Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Dystrophin is a tumor suppressor in human cancers with myogenic programs

Abstract

Many common human mesenchymal tumors, including gastrointestinal stromal tumor (GIST), rhabdomyosarcoma (RMS) and leiomyosarcoma (LMS), feature myogenic differentiation1,2,3. Here we report that intragenic deletion of the dystrophin-encoding and muscular dystrophy–associated DMD gene is a frequent mechanism by which myogenic tumors progress to high-grade, lethal sarcomas. Dystrophin is expressed in the non-neoplastic and benign counterparts of GIST, RMS and LMS tumors, and DMD deletions inactivate larger dystrophin isoforms, including 427-kDa dystrophin, while preserving the expression of an essential 71-kDa isoform. Dystrophin inhibits myogenic sarcoma cell migration, invasion, anchorage independence and invadopodia formation, and dystrophin inactivation was found in 96%, 100% and 62% of metastatic GIST, embryonal RMS and LMS samples, respectively. These findings validate dystrophin as a tumor suppressor and likely anti-metastatic factor, suggesting that therapies in development for muscular dystrophies may also have relevance in the treatment of cancer.

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

Figure 1: Identification of somatic intragenic DMD deletions in human myogenic cancers.
Figure 2: MLPA evaluation of DMD exons 1–79 shows intragenic deletions in 24 myogenic cancers.
Figure 3: Loss of expression of 427-kDa dystrophin in most metastatic GIST, RMS and LMS tumors.
Figure 4: Expression of Dp71 dystrophin in myogenic tumors.
Figure 5: Restoration of dystrophin expression inhibits invasiveness and migration in DMD-inactivated GIST, RMS and LMS cells but not in a comparator non-myogenic fibrosarcoma cell line (HT-1080).
Figure 6: Restoration of dystrophin expression inhibits anchorage-independent growth and invadopodia formation in DMD-inactivated myogenic cancer cells.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Corless, C.L., Barnett, C.M. & Heinrich, M.C. Gastrointestinal stromal tumours: origin and molecular oncology. Nat. Rev. Cancer 11, 865–878 (2011).

    Article  CAS  Google Scholar 

  2. Arndt, C.A. & Crist, W.M. Common musculoskeletal tumors of childhood and adolescence. N. Engl. J. Med. 341, 342–352 (1999).

    Article  CAS  Google Scholar 

  3. Hernando, E. et al. The AKT-mTOR pathway plays a critical role in the development of leiomyosarcomas. Nat. Med. 13, 748–753 (2007).

    Article  CAS  Google Scholar 

  4. Corless, C.L., Fletcher, J.A. & Heinrich, M.C. Biology of gastrointestinal stromal tumors. J. Clin. Oncol. 22, 3813–3825 (2004).

    Article  CAS  Google Scholar 

  5. Taylor, B.S. et al. Advances in sarcoma genomics and new therapeutic targets. Nat. Rev. Cancer 11, 541–557 (2011).

    Article  CAS  Google Scholar 

  6. Hettmer, S. & Wagers, A.J. Muscling in: uncovering the origins of rhabdomyosarcoma. Nat. Med. 16, 171–173 (2010).

    Article  CAS  Google Scholar 

  7. Edris, B. et al. Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma. Proc. Natl. Acad. Sci. USA 109, 6656–6661 (2012).

    Article  CAS  Google Scholar 

  8. Würl, P. et al. Frequent occurrence of p53 mutations in rhabdomyosarcoma and leiomyosarcoma, but not in fibrosarcoma and malignant neural tumors. Int. J. Cancer 69, 317–323 (1996).

    Article  Google Scholar 

  9. Ognjanovic, S., Olivier, M., Bergemann, T.L. & Hainaut, P. Sarcomas in TP53 germline mutation carriers: a review of the IARC TP53 database. Cancer 118, 1387–1396 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Heinrich, M.C. et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 299, 708–710 (2003).

