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.

  • Original Article
  • Published:

The induction of MIG6 under hypoxic conditions is critical for dormancy in primary cultured lung cancer cells with activating EGFR mutations

Abstract

The biologic activity of individual cancer cells is highly heterogeneous. Hypoxia, one of the prominent features of a tumor microenvironment, is thought to be causal in generating this cellular heterogeneity. In this study, we revealed that primary lung cancer cells harboring activating epidermal growth factor receptor (EGFR) mutations generally entered a dormant state when hypoxic. We found that heterodimer formation of the ERBB family receptor tyrosine kinases (RTKs), and their subsequent downstream signaling, was diminished under hypoxic conditions, although phosphorylation of the EGFR was retained. Dormant lung cancer cells were found to be resistant to EGFR tyrosine kinase inhibitor (TKI) treatment. In terms of mechanism, we found that a negative regulator of ERBB signaling, MIG6/ERRFI1/RALT/Gene33, was induced by hypoxia both in vitro and in vivo. MIG6 expression prevented heterodimer formation of ERBB family RTKs, and suppressed their downstream signaling. Knockdown of MIG6 enhanced tumor cell growth under hypoxic conditions, and promoted the phosphorylation of ERK and AKT via increased EGFR-HER3 binding. Critically, sensitivity to an EGFR-TKI, as well as to irradiation under hypoxic conditions, was increased in MIG6 knockdown cells. The expression of MIG6 was partly correlated with a pS6 negative zone in patient tumors. Analyses of tumor sections from 68 patients with activating EGFR mutations showed that patients with high MIG6 expression showed significantly shorter survival after EGFR-TKI treatment than other groups. Collectively, our data suggest that dormant cancer cells with a high MIG6 expression level might be one of the causes of EGFR-TKI resistance in EGFR mutant lung cancer cells.

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
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

References

  1. Marusyk A, Almendro V, Polyak K . Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 2012; 12: 323–334.

    Article  CAS  Google Scholar 

  2. Gilkes DM, Semenza GL, Wirtz D . Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat Rev Cancer 2014; 14: 430–439.

    Article  CAS  Google Scholar 

  3. Harada H, Inoue M, Itasaka S, Hirota K, Morinibu A, Shinomiya K et al. Cancer cells that survive radiation therapy acquire HIF-1 activity and translocate towards tumour blood vessels. Nat Commun 2012; 3: 783.

    Article  Google Scholar 

  4. Semenza GL . The hypoxic tumor microenvironment: a driving force for breast cancer progression. Biochim Biophys Acta 2016; 1863: 382–391.

    Article  CAS  Google Scholar 

  5. Sosa MS, Bragado P, Aguirre-Ghiso JA . Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat Rev Cancer 2014; 14: 611–622.

    Article  CAS  Google Scholar 

  6. Endo H, Okuyama H, Ohue M, Inoue M . Dormancy of cancer cells with suppression of AKT activity contributes to survival in chronic hypoxia. PLoS One 2014; 9: e98858.

    Article  Google Scholar 

  7. Lowe SW, Cepero E, Evan G . Intrinsic tumour suppression. Nature 2004; 432: 307–315.

    Article  CAS  Google Scholar 

  8. Hanahan D, Weinberg RA . Hallmarks of cancer: the next generation. Cell 2011; 144: 646–674.

    Article  CAS  Google Scholar 

  9. Arsham AM, Howell JJ, Simon MC . A novel hypoxia-inducible factor-independent hypoxic response regulating mammalian target of rapamycin and its targets. J Biol Chem 2003; 278: 29655–29660.

    Article  CAS  Google Scholar 

  10. Endo H, Murata K, Mukai M, Ishikawa O, Inoue M . Activation of insulin-like growth factor signaling induces apoptotic cell death under prolonged hypoxia by enhancing endoplasmic reticulum stress response. Cancer Res 2007; 67: 8095–8103.

    Article  CAS  Google Scholar 

  11. Okuyama H, Endo H, Akashika T, Kato K, Inoue M . Downregulation of c-MYC protein levels contributes to cancer cell survival under dual deficiency of oxygen and glucose. Cancer Res 2010; 70: 10213–10223.

    Article  CAS  Google Scholar 

  12. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004; 350: 2129–2139.

    Article  CAS  Google Scholar 

  13. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004; 304: 1497–1500.

    Article  CAS  Google Scholar 

  14. Kondo J, Endo H, Okuyama H, Ishikawa O, Iishi H, Tsujii M et al. Retaining cell-cell contact enables preparation and culture of spheroids composed of pure primary cancer cells from colorectal cancer. Proc Natl Acad Sci USA 2011; 108: 6235–6240.

    Article  CAS  Google Scholar 

  15. Endo H, Okami J, Okuyama H, Kumagai T, Uchida J, Kondo J et al. Spheroid culture of primary lung cancer cells with neuregulin 1/HER3 pathway activation. J Thorac Oncol 2013; 8: 131–139.

    Article  CAS  Google Scholar 

  16. Ruvinsky I, Meyuhas O . Ribosomal protein S6 phosphorylation: from protein synthesis to cell size. Trends Biochem Sci 2006; 31: 342–348.

    Article  CAS  Google Scholar 

  17. Pinkas-Kramarski R, Shelly M, Glathe S, Ratzkin BJ, Yarden Y . Neu differentiation factor/neuregulin isoforms activate distinct receptor combinations. J Biol Chem 1996; 271: 19029–19032.

    Article  CAS  Google Scholar 

  18. Zhang X, Pickin KA, Bose R, Jura N, Cole PA, Kuriyan J . Inhibition of the EGF receptor by binding of MIG6 to an activating kinase domain interface. Nature 2007; 450: 741–744.

    Article  CAS  Google Scholar 

  19. Swinson DEB . Carbonic anhydrase IX expression, a novel surrogate marker of tumor hypoxia, is associated with a poor prognosis in non-small-cell lung cancer. J Clin Oncol 2003; 21: 473–482.

    Article  CAS  Google Scholar 

  20. Silvera D, Formenti SC, Schneider RJ . Translational control in cancer. Nat Rev Cancer 2010; 10: 254–266.

    Article  CAS  Google Scholar 

  21. Braunstein S, Karpisheva K, Pola C, Goldberg J, Hochman T, Yee H et al. A hypoxia-controlled cap-dependent to cap-independent translation switch in breast cancer. Mol Cell 2007; 28: 501–512.

    Article  CAS  Google Scholar 

  22. Uniacke J, Holterman CE, Lachance G, Franovic A, Jacob MD, Fabian MR et al. An oxygen-regulated switch in the protein synthesis machinery. Nature 2012; 486: 126–129.

    Article  CAS  Google Scholar 

  23. Young RM, Wang SJ, Gordan JD, Ji X, Liebhaber SA, Simon MC . Hypoxia-mediated selective mRNA translation by an internal ribosome entry site-independent mechanism. J Biol Chem 2008; 283: 16309–16319.

    Article  CAS  Google Scholar 

  24. Izumchenko E, Chang X, Michailidi C, