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.

  • Brief Communication
  • Published:

Spontaneous expression of the CIC::DUX4 fusion oncoprotein from a conditional allele potently drives sarcoma formation in genetically engineered mice

Abstract

CIC::DUX4 sarcoma (CDS) is a rare but highly aggressive undifferentiated small round cell sarcoma driven by a fusion between the tumor suppressor Capicua (CIC) and DUX4. Currently, there are no effective treatments and efforts to identify and translate better therapies are limited by the scarcity of patient tumor samples and cell lines. To address this limitation, we generated three genetically engineered mouse models of CDS (Ch7CDS, Ai9CDS, and TOPCDS). Remarkably, chimeric mice from all three conditional models developed spontaneous soft tissue tumors and disseminated disease in the absence of Cre-recombinase. The penetrance of spontaneous (Cre-independent) tumor formation was complete irrespective of bi-allelic Cic function and the distance between adjacent loxP sites. Characterization of soft tissue and presumed metastatic tumors showed that they consistently expressed the CIC::DUX4 fusion protein and many downstream markers of the disease credentialing the models as CDS. In addition, tumor-derived cell lines were generated and ChIP-seq was preformed to map fusion-gene specific binding using an N-terminal HA epitope tag. These datasets, along with paired H3K27ac ChIP-sequencing maps, validate CIC::DUX4 as a neomorphic transcriptional activator. Moreover, they are consistent with a model where ETS family transcription factors are cooperative and redundant drivers of the core regulatory circuitry in CDS.

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: Fusion of a human DUX4 C-terminal domain to endogenous Cic is sufficient to generate small round cell sarcomas.
Fig. 2: Cic haploinsufficiency is not required for CIC::DUX4 sarcomagenesis.
Fig. 3: Mouse tumors express CIC::DUX4 and a transcriptional signature consistent with CDS.
Fig. 4: CIC::DUX4 behaves as a neomorphic transcriptional activator.

Similar content being viewed by others

Data availability

All RNA-sequencing and ChIP-sequencing data is freely accessible through the Gene Expression Omnibus (GEO) under the series ID: GSE241371.

References

  1. Yoshimoto M, Graham C, Chilton-MacNeill S, Lee E, Shago M, Squire J, et al. Detailed cytogenetic and array analysis of pediatric primitive sarcomas reveals a recurrent CIC-DUX4 fusion gene event. Cancer Genet Cytogenet. 2009;195:1–11.

    Article  CAS  PubMed  Google Scholar 

  2. Italiano A, Sung YS, Zhang L, Singer S, Maki RG, Coindre J-M, et al. High prevalence of CIC fusion with double-homeobox (DUX4) transcription factors in EWSR1-negative undifferentiated small blue round cell sarcomas. Genes Chromosomes Cancer. 2012;51:207–18.

    Article  CAS  PubMed  Google Scholar 

  3. Antonescu CR, Owosho AA, Zhang L, Chen S, Deniz K, Huryn JM, et al. Sarcomas With CIC-rearrangements are a distinct pathologic entity with aggressive outcome: a clinicopathologic and molecular study of 115 cases. Am J Surg Pathol. 2017;41:941–9.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Hendrickson PG, Doráis JA, Grow EJ, Whiddon JL, Lim J-W, Wike CL, et al. Conserved roles of mouse DUX and human DUX4 in activating cleavage-stage genes and MERVL/HERVL retrotransposons. Nat Genet. 2017;49:925–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kawamura-Saito M, Yamazaki Y, Kaneko K, Kawaguchi N, Kanda H, Mukai H, et al. Fusion between CIC and DUX4 up-regulates PEA3 family genes in Ewing-like sarcomas with t(4;19)(q35;q13) translocation. Hum Mol Genet. 2006;15:2125–37.

    Article  CAS  PubMed  Google Scholar 

  6. Yoshimoto T, Tanaka M, Homme M, Yamazaki Y, Takazawa Y, Antonescu CR, et al. CIC-DUX4 induces small round cell sarcomas distinct from ewing sarcoma. Cancer Res. 2017;77:2927–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wisdom AJ, Mowery YM, Hong CS, Himes JE, Nabet BY, Qin X, et al. Single cell analysis reveals distinct immune landscapes in transplant and primary sarcomas that determine response or resistance to immunotherapy. Nat Commun. 2020;11:6410.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Connolly EA, Bhadri VA, Wake J, Ingley KM, Lewin J, Bae S, et al. Systemic treatments and outcomes in CIC-rearranged Sarcoma: A national multi-centre clinicopathological series and literature review. Cancer Med. 2022;11:1805–16.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Graham C, Chilton-MacNeill S, Zielenska M, Somers GR. The CIC-DUX4 fusion transcript is present in a subgroup of pediatric primitive round cell sarcomas. Hum Pathol. 2012;43:180–9.