    Article  CAS  Google Scholar 

  12. Rossi, S. et al. Molecular and clinicopathologic characterization of gastrointestinal stromal tumors (GISTs) of small size. Am. J. Surg. Pathol. 34, 1480–1491 (2010).

    Article  Google Scholar 

  13. Schneider-Stock, R. et al. Loss of p16 protein defines high-risk patients with gastrointestinal stromal tumors: a tissue microarray study. Clin. Cancer Res. 11, 638–645 (2005).

    CAS  PubMed  Google Scholar 

  14. Henze, J. et al. p53 modulation as a therapeutic strategy in gastrointestinal stromal tumors. PLoS ONE 7, e37776 (2012).

    Article  CAS  Google Scholar 

  15. Hoffman, E.P., Brown, R.H. Jr. & Kunkel, L.M. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 51, 919–928 (1987).

    Article  CAS  Google Scholar 

  16. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  Google Scholar 

  17. Duszczyk, M.M., Wutz, A., Rybin, V. & Sattler, M. The Xist RNA A-repeat comprises a novel AUCG tetraloop fold and a platform for multimerization. RNA 17, 1973–1982 (2011).

    Article  CAS  Google Scholar 

  18. Muntoni, F., Torelli, S. & Ferlini, A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol. 2, 731–740 (2003).

    Article  CAS  Google Scholar 

  19. Tadayoni, R., Rendon, A., Soria-Jasso, L.E. & Cisneros, B. Dystrophin Dp71: the smallest but multifunctional product of the Duchenne muscular dystrophy gene. Mol. Neurobiol. 45, 43–60 (2012).

    Article  CAS  Google Scholar 

  20. Chapdelaine, P. et al. Functional EGFP-dystrophin fusion proteins for gene therapy vector development. Protein Eng. 13, 611–615 (2000).

    Article  CAS  Google Scholar 

  21. Tang, Y., Cummins, J., Huard, J. & Wang, B. AAV-directed muscular dystrophy gene therapy. Expert Opin. Biol. Ther. 10, 395–408 (2010).

    Article  CAS  Google Scholar 

  22. Ervasti, J.M. & Campbell, K.P. Membrane organization of the dystrophin-glycoprotein complex. Cell 66, 1121–1131 (1991).

    Article  CAS  Google Scholar 

  23. Berk, B.C., Fujiwara, K. & Lehoux, S. ECM remodeling in hypertensive heart disease. J. Clin. Invest. 117, 568–575 (2007).

    Article  CAS  Google Scholar 

  24. Valastyan, S. & Weinberg, R.A. Tumor metastasis: molecular insights and evolving paradigms. Cell 147, 275–292 (2011).

    Article  CAS  Google Scholar 

  25. Hanahan, D. & Weinberg, R.A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  Google Scholar 

  26. Murphy, D.A. & Courtneidge, S.A. The 'ins' and 'outs' of podosomes and invadopodia: characteristics, formation and function. Nat. Rev. Mol. Cell Biol. 12, 413–426 (2011).

    Article  CAS  Google Scholar 

  27. Fernandez, K., Serinagaoglu, Y., Hammond, S., Martin, L.T. & Martin, P.T. Mice lacking dystrophin or α sarcoglycan spontaneously develop embryonal rhabdomyosarcoma with cancer-associated p53 mutations and alternatively spliced or mutant Mdm2 transcripts. Am. J. Pathol. 176, 416–434 (2010).

    Article  CAS  Google Scholar 

  28. Chamberlain, J.S., Metzger, J., Reyes, M., Townsend, D. & Faulkner, J.A. Dystrophin-deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma. FASEB J. 21, 2195–2204 (2007).

    Article  CAS  Google Scholar 

  29. Schmidt, W.M. et al. DNA damage, somatic aneuploidy, and malignant sarcoma susceptibility in muscular dystrophies. PLoS Genet. 7, e1002042 (2011).

