CAS  PubMed  Google Scholar 

  56. Eckert, K. A. & Hile, S. E. Every microsatellite is different: Intrinsic DNA features dictate mutagenesis of common microsatellites present in the human genome. Mol. Carcinog. 48, 379–388 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Jawad, M. U. et al. Ewing sarcoma demonstrates racial disparities in incidence‐related and sex‐related differences in outcome. Cancer 115, 3526–3536 (2009).

    Article  PubMed  Google Scholar 

  58. Monument, M. J. et al. Clinical and biochemical function of polymorphic NR0B1 GGAA-microsatellites in Ewing sarcoma: a report from the Children’s Oncology Group. PLoS ONE 9, e104378 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Monument, M. J. et al. Microsatellites with macro-influence in Ewing sarcoma. Genes 3, 444–460 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Maurer, L. M. et al. BRCA1-associated RING domain-1 (BARD1) loss and GBP1 expression enhance sensitivity to DNA damage in Ewing sarcoma. Cancer Res. Commun. 2, 220–232 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Tarumoto, Y. et al. LKB1, salt-inducible kinases, and MEF2C are linked dependencies in acute myeloid leukemia. Mol. Cell 69, 1017–1027.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Almeida, Mde et al. AKIRIN2 controls the nuclear import of proteasomes in vertebrates. Nature 599, 491–496 (2021).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  64. 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).

    Article  CAS  PubMed  Google Scholar 

  65. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Feng, J., Liu, T., Qin, B., Zhang, Y. & Liu, X. S. Identifying ChIP–seq enrichment using MACS. Nat. Protoc. 7, 1728–1740 (2012).

    Article  CAS  PubMed  Google Scholar 

  70. Zhang, Y. et al. Model-based analysis of ChIP–seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Schrödinger, L. The PyMOL Molecular Graphics System, Version 2.0. (2019).

  75. Tran, H. H., Kim, C. A., Faham, S., Siddall, M.-C. & Bowie, J. U. Native interface of the SAM domain polymer of TEL. BMC Struct. Biol. 2, 5 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  76. De, S. et al. Steric mechanism of auto-inhibitory regulation of specific and non-specific DNA binding by the ETS transcriptional repressor ETV6. J. Mol. Biol. 426, 1390–1406 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by Cold Spring Harbor Laboratory NCI Cancer Center Support grants P30-CA045508 and 5P01CA013106-49. Additional funding was provided to C.R.V. by the Pershing Square Sohn Cancer Research Alliance, National Institutes of Health grants CA013106 and CA245859, Friends of T.J. Foundation, Christina Renna Foundation, Michelle Paternoster Foundation, and the William J. Riley Foundation, to M.E. by the National Institutes of Health (NIH) (5R01CA237413). K.M.B. is supported by the NCI award (K08CA252178). L.J. is an investigator of the Howard Hughes Medical Institute. X.-Y.H. is supported by the 2021 AACR-AstraZeneca Breast Cancer Research Fellowship (grant number 21-40-12-HE). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Y.G., X.-Y.H., X.S.W., Y.-H.H., S.T., J.J.I., T.H. and P.K.K., performed experiments and/or analysed the data; L.J.-T., K.M.B., M.E. and C.R.V. supervised the experiments and analysis; Y.G. and C.R.V. wrote the manuscript.

Corresponding author

Correspondence to Christopher R. Vakoc.

Ethics declarations

Competing interests

C.R.V. has received consulting fees from Flare Therapeutics, Roivant Sciences and C4 Therapeutics; has served on the advisory boards of KSQ Therapeutics, Syros Pharmaceuticals and Treeline Biosciences; has received research funding from Boehringer-Ingelheim and Treeline Biosciences; and owns a stock option from Treeline Biosciences. M.E. is a member of the research advisory board for brensocatib for Insmed, Inc.; a member of the scientific advisory board for Vividion Therapeutics, Inc.; a consultant for Protalix, Inc.; and holds shares in Agios Pharmaceuticals, Inc. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Thomas Grunewald, Tiebang Kang and Elizabeth Lawlor for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 ETV6 is a dependency in Ewing sarcoma cell lines.