    Article  CAS  PubMed  Google Scholar 

  10. Leidenroth A, Hewitt JE. A family history of DUX4: phylogenetic analysis of DUXA, B, C and Duxbl reveals the ancestral DUX gene. BMC Evol Biol. 2010;10:364.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chebib I, Jo VY. Round cell sarcoma with CIC-DUX4 gene fusion: Discussion of the distinctive cytomorphologic, immunohistochemical, and molecular features in the differential diagnosis of round cell tumors. Cancer Cytopathol. 2016;124:350–61.

    Article  CAS  PubMed  Google Scholar 

  12. Choi E-YK, Thomas DG, McHugh JB, Patel RM, Roulston D, Schuetze SM, et al. Undifferentiated Small Round Cell Sarcoma With t(4;19)(q35;q13.1) CIC-DUX4 Fusion: A Novel Highly Aggressive Soft Tissue Tumor With Distinctive Histopathology. Am J Surg Pathol. 2013;37:1379–86.

    Article  PubMed  Google Scholar 

  13. Specht K, Sung Y-S, Zhang L, Richter GHS, Fletcher CD, Antonescu CR. Distinct transcriptional signature and immunoprofile of CIC-DUX4 fusion-positive round cell tumors compared to EWSR1 -rearranged ewing sarcomas: Further evidence toward distinct pathologic entities: Gene Expression in CIC-DUX4 Sarcomas. Genes Chromosomes Cancer. 2014;53:622–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Simón-Carrasco L, Jiménez G, Barbacid M, Drosten M. The Capicua tumor suppressor: a gatekeeper of Ras signaling in development and cancer. Cell Cycle Georget Tex. 2018;17:702–11.

    Article  Google Scholar 

  15. Kim JW, Ponce RK, Okimoto RA. Capicua in Human Cancer. Trends Cancer. 2021;7:77–86.

    Article  CAS  PubMed  Google Scholar 

  16. Zheng B, Sage M, Sheppeard EA, Jurecic V, Bradley A. Engineering mouse chromosomes with Cre-loxP: range, efficiency, and somatic applications. Mol Cell Biol. 2000;20:648–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jackson EL, Willis N, Mercer K, Bronson RT, Crowley D, Montoya R, et al. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev. 2001;15:3243–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Politi K, Kljuic A, Szabolcs M, Fisher P, Ludwig T, Efstratiadis A. Designer’ tumors in mice. Oncogene. 2004;23:1558–65.

    Article  CAS  PubMed  Google Scholar 

  19. Le Guellec S, Velasco V, Pérot G, Watson S, Tirode F, Coindre J-M. ETV4 is a useful marker for the diagnosis of CIC-rearranged undifferentiated round-cell sarcomas: a study of 127 cases including mimicking lesions. Mod Pathol. 2016;29:1523–31.

    Article  PubMed  Google Scholar 

  20. Siegele B, Roberts J, Black JO, Rudzinski E, Vargas SO, Galambos C. DUX4 Immunohistochemistry Is a Highly Sensitive and Specific Marker for CIC-DUX4 Fusion-positive Round Cell Tumor. Am J Surg Pathol. 2017;41:423–9.

    Article  PubMed  Google Scholar 

  21. Lin YK, Wu W, Ponce RK, Kim JW, Okimoto RA. Negative MAPK-ERK regulation sustains CIC-DUX4 oncoprotein expression in undifferentiated sarcoma. Proc Natl Acad Sci USA. 2020;117:20776–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kirsch DG, Dinulescu DM, Miller JB, Grimm J, Santiago PM, Young NP, et al. A spatially and temporally restricted mouse model of soft tissue sarcoma. Nat Med. 2007;13:992–7.

    Article  CAS  PubMed  Google Scholar 

  23. The Tabula Muris Consortium, Overall coordination, Logistical coordination, Organ collection and processing, Library preparation and sequencing, Computational data analysis. et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature. 2018;562:367–72.

    Article  Google Scholar 

  24. Comitani F, Nash JO, Cohen-Gogo S, Chang AI, Wen TT, Maheshwari A, et al. Diagnostic classification of childhood cancer using multiscale transcriptomics. Nat Med. 2023;29:656–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Okimoto RA, Wu W, Nanjo S, Olivas V, Lin YK, Ponce RK, et al. CIC-DUX4 oncoprotein drives sarcoma metastasis and tumorigenesis via distinct regulatory programs. J Clin Invest. 2019;129:3401–6.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Kim JW, Luck C, Wu W, Ponce RK, Lin YK, Gupta N, et al. Capicua suppresses YAP1 to limit tumorigenesis and maintain drug sensitivity in human cancer. Cell Rep. 2022;41:111443.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Carrabotta M, Laginestra MA, Durante G, Mancarella C, Landuzzi L, Parra A, et al. Integrated Molecular Characterization of Patient-Derived Models Reveals Therapeutic Strategies for Treating CIC-DUX4 Sarcoma. Cancer Res. 2022;82:708–20.