    Article  CAS  Google Scholar 

  30. Rossbach, H.C., Lacson, A., Grana, N.H. & Barbosa, J.L. Duchenne muscular dystrophy and concomitant metastatic alveolar rhabdomyosarcoma. J. Pediatr. Hematol. Oncol. 21, 528–530 (1999).

    Article  CAS  Google Scholar 

  31. Jakab, Z., Szegedi, I., Balogh, E., Kiss, C. & Oláh, E. Duchenne muscular dystrophy–rhabdomyosarcoma, ichthyosis vulgaris/acute monoblastic leukemia: association of rare genetic disorders and childhood malignant diseases. Med. Pediatr. Oncol. 39, 66–68 (2002).

    Article  Google Scholar 

  32. Barretina, J. et al. Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat. Genet. 42, 715–721 (2010).

    Article  CAS  Google Scholar 

  33. Gottipati, P. et al. Transcription-associated recombination is dependent on replication in mammalian cells. Mol. Cell. Biol. 28, 154–164 (2008).

    Article  CAS  Google Scholar 

  34. Körner, H. et al. Digital karyotyping reveals frequent inactivation of the dystrophin/DMD gene in malignant melanoma. Cell Cycle 6, 189–198 (2007).

    Article  Google Scholar 

  35. Li, Y. et al. UTRN on chromosome 6q24 is mutated in multiple tumors. Oncogene 26, 6220–6228 (2007).

    Article  CAS  Google Scholar 

  36. Tinsley, J.M. et al. Daily treatment with SMTC1100, a novel small molecule utrophin upregulator, dramatically reduces the dystrophic symptoms in the mdx mouse. PLoS ONE 6, e19189 (2011).

    Article  CAS  Google Scholar 

  37. Cirak, S. et al. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet 378, 595–605 (2011).

    Article  CAS  Google Scholar 

  38. Kawahara, G. et al. Drug screening in a zebrafish model of Duchenne muscular dystrophy. Proc. Natl. Acad. Sci. USA 108, 5331–5336 (2011).

    Article  CAS  Google Scholar 

  39. Gehrig, S.M. et al. Hsp72 preserves muscle function and slows progression of severe muscular dystrophy. Nature 484, 394–398 (2012).

    Article  CAS  Google Scholar 

  40. Seto, J.T., Ramos, J.N., Muir, L., Chamberlain, J.S. & Odom, G.L. Gene replacement therapies for Duchenne muscular dystrophy using adeno-associated viral vectors. Curr. Gene Ther. 12, 139–151 (2012).

    Article  CAS  Google Scholar 

  41. Bardsley, M.R. et al. Kitlow stem cells cause resistance to Kit/platelet-derived growth factor α inhibitors in murine gastrointestinal stromal tumors. Gastroenterology 139, 942–952 (2010).

    Article  CAS  Google Scholar 

  42. Izbeki, F. et al. Loss of Kitlow progenitors, reduced stem cell factor and high oxidative stress underlie gastric dysfunction in progeric mice. J. Physiol. (Lond.) 588, 3101–3117 (2010).

    Article  CAS  Google Scholar 

  43. 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).

    Article  Google Scholar 

  44. Rasheed, S., Nelson-Rees, W.A., Toth, E.M., Arnstein, P. & Gardner, M.B. Characterization of a newly derived human sarcoma cell line (HT-1080). Cancer 33, 1027–1033 (1974).

