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:

Positively selected enhancer elements endow osteosarcoma cells with metastatic competence

A Corrigendum to this article was published on 01 April 2018

This article has been updated

Abstract

Metastasis results from a complex set of traits acquired by tumor cells, distinct from those necessary for tumorigenesis. Here, we investigate the contribution of enhancer elements to the metastatic phenotype of osteosarcoma. Through epigenomic profiling, we identify substantial differences in enhancer activity between primary and metastatic human tumors and between near isogenic pairs of highly lung metastatic and nonmetastatic osteosarcoma cell lines. We term these regions metastatic variant enhancer loci (Met-VELs). Met-VELs drive coordinated waves of gene expression during metastatic colonization of the lung. Met-VELs cluster nonrandomly in the genome, indicating that activity of these enhancers and expression of their associated gene targets are positively selected. As evidence of this causal association, osteosarcoma lung metastasis is inhibited by global interruptions of Met-VEL-associated gene expression via pharmacologic BET inhibition, by knockdown of AP-1 transcription factors that occupy Met-VELs, and by knockdown or functional inhibition of individual genes activated by Met-VELs, such as that encoding coagulation factor III/tissue factor (F3). We further show that genetic deletion of a single Met-VEL at the F3 locus blocks metastatic cell outgrowth in the lung. These findings indicate that Met-VELs and the genes they regulate play a functional role in metastasis and may be suitable targets for antimetastatic therapies.

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 Met-VELs and Met-VEL clusters through H3K4me1 ChIP–seq.
Figure 2: Met-VELs modulate gene expression during metastatic colonization of the lung.
Figure 3: Results from an in vivo high-throughput RNAi functional assay of candidate metastasis dependency genes.
Figure 4: F3 mediates lung metastasis of osteosarcoma.
Figure 5: Deletion of a single gained Met-VEL blunts F3 expression and mitigates lung metastasis of osteosarcoma cells.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Change history

  • 07 February 2018

    In the version of this article initially published, two of the authors are incorrectly identified as John Stamatoyannopolus and Henri Versteeg. The authors' names are John A Stamatoyannopoulos and Henri H Versteeg. Also, the affiliation "Research Laboratory, Istituto Ortopedico Rizzoli, Bologna, Italy" is incorrect. The correct affiliation is "Laboratory of Experimental Oncology, Istituto Ortopedico Rizzoli, Bologna, Italy". The errors have been corrected in the HTML and PDF versions of the article.

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Chambers, A.F., Groom, A.C. & MacDonald, I.C. Dissemination and growth of cancer cells in metastatic sites. Nat. Rev. Cancer 2, 563–572 (2002).

    Article  CAS  PubMed  Google Scholar 

  3. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hong, M.K. et al. Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer. Nat. Commun. 6, 6605 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Bos, P.D. et al. Genes that mediate breast cancer metastasis to the brain. Nature 459, 1005–1009 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kang, Y. et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3, 537–549 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Minn, A.J. et al. Genes that mediate breast cancer metastasis to lung. Nature 436, 518–524 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Factor, D.C. et al. Epigenomic comparison reveals activation of “seed” enhancers during transition from naive to primed pluripotency. Cell Stem Cell 14, 854–863 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gifford, C.A. et al. Transcriptional and epigenetic dynamics during specification of human embryonic stem cells. Cell 153, 1149–1163 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhu, J. et al. Genome-wide chromatin state transitions associated with developmental and environmental cues. Cell 152, 642–654 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Heintzman, N.D. et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Akhtar-Zaidi, B. et al. Epigenomic enhancer profiling defines a signature of colon cancer. Science 336, 736–739 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    CAS  PubMed  Google Scholar 

  15. Lovén, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Ramaswamy, S., Ross, K.N., Lander, E.S. & Golub, T.R. A molecular signature of metastasis in primary solid tumors. Nat. Genet. 33, 49–54 (2003).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kansara, M., Teng, M.W., Smyth, M.J. & Thomas, D.M. Translational biology of osteosarcoma. Nat. Rev. Cancer 14, 722–735 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Huang, Y.M., Hou, C.H., Hou, S.M. & Yang, R.S. The metastasectomy and timing of pulmonary metastases on the outcome of osteosarcoma patients. Clin. Med. Oncol. 3, 99–105 (2009).

    PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ren, L. et al. Characterization of the metastatic phenotype of a panel of established osteosarcoma cells. Oncotarget 6, 29469–29481 (2015).

