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:

Inactivation of Capicua drives cancer metastasis

Abstract

Metastasis is the leading cause of death in people with lung cancer, yet the molecular effectors underlying tumor dissemination remain poorly defined. Through the development of an in vivo spontaneous lung cancer metastasis model, we show that the developmentally regulated transcriptional repressor Capicua (CIC) suppresses invasion and metastasis. Inactivation of CIC relieves repression of its effector ETV4, driving ETV4-mediated upregulation of MMP24, which is necessary and sufficient for metastasis. Loss of CIC, or an increase in levels of its effectors ETV4 and MMP24, is a biomarker of tumor progression and worse outcomes in people with lung and/or gastric cancer. Our findings reveal CIC as a conserved metastasis suppressor, highlighting new anti-metastatic strategies that could potentially improve patient outcomes.

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: Our in vivo orthotopic model identifies novel effectors of lung cancer metastasis.
Figure 2: CIC is altered in advanced-stage LA.
Figure 3: Inactivation of CIC de-represses an ETV4–MMP24 pro-metastatic circuit.
Figure 4: CIC effector MMP24 drives lung cancer metastasis.
Figure 5: MAPK pathway activation functionally suppresses CIC.
Figure 6: The CIC–ETV4–MMP24 metastatic axis is deregulated in gastric cancer.
Figure 7: CIC suppresses cancer metastasis.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Herbst, R.S., Heymach, J.V. & Lippman, S.M. Lung cancer. N. Engl. J. Med. 359, 1367–1380 (2008).

    Article  CAS  Google Scholar 

  2. Chaffer, C.L. & Weinberg, R.A. A perspective on cancer cell metastasis. Science 331, 1559–1564 (2011).

    Article  CAS  Google Scholar 

  3. Khanna, C. & Hunter, K. Modeling metastasis in vivo. Carcinogenesis 26, 513–523 (2005).

    Article  CAS  Google Scholar 

  4. Francia, G., Cruz-Munoz, W., Man, S., Xu, P. & Kerbel, R.S. Mouse models of advanced spontaneous metastasis for experimental therapeutics. Nat. Rev. Cancer 11, 135–141 (2011).

    Article  CAS  Google Scholar 

  5. Sequist, L.V. et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl. Med. 3, 75ra26 (2011).

    Article  Google Scholar 

  6. Walter, A.O. et al. Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC. Cancer Discov. 3, 1404–1415 (2013).

    Article  CAS  Google Scholar 

  7. Zhang, Z. et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 44, 852–860 (2012).

    Article  CAS  Google Scholar 

  8. Madero-Visbal, R.A. et al. Bioluminescence imaging correlates with tumor progression in an orthotopic mouse model of lung cancer. Surg. Oncol. 21, 23–29 (2012).

    Article  Google Scholar 

  9. Mordant, P. et al. Bioluminescent orthotopic mouse models of human localized non-small cell lung cancer: feasibility and identification of circulating tumour cells. PLoS One 6, e26073 (2011).

    Article  CAS  Google Scholar 

  10. Gleize, V. et al. CIC inactivating mutations identify aggressive subset of 1p19q codeleted gliomas. Ann. Neurol. 78, 355–374 (2015).

    Article  CAS  Google Scholar 

  11. Choi, N. et al. miR-93/miR-106b/miR-375-CIC-CRABP1: a novel regulatory axis in prostate cancer progression. Oncotarget 6, 23533–23547 (2015).

    PubMed  PubMed Central  Google Scholar 

  12. Jin, Y. et al. EGFR/Ras signaling controls Drosophila intestinal stem cell proliferation via Capicua-regulated genes. PLoS Genet. 11, e1005634 (2015).

    Article  Google Scholar 

  13. Bettegowda, C. et al. Mutations in CIC and FUBP1 contribute to human oligodendroglioma. Science 333, 1453–1455 (2011).

    Article  CAS  Google Scholar 

  14. Rizvi, N.A. et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).

    Article  CAS  Google Scholar 

  15. Lee, Y. et al. ATXN1 protein family and CIC regulate extracellular matrix remodeling and lung alveolarization. Dev. Cell 21, 746–757 (2011).

    Article  CAS  Google Scholar 

  16. Greenlee, K.J., Werb, Z. & Kheradmand, F. Matrix metalloproteinases in lung: multiple, multifarious, and multifaceted. Physiol. Rev. 87, 69–98 (2007).

    Article  CAS  Google Scholar 

  17. Jiménez, G., Shvartsman, S.Y. & Paroush, Z. The Capicua repressor—a general sensor of RTK signaling in development and disease. J. Cell Sci. 125, 1383–1391 (2012).

    Article  Google Scholar 

  18. Egeblad, M. & Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer 2, 161–174 (2002).

    Article  CAS  Google Scholar 

  19. Gyoő rffy, B., Surowiak, P., Budczies, J. & Lánczky, A. Online survival analysis software to assess the prognostic value of biomarkers using transcriptomic data in non-small-cell lung cancer. PLoS One 8, e82241 (2013).

    Article  Google Scholar 

  20. Hayashita-Kinoh, H. et al. Membrane-type 5 matrix metalloproteinase is expressed in differentiated neurons and regulates axonal growth. Cell Growth Differ. 12, 573–580 (2001).

    CAS  PubMed  Google Scholar 

  21. Elkin, M. & Vlodavsky, I. Tail vein assay of cancer metastasis. Curr. Protoc. Cell Biol. Chapter 19, Unit 19.2 (2001).

  22. Porlan, E. et al. MT5-MMP regulates adult neural stem cell functional quiescence through the cleavage of N-cadherin. Nat. Cell Biol. 16, 629–638 (2014).

    Article  CAS  Google Scholar 

  23. Blakely, C.M. et al. NF-κB-activating complex engaged in response to EGFR oncogene inhibition drives tumor cell survival and residual disease in lung cancer. Cell Rep. 11, 98–110 (2015).

    Article  CAS  Google Scholar 

  24. Schoppmann, S.F. et al. Downregulation of CIC does not associate with overexpression of ETV1 or MAP kinase pathway activation in gastrointestinal stromal tumors. Cancer Invest. 32, 363–367 (2014).

    Article  CAS  Google Scholar 

  25. Astigarraga, S. et al. A MAPK docking site is critical for downregulation of Capicua by Torso and EGFR RTK signaling. EMBO J. 26, 668–677 (2007).

    Article  CAS  Google Scholar 

  26. Futran, A.S., Kyin, S., Shvartsman, S.Y. & Link, A.J. Mapping the binding interface of ERK and transcriptional repressor Capicua using photocrosslinking. Proc. Natl. Acad. Sci. USA 112, 8590–8595 (2015).

    Article  CAS  Google Scholar 

  27. Grimm, O. et al. Torso RTK controls Capicua degradation by changing its subcellular localization. Development 139, 3962–3968 (2012).

    Article  CAS  Google Scholar 

  28. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014).

  29. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513, 202–209 (2014).

  30. Szász, A.M. et al. Cross-validation of survival associated biomarkers in gastric cancer using transcriptomic data of 1,065 patients. Oncotarget http://dx.doi.org/10.18632/oncotarget.10337 (2016).

  31. Weinstein, J.N. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 45, 1113–1120 (2013).

    Article  Google Scholar 

  32. Wei, W. et al. Single-cell phosphoproteomics resolves adaptive signaling dynamics and informs targeted combination therapy in glioblastoma. Cancer Cell 29, 563–573 (2016).

    Article  CAS  Google Scholar 

  33. Li, B. & Dewey, C.N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    Article  CAS  Google Scholar 

  34. Huang, W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  35. Backes, C. et al. GeneTrail—advanced gene set enrichment analysis. Nucleic Acids Res. 35, W186–W192 (2007).

    Article  Google Scholar 

  36. Talevich, E., Shain, A.H., Botton, T. & Bastian, B.C. CNVkit: copy number detection and visualization for targeted sequencing using off-target reads. bioRxiv Preprint available at http://dx.doi.org/10.1101/010876 (2014).

  37. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  Google Scholar 

  38. Boeva, V. et al. Control-FREEC: a tool for assessing copy number and allelic content using next-generation sequencing data. Bioinformatics 28, 423–425 (2012).

    Article  CAS  Google Scholar 

  39. Fransson, S. et al. Estimation of copy number aberrations: comparison of exome sequencing data with SNP microarrays identifies homozygous deletions of 19q13.2 and CIC in neuroblastoma. Int. J. Oncol. 48, 1103–1116 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

R.A.O. was supported by the A.P. Giannini Foundation and NIHT32CA177555-01. T.G.B. acknowledges support from NIH Director's New Innovator Award DP2 CA174497, NIH/NCI RO1 CA169338, and the Pew-Stewart Foundation Trusts. We thank B. Hann (UCSF Preclinical Therapeutics Core) for helpful discussions. The lentiviral GFP-Luc vector was a kind gift from M. Jensen (Seattle Children's Research Institute, Seattle, Washington, USA).

Author information

Authors and Affiliations

Authors

Contributions

R.A.O. designed and performed the experiments, analyzed the data, and wrote the manuscript. R.A.O., F.B., V.R.O., and B.G. performed in vivo studies. M.H. and W.W. performed TCGA analysis. K.G., J.S., V.A.M., and S. Ali provided lung cancer data sets. S. Asthana, J.F., H.J.H., and A.D.S. analyzed RNA-seq and CNA data. G.H., A.T., C.M.B., and M.S. provided clinical samples. T.G.B. directed the project, designed and analyzed experiments, and wrote the manuscript.

Corresponding author

Correspondence to Trever G Bivona.

Ethics declarations

Competing interests

H.J.H. and A.D.S. are employees of Clovis Oncology. K.G., J.S., V.A.M., and S. Ali are employees of Foundation Medicine. T.G.B. is a consultant to Novartis, Astellas, Array Biopharma, Ariad, Teva, and Astrazeneca, and has received research funding from Ignyta.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 and Supplementary Tables 1 and 5 (PDF 41965 kb)

Supplementary Table 2

Top 1,500 differentially expressed genes between H1975 M1 (CIC null) and H1975 M1 with reconstituted CIC expression (XLSX 137 kb)

Supplementary Table 3a

Top 1,000 differentially expressed genes between H1975 and H1975 M1 cells. Ranked by P value. Fold change and log2 fold change are also provided. (XLSX 117 kb)

Supplementary Table 3b

Top 1,000 differentially expressed genes between H1975 and H1975 M2 cells. Ranked by P value. Fold change and log2 fold change are also provided. (XLSX 121 kb)

Supplementary Table 4

267 putative CIC response genes. (XLSX 82 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Okimoto, R., Breitenbuecher, F., Olivas, V. et al. Inactivation of Capicua drives cancer metastasis. Nat Genet 49, 87–96 (2017). https://doi.org/10.1038/ng.3728

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3728

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