(a) Scatterplot showing the correlation between patient survival time and ETV6 expression level in primary tumours from the ICGC BOCA-FR dataset containing 57 Ewing sarcoma patients. Blue lines mark the mean with 95% confidence interval. (b) Comparison of patient survival time between ETV6-high (Top 25%, n = 15) and ETV6-low patients (Bottom 25%, n = 15). Data are mean ± SEM. P value was calculated using two-tailed unpaired Student’s t-test. (c) Western blot of ETV6 and EWS-FLI1 levels confirming the knockout of ETV6 in different cancer cell lines. Beta-ACTIN (ACTB) was used as a loading control. (d) Photographs of collected tumours at end time point. (e) Western blot indicates that the resulting tumours from A673 xenograft experiments maintain similar levels of ETV6 and EWS-FLI1. ACTB was used as a loading control. (f) Representative images of immunofluorescence staining for PCNA (red) in the resulting tumours from A673 xenograft experiments (left panel). Percentage of PCNA-positive over DAPI-positive cells was quantified in the right panel. Data are mean ± SEM. P values were calculated using one-way ANOVA, Tukey’s multiple comparison tests. (n = 5 tumours) (g) Representative images of immunofluorescence staining for Cleaved Caspase-3 (red) in the resulting tumours from A673 xenografts (left panel). Percentage of Cleaved Caspase-3 positive over DAPI stained cells was quantified in the right panel. Data are mean ± SEM. P values were calculated using one-way ANOVA, Tukey’s multiple comparison tests. (n = 5 tumours).

Source data

Extended Data Fig. 2 ETV6 knockout in Ewing sarcoma drives mesenchymal differentiation.

(a) Representative images of immunofluorescence staining for collagen I (red), alpha smooth muscle actin (α-SMA, green) and the cytoskeleton component F-actin (white) in RH1 cells infected with indicated sgRNAs. (b, c) Representative images of immunofluorescence staining for alpha smooth muscle actin (α-SMA, red) and the cytoskeleton component F-actin (white) in two independent patient-derived Ewing sarcoma tumour cells PSaRC219 (b) and PSaRC318 (c) infected with indicated sgRNAs. (d) Quantification of cell size based on F-actin staining (μm2) in RH1, PSaRC219 and PSaRC318 cells. Data are mean ± SEM. P values were calculated using one-way ANOVA, Dunnett’s multiple comparison tests. (e) Representative images of immunofluorescence staining for collagen I (green) in the resulting tumours from A673 xenografts (left). Percentage of Collagen I stained area was quantified (right). (n = 5 tumours) Data are mean ± SEM. P values were calculated using one-way ANOVA, Dunnett’s multiple comparison tests.

Source data

Extended Data Fig. 3 ETV6 knockout up-regulates mesenchymal differentiation program in Ewing sarcoma cells, but not in non-Ewing cancer cell lines.

(a) Volcano plots showing the gene expression changes upon ETV6 knockout in 3 different non-Ewing cancer cell lines (RD, U2OS and SUIT2) assessed by RNA-seq. UP-regulated Genes (Log2FC > 0.5, p-adj<0.01, red); DOWN-regulated Genes (Log2FC < −0.5, p-adj<0.01, blue). (b) Venn diagram showing the overlap in up-regulated (Log2FC > 1) and down-regulated (Log2FC < −1) genes upon ETV6 knockout among three Ewing sarcoma cell lines (A673, RH1 and TC71). (c) Heatmap showing the expression changes of the mesenchymal differentiation genes, that are up-regulated upon ETV6 knockout in A673 cells, among 6 cancer cell lines (3 Ewing sarcoma and 3 non-Ewing lines). (d) Venn diagram showing the overlap between EWS-FLI1 repressed signature from Tirode et al. 2007. (n = 297, Log2FC > 1, P value<0.01) and the ETV6 repressed genes (n = 363, Log2FC > 1, P value<0.01) from our study in A673 cells.

Source data

Extended Data Fig. 4 ETV6 knockout does not enhance Ewing sarcoma cell migration or invasion phenotype.