    Article  CAS  Google Scholar 

  45. Patial, S., Luo, J., Porter, K.J., Benovic, J.L. & Parameswaran, N. G-protein-coupled-receptor kinases mediate TNFα-induced NFκB signalling via direct interaction with and phosphorylation of IκBα. Biochem. J. 425, 169–178 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Tremblay (Quebec University Hospital) for the pCR3.1-miniDMD construct; T. Taguchi (Kochi Medical School) for the GIST-T1 cell line; Y. Hayashi, M. Bardsley and H. Qiu for their expert technical assistance; and S. Bauer (West German Cancer Center) for useful discussions and for funding support of J.H. This work was supported by grants from the US National Institutes of Health, including 1P50CA127003-07 (J.A.F. and G.D.D.), 1P50CA168512-01 (J.A.F., G.D.D. and A.M.-E.) and 5R01DK058185-12 (T.O.), and by the Virginia and Daniel K. Ludwig Trust for Cancer Research (J.A.F. and G.D.D.), the GIST Cancer Research Fund (J.A.F.), the Life Raft Group (J.A.F., T.O. and M.v.d.R.), the Cesarini Pan-Mass Challenge for GIST (J.A.F. and G.D.D.), Paul's Posse of the Pan-Mass Challenge (J.A.F. and G.D.D.), the Bernard F. and Alva B. Gimbel Foundation (L.M.K.), the Sarcoma Alliance for Research Through Collaboration (A.M.-E.) and the Erica Kaitz LMS Research NOW Fund (J.A.F., A.M.-E. and G.D.D.).

Author information

Authors and Affiliations

Authors

Contributions

J.A.F. supervised the project. Y.W. and J.A.F. generated the original hypothesis and designed the study. Y.W., A.M.-E., R.R.B., M.Z., Y.S., G.E., J.-C.L., J.H., B.S.F., Z.G., Y.S., X.G. and T.O. performed experiments. A.M.-E., C.R.A., C.D.M.F., C.P.R., M.v.d.R. and J.A.F. provided samples and clinical data. Y.W., A.M.-E., R.R.B., M.Z., Y.S., G.E., J.-C.L., B.S.F., Z.G., E.A.F., X.G., M.v.d.R., T.O., L.M.K. and J.A.F. analyzed data. C.R.A., C.D.M.F., G.D.D., M.v.d.R. and L.M.K. provided scientific advice and helpful comments on the project. Y.W., A.M.-E., G.E. and J.A.F. wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Jonathan A Fletcher.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Identical DMD deletions in a primary GIST and subsequent metastasis.

Identical DMD deletions in a primary gastric GIST and a subsequent metastasis, diagnosed 1 year later, from patient 31. SNP profiles of non-neoplastic DNA from the patient, the primary gastric GIST and the subsequent metastasis are shown. The top panel (a) shows the entire chromosome X; the bottom panel (b) focuses on the DMD locus. Data are shown as dChip SNP log2 ratio copy number.

Supplementary Figure 2 Identical DMD deletions in multiple GIST metastases from one patient.

Identical DMD deletions in each of 28 GIST metastases from patient 61. The metastases have a DMD deletion not present in non-neoplastic DNA from the patient, demonstrating the somatic nature of DMD deletion. The top panel (a) shows the entire chromosome X; the bottom panel (b) focuses on the DMD locus. Data are shown as dChip SNP log2 ratio copy number, with a representative SNP profile (metastasis 10) shown to the right.

Supplementary Figure 3 DMD intragenic deletions in 1,003 human cancers.

DMD intragenic deletions in 1,003 human cancers, including 40 myogenic sarcomas, 58 non-myogenic sarcomas and 905 non-sarcoma cancers. The 58 non-myogenic sarcomas (Supplementary Table 2) include 4 malignant peripheral nerve sheath tumors, 10 liposarcomas, 15 Ewing's sarcomas, 1 fibrosarcoma, 1 clear cell sarcoma, 1 spindle cell sarcoma, 1 dermatofibrosarcoma protuberans, 1 giant cell tumor, 1 mesenchymal chondrosarcoma and 23 osteosarcomas. The 905 non-sarcoma cancers (Cancer Cell Line Encyclopedia; Barretina et al. Nature, 2012) include 591 carcinomas, 175 hematologic neoplasms, 59 melanomas, 43 gliomas, 15 neuroblastomas and 22 other tumors. DMD intragenic deletions are significantly more frequent in myogenic sarcomas than in non-myogenic sarcomas (P < 0.0001, two-tailed Fisher's test) and non-sarcoma cancers (P < 0.0001, two-tailed Fisher's test).