    PubMed  PubMed Central  Google Scholar 

  22. Zentner, G.E., Tesar, P.J. & Scacheri, P.C. Epigenetic signatures distinguish multiple classes of enhancers with distinct cellular functions. Genome Res. 21, 1273–1283 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. Huang, H., Bhat, A., Woodnutt, G. & Lappe, R. Targeting the ANGPT–TIE2 pathway in malignancy. Nat. Rev. Cancer 10, 575–585 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Clayton, P.E., Banerjee, I., Murray, P.G. & Renehan, A.G. Growth hormone, the insulin-like growth factor axis, insulin and cancer risk. Nat. Rev. Endocrinol. 7, 11–24 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Pinski, J. et al. Inhibition of growth of human osteosarcomas by antagonists of growth hormone-releasing hormone. J. Natl. Cancer Inst. 87, 1787–1794 (1995).

    Article  CAS  PubMed  Google Scholar 

  27. Li, N. et al. Phosphodiesterase 10A: a novel target for selective inhibition of colon tumor cell growth and β-catenin-dependent TCF transcriptional activity. Oncogene 34, 1499–1509 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. van den Berg, Y.W., Osanto, S., Reitsma, P.H. & Versteeg, H.H. The relationship between tissue factor and cancer progression: insights from bench and bedside. Blood 119, 924–932 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Mendoza, A. et al. Modeling metastasis biology and therapy in real time in the mouse lung. J. Clin. Invest. 120, 2979–2988 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Corradin, O. et al. Combinatorial effects of multiple enhancer variants in linkage disequilibrium dictate levels of gene expression to confer susceptibility to common traits. Genome Res. 24, 1–13 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Leaner, V.D. et al. Inhibition of AP-1 transcriptional activity blocks the migration, invasion, and experimental metastasis of murine osteosarcoma. Am. J. Pathol. 174, 265–275 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lamoureux, F. et al. Selective inhibition of BET bromodomain epigenetic signalling interferes with the bone-associated tumour vicious cycle. Nat. Commun. 5, 3511 (2014).

    Article  PubMed  CAS  Google Scholar 

  34. Puissant, A. et al. Targeting MYCN in neuroblastoma by BET bromodomain inhibition. Cancer Discov. 3, 308–323 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Bandopadhayay, P. et al. BET bromodomain inhibition of MYC-amplified medulloblastoma. Clin. Cancer Res. 20, 912–925 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Fellmann, C. et al. An optimized microRNA backbone for effective single-copy RNAi. Cell Rep. 5, 1704–1713 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Versteeg, H.H. et al. Inhibition of tissue factor signaling suppresses tumor growth. Blood 111, 190–199 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. You, J.S. & Jones, P.A. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell 22, 9–20 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Jones, S. et al. Comparative lesion sequencing provides insights into tumor evolution. Proc. Natl. Acad. Sci. USA 105, 4283–4288 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liu, W. et al. Copy number analysis indicates monoclonal origin of lethal metastatic prostate cancer. Nat. Med. 15, 559–565 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Campbell, P.J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yachida, S. et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467, 1114–1117 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Navin, N. et al. Tumour evolution inferred by single-cell sequencing. Nature 472, 90–94 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Moelans, C.B. et al. Genomic evolution from primary breast carcinoma to distant metastasis: Few copy number changes of breast cancer related genes. Cancer Lett. 344, 138–146 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Kerbel, R.S., Frost, P., Liteplo, R., Carlow, D.A. & Elliott, B.E. Possible epigenetic mechanisms of tumor progression: induction of high-frequency heritable but phenotypically unstable changes in the tumorigenic and metastatic properties of tumor cell populations by 5-azacytidine treatment. J. Cell. Physiol. Suppl. 3, 87–97 (1984).

    Article  CAS  PubMed  Google Scholar 

  46. Rodenhiser, D.I. Epigenetic contributions to cancer metastasis. Clin. Exp. Metastasis 26, 5–18 (2009).

    Article  CAS  PubMed  Google Scholar 

  47. Javaid, S. et al. Dynamic chromatin modification sustains epithelial-mesenchymal transition following inducible expression of Snail-1. Cell Rep. 5, 1679–1689 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Latil, M. et al. Cell-type-specific chromatin states differentially prime squamous cell carcinoma tumor-initiating cells for epithelial to mesenchymal transition. Cell Stem Cell 20, 191–204.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Denny, S.K. et al. Nfib promotes metastasis through a widespread increase in chromatin accessibility. Cell 166, 328–342 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Roe, J.S. et al. Enhancer reprogramming promotes pancreatic cancer metastasis. Cell 170, 875–888.e20 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Khanna, C. et al. An orthotopic model of murine osteosarcoma with clonally related variants differing in pulmonary metastatic potential. Clin. Exp. Metastasis 18, 261–271 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Schmidt, D. et al. ChIP–seq: using high-throughput sequencing to discover protein-DNA interactions. Methods 48, 240–248 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. McLean, C.Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Reimand, J., Arak, T. & Vilo, J. g:Profiler—a web server for functional interpretation of gene lists (2011 update). Nucleic Acids Res. 39, W307–15 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G.D. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS One 5, e13984 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Song, L. & Crawford, G.E. DNase-seq: a high-resolution technique for mapping active gene regulatory elements across the genome from mammalian cells. Cold Spring Harb. Protoc. https://doi.org/10.1101/pdb.prot5384 (2010).

    Article  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  58. Liu, T. et al. Cistrome: an integrative platform for transcriptional regulation studies. Genome Biol. 12, R83 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Knott, S.R.V. et al. A computational algorithm to predict shRNA potency. Mol. Cell 56, 796–807 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zuber, J. et al. Toolkit for evaluating genes required for proliferation and survival using tetracycline-regulated RNAi. Nat. Biotechnol. 29, 79–83 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Goecks, J., Nekrutenko, A. & Taylor, J. Galaxy: a comprehensive approach for supporting accessible, reproducible, and transparent computational research in the life sciences. Genome Biol. 11, R86 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Osborne, T.S. et al. Evaluation of eIF4E expression in an osteosarcoma-specific tissue microarray. J. Pediatr. Hematol. Oncol. 33, 524–528 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Sakuma, T. et al. Efficient TALEN construction and evaluation methods for human cell and animal applications. Genes Cells 18, 315–326 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Cermak, T. et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 39, e82 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank the members of the Tesar, Kaplan, and Helman laboratories for their input throughout the course of the project as well as B. Decker for his input on the manuscript text. Additional support was provided by the Genomics Core Facility of the Case Western Reserve University School of Medicine's Genetics and Genome Sciences Department and the Case Comprehensive Cancer Center (P30CA043703). This work was supported by the Liddy Shriver Sarcoma Initiative (P.C.S., C.K., J.J.M.), the QuadW Foundation (P.C.S.), Sarcoma Foundation of America (P.C.S.), St. Baldrick's Foundation (A.Y.H.), Alex's Lemonade Stand Foundation (A.Y.H.), Hyundai Hope-on-Wheels Program (A.Y.H.), Pediatric Cancer Research Foundation (A.Y.H.), CCCC AYA Oncology Pilot Grant (A.Y.H.), National Institutes of Health (NIH) grants F30 CA186633 (J.J.M.), F30 CA183510 (T.E.M.), T32 GM007250 (J.J.M., T.E.M., S.H.), R01CA193677 (P.C.S.), R01CA204279 (P.C.S.), R01CA160356 (P.C.S.), F31CA192874 (F.A.), R21CA218790 (A.Y.H.), NIH Intramural Visiting Fellow Program 15335 (M.M.L.), and NIH Intramural Research Program (C.K.).

Author information

Authors and Affiliations

Authors

Contributions

J.J.M., C.K., and P.C.S. conceived the overall experimental design. J.J.M., C.F.B., and G.D. generated ChIP–seq, RNA-seq, and DHS-seq data. J.J.M., A.S., S.H., and P.C.S. completed analyses of ChIP–seq, RNA-seq, and DHS-seq data. J.J.M. and T.E.M. designed and completed the shRNA screening experiment and subsequent analysis. T.E.M. completed functional enrichment analysis of RNA-seq data. J.J.M. and I.B. generated 4C-seq data. J.J.M. and A.S. analyzed 4C-seq data. J.J.M., A.M., and I.B. and M.M.L. completed the in vivo and ex vivo metastasis experiments. J.J.M., J.T.M., and F.A. designed and completed the orthotopic metastasis experiments. J.J.M., D.R.C., and A.P.W.F. designed and completed the TALEN deletion experiments. M.Y.K. completed the in vitro F3 experiments. M.G., A.R., and P.P. provided subject tumor samples and clinical data. B.P.R. assessed F3 staining in subject tissue microarrays. A.D., A.Y.H., P.S.M., L.J.H., H.H.V., J.A.S., C.K., and P.C.S. provided the technical expertise and facilities to complete the experiments. J.J.M. and P.C.S. analyzed all data and wrote the paper. All authors provided intellectual input, edited, and approved the final manuscript.

Corresponding author

Correspondence to Peter C Scacheri.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures & Tables

Supplementary Figures 1–13 and Supplementary Table 1 (PDF 2043 kb)

Life Sciences Reporting Summary (PDF 162 kb)

Supplementary Table 2

Met-VEL gene overlaps across patient tumors and cell lines (XLSX 468 kb)

Supplementary Table 3

Hairpins used in high-throughput in vivo RNAi screen (XLSX 37 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morrow, J., Bayles, I., Funnell, A. et al. Positively selected enhancer elements endow osteosarcoma cells with metastatic competence. Nat Med 24, 176–185 (2018). https://doi.org/10.1038/nm.4475

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4475

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