(a) Representative images of migrated cells toward a high serum environment after 24 hours seeded in serum-free media in three Ewing sarcoma cell lines (A673, RH1 and TC71) (left). Comparison of the cell counts that migrate through the pores. Data are mean ± SEM. P values were calculated using one-way ANOVA, Tukey’s multiple comparison tests. (n = 3 biological replicates) (b) Representative images (left) and quantification (right) of trans-well invasion assay measuring the number of cells invade through a Matrigel matrix (24 hours). Data are mean ± SEM. P values were calculated using one-way ANOVA, Tukey’s multiple comparison tests. (n = 3 biological replicates).

Source data

Extended Data Fig. 5 ETV6 represses a unique set of genes that contributes to the dependency in Ewing sarcoma cells.

(a) Venn diagram showing the overlap of ETV6 ChIP-seq peaks between Ewing (A673) and non-Ewing cell lines. The ETV6 peaks in non-Ewing cells were annotated by merging ETV6 peaks from 3 different non-Ewing cancer cell lines. (b) The schematic diagram of genetic bypass screening. (c) Competition-based proliferation assay to validate the genetic bypass screening results using single sgRNAs. The sgNEG-mCherry control related to Fig. 3g. Data are mean ± SEM. (n = 3 biological replicates) (d) Competition-based proliferation assay to validate the genetic bypass screening results in two additional Ewing sarcoma cell lines (TC71 and RH1). Cells were infected with a dual-sgRNA linked to a GFP reporter. The percentage of cells that are positive for GFP were monitored during culturing. Data are mean ± SEM. P values were calculated using two-way ANOVA. (n = 3 biological replicates) (e)Western blot confirming the knockout of ETV6 and SOX11 in A673 cells prior to subcutaneous injections. ACTB was used as a loading control. (f) Average growth curves of ETV6 or/and SOX11, NTRK1 knockout A673 xenografts in immunodeficient mice. Data are mean ± SEM. Linear mixed-effects model with sgRNA, time and sgRNA by time interaction as fixed effects and sample specific random intercept was used to fit the longitudinal tumour volume data. Differences in tumour volume were examined using simultaneous tests for general linear hypotheses of contrasts of interest. P values were adjusted for multiple comparisons using the Bonferroni-Holm method. (sgNEG;sgNEG#2 (n = 11), sgNEG;sgETV6 (n = 12), sgETV6;sgSOX11 (n = 20), sgETV6;sgNTRK1 (n = 16)). (g) Weight of the resected tumours at the end point. Data are mean ± SEM. Log-transformed tumour weight in different groups were compared using Welch’s ANOVA test followed by Dunnett’s T3 multiple-comparison tests. (sgNEG;sgNEG#2 (n = 11), sgNEG;sgETV6 (n = 12), sgETV6;sgSOX11 (n = 20), sgETV6;sgNTRK1 (n = 16)).

Source data

Extended Data Fig. 6 Activation of ETV6 target genes specifically impairs Ewing sarcoma cell growth.

(a) RT-PCR analysis of SOX11 and NTRK1 levels in the CRISPR activation experiments. (-), no virus infection control. NT, non-targeting sgRNA control. Data are mean ± SEM. (n = 3 biological replicates) (b) Western blot confirming the elevation of SOX11 level upon CRISPR activation. ACTB was used as a loading control. (c) Competition-based proliferation assay evaluating the effects of candidate gene activation, using CRISPR activation (CRISPRa), to cell fitness in non-Ewing cancer cell lines (RD, U2OS and SUIT2). The percentage of GFP positive cells was monitored during culturing. Data are mean ± SEM. (n = 3 biological replicates) (d) Gene expression changes of SOX11 and NTRK1 in different cell lines assessed by RNA-seq. The significant expression changes (p < 0.05) were highlighted in red. (NA: mRNA not detected).

Source data

Extended Data Fig. 7 ETV6 antagonizes EWS-FLI1 function at select DNA elements.

(a) Venn diagram showing the overlap between ETV6 and EWS-FLI1 peaks in A673 cells. (b)Western blot assessing EWS-FLI1 and ETV6 levels in A673 cells transduced with indicated dual-sgRNA constructs. (c)Western blot of EWS-FLI1 and ETV6 in EWS-FLI1 reprogrammed RD cells. RD cells were transduced with EV (empty vector) control or EWS-FLI1 linked to indicated sgRNAs. (d) Metagene representation of the mean EWS-FLI1 signal differences at R sites versus microsatellites. (e) Metagene representation of the mean EWS-FLI1 signal differences at all ETV6 binding sites in A673 cells upon targeting ETV6. (f) Gene tracks of ETV6 and EWS-FLI1 ChIP-seq occupancy upon ETV6 knockout at DPEP1 and NTRK1 sites at two different scales. (g) Metagene representation of mean EWS-FLI1 ChIP-seq signal changes across R sites, microsatellites and all ETV6 binding regions in A673 cells upon targeting ETV6 using sgETV6 #2. (h) Western blot of ETV6 and EWS-FLI1 levels in patient-derived tumour cells (PSaRC219, left; PSaRC318, right) infected with indicated sgRNAs. ACTB was used as a loading control. (i) CellTiter Glo assay evaluating the effects of ETV6 knockout to proliferation in the patient-derived tumour cells (PSaRC219, left; PSaRC318, right). The relative luminescence fold changes (FC) to day 1 were plotted. Data are mean ± SEM. P values were calculated using two-way ANOVA. (n = 4 biological replicates) (j) EWS-FLI1 ChIP-qPCR analysis assessing the EWS-FLI1 occupancy changes at the ACTA2 site in the patient-derived Ewing tumour cells (PSaRC219 and PSaRC318) upon ETV6 knockout. A site 5 kb downstream of the ACTA2 site was used as a negative control. Data are mean ± SEM. P values were calculated using one-way ANOVA, Dunnett’s multiple comparison tests. (n = 3 biological replicates).

Source data

Extended Data Fig. 8 EWS-FLI1 does not bind to ETV6.

Immunoprecipitation assay showing ETV6 interacts with wild-type FLI1, but not EWS-FLI1 fusion. HEK293T cells were co-transfected with FLAG tagged ETV6 and HA tagged FLI1 or EWS-FLI1. FLAG IP was performed.

Source data

Extended Data Fig. 9 ETV6 oligomerization is essential in Ewing sarcoma.

(a) Illustration of ETV6 domain structure and SAM induction altering endogenous ETV6 function. (a.a: amino acid) (b) Western blot of A673 cells transduced with EV (empty vector), 3×FLAG tagged sgRNA resistant wild-type or (A94D, V113E) mutant ETV6 cDNA. Beta-ACTIN was used as a loading control. (c) Immunoprecipitation assay showing wild-type, but not (A94D, V113E) mutant SAM domain is able to associate with full-length ETV6. HEK293T cells were co-transfected with HA tagged ETV6 and FLAG tagged wild-type or (A94D, V113E) mutant SAM domain. FLAG IP was performed. (d) Western blot of A673 and RD cells transduced with Dox-inducible GFP or FLAG tagged SAM fragment. The samples were collected 48 hours after control or doxycycline treatment (1 μg/mL). ACTB was used as a loading control. (e) Metagene representation of the mean ETV6 and EWS-FLI1 signal changes upon SAM induction across microsatellites and all ETV6 binding sites. (f) Gene tracks of EWS-FLI1 ChIP-seq occupancy upon doxycycline treatment at ACTA2, DPEP1 and SOX11 sites in two different scales.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 1

Unprocessed western blots for Fig. 1.

Source Data Fig. 2

Statistical source data for Fig. 2.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Fig. 5

Statistical source data for Fig. 5.

Source Data Fig. 5

Unprocessed western blots for Fig. 5

Source Data Fig. 6

Statistical source data for Fig. 6.

Source Data Fig. 6

Unprocessed western blots for Fig. 6.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 1

Unprocessed western blots for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 5

Unprocessed western blots for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 6

Unprocessed western blots for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 7

Unprocessed western blots for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Unprocessed western blots for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 9

Unprocessed western blots for Extended Data Fig. 9.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gao, Y., He, XY., Wu, X.S. et al. ETV6 dependency in Ewing sarcoma by antagonism of EWS-FLI1-mediated enhancer activation. Nat Cell Biol 25, 298–308 (2023). https://doi.org/10.1038/s41556-022-01060-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-01060-1

This article is cited by

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