Supplementary Figure 4 Detailed characterization of intragenic DMD deletions by MLPA.

DMD MLPA capillary electropherograms are normal in low-risk GISTs from male (a) and female (b) patients. MLPA shows intragenic hemizygous, homozygous and heterozygous deletions in metastatic GISTs from a male (c) and two female (d,e) patients, respectively. Each peak represents a single DMD exon, displayed non-consecutively on the basis of the size of the ligated stuffer sequence. The reference trace is shown in red, and the tumor sample is shown in blue.

Supplementary Figure 5 Expression of Dp71 dystrophin in non-myogenic sarcomas.

Protein blotting with dystrophin antibody 7A10 demonstrates Dp71 expression in malignant peripheral nerve sheath tumor (MPNST), liposarcoma (LPS), dermatofibrosarcoma protuberans (DFSP) and malignant solitary fibrous tumor (SFT).

Supplementary Figure 6 Dp71 dystrophin is a positive regulator of cell viability in myogenic sarcoma.

(a) Protein blot validation of siRNA-mediated inhibition of Dp71 in RMS176 (eRMS with DMD deletion) and RMS843 (aRMS without DMD deletion). (b) Dp71 inhibition reduces cell viability, as assessed by CellTiter-Glo assay. The control is a non-targeting siRNA. Data are shown as the mean ± s.d. from three replicates. Dp71 and Dp427 were detected with dystrophin antibodies 7A10 and DYS1, respectively.

Supplementary Figure 7 miniDMD expression in GIST, RMS and LMS cells at levels physiological for dystrophin in the corresponding cell lineages.

Stable transfection with miniDMD of GIST, RMS and LMS cells induces expression of 240-kDa miniDMD dystrophin at levels comparable to those of endogeneous 427-kDa dystrophin in low-risk GIST, skeletal muscle and myometrium cells, respectively. miniDMD was detected with DYS2 antibody.

Supplementary Figure 8 Effect of miniDMD expression on cell viability in DMD-inactivated GIST, RMS and LMS cells.

Stable transfection with miniDMD reduces cell viability in RMS176 and LMS04 cells, as assessed by CellTiter-Glo viability assay, but does not alter the viability of GIST (GIST-T1 and GIST430) or non-myogenic cells: fibrosarcoma (HT-1080), Ewing's sarcoma (EWS502) and HEK 293. Data are shown as the mean ± s.d. of three replicates.

Supplementary Figure 9 Dystrophin is not expressed in Ewing's sarcoma (EWS502 and EWS894), fibrosarcoma (HT-1080) and HEK 293 cells.

Dystrophin was detected with DYS1 antibody.

Supplementary Figure 10 Dystrophin immunohistochemistry in clinical samples.

Immunohistochemical detection in formalin-fixed, paraffin-embedded surgical samples shows robust dystrophin expression in skeletal, cardiac and smooth muscle (a) and in benign smooth muscle neoplasms (b, top), whereas dystrophin expression is inhibited in malignant smooth muscle tumors (b, bottom; LMS, leiomyosarcoma). (c) Contingency table summarizing dystrophin expression assessed by immunohistochemistry in 20 benign and 57 malignant smooth muscle tumors in a tissue microarray. Loss of dystrophin expression correlates with malignancy in smooth muscle tumors (P < 0.0001, two-tailed Fisher's test).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Tables 1–6 (PDF 7649 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Marino-Enriquez, A., Bennett, R. et al. Dystrophin is a tumor suppressor in human cancers with myogenic programs. Nat Genet 46, 601–606 (2014). https://doi.org/10.1038/ng.2974

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2974

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