    Article  CAS  PubMed  Google Scholar 

  28. Watson S, Kendall GC, Rakheja D, McFaul ME, Draper BW, Tirode F, et al. CIC-DUX4 expression drives the development of small round cell sarcoma in transgenic zebrafish: a new model revealing a role for ETV4 in CIC-mediated sarcomagenesis. Cancer Biology, https://doi.org/10.1101/517722. (2019).

  29. Mancarella C, Carrabotta M, Toracchio L, Scotlandi K. CIC-rearranged sarcomas: an intriguing entity that may lead the way to the comprehension of more common cancers. Cancers. 2022;14:5411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Johnson L, Mercer K, Greenbaum D, Bronson RT, Crowley D, Tuveson DA, et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature. 2001;410:1111–6.

    Article  CAS  PubMed  Google Scholar 

  31. Bosnakovski D, Ener ET, Cooper MS, Gearhart MD, Knights KA, Xu NC, et al. Inactivation of the CIC-DUX4 oncogene through P300/CBP inhibition, a therapeutic approach for CIC-DUX4 sarcoma. Oncogenesis. 2021;10:68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Durall RT, Huang J, Wojenski L, Huang Y, Gokhale PC, Leeper BA, et al. The BRD4–NUT fusion alone drives malignant transformation of NUT Carcinoma. Cancer Res. 2023;83:3846–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mesquita D, Barros-Silva JD, Santos J, Skotheim RI, Lothe RA, Paulo P, et al. Specific and redundant activities of ETV1 and ETV4 in prostate cancer aggressiveness revealed by co-overexpression cellular contexts. Oncotarget. 2015;6:5217–36.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Wei G-H, Badis G, Berger MF, Kivioja T, Palin K, Enge M, et al. Genome-wide analysis of ETS-family DNA-binding in vitro and in vivo. EMBO J. 2010;29:2147–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ramachandran B, Rajkumar T, Gopisetty G. Challenges in modeling EWS-FLI1-driven transgenic mouse model for Ewing sarcoma. Am J Transl Res. 2021;13:12181–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Gao Y, He X-Y, Wu XS, Huang Y-H, Toneyan S, Ha T, et al. ETV6 dependency in Ewing sarcoma by antagonism of EWS-FLI1-mediated enhancer activation. Nat Cell Biol. https://doi.org/10.1038/s41556-022-01060-1. (2023).

  37. Lu DY, Ellegast JM, Ross KN, Malone CF, Lin S, Mabe NW, et al. The ETS transcription factor ETV6 constrains the transcriptional activity of EWS–FLI to promote Ewing sarcoma. Nat Cell Biol. https://doi.org/10.1038/s41556-022-01059-8. (2023).

Download references

Author information

Authors and Affiliations

Authors

Contributions

The study was conceived and designed by PGH, KMO, and DGK. Animal data was collected and analyzed by PGH and KMO with help from MRB, LL, YM, DMC, and CML. RNA-sequencing analysis and neural network classification was performed by JON, PLB, SD, and AS. The paper with written by PGH, KMO, and DGK. This work was supported by grants from the Department of Defense (W81XWH-22-1-0454) to DGK, Alex’s Lemonade Stand Foundation to CML, the National Cancer Institute (7R35CA197616) to DGK and (1R38CA245204) PGH, and the American Society of Radiation Oncology (852948) to PGH.

Corresponding author

Correspondence to David G. Kirsch.

Ethics declarations

Competing interests

DGK is a cofounder of and stockholder in XRAD Therapeutics, which is developing radiosensitizers. DGK is a member of the scientific advisory board and owns stock in Lumicell Inc, a company commercializing intraoperative imaging technology. None of these affiliations represents a conflict of interest with respect to the work described in this manuscript. DGK is a coinventor on a patent for a handheld imaging device and is a coinventor on a patent for radiosensitizers. XRAD Therapeutics, Merck, Bristol Myers Squibb, and Varian Medical Systems have provided research support to DGK, but this did not support the research described in this manuscript. The other authors have no conflicting financial interests.

Additional information

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

Supplementary information

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

Hendrickson, P.G., Oristian, K.M., Browne, M.R. et al. Spontaneous expression of the CIC::DUX4 fusion oncoprotein from a conditional allele potently drives sarcoma formation in genetically engineered mice. Oncogene 43, 1223–1230 (2024). https://doi.org/10.1038/s41388-024-02984-8

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-024-02984-8

Search

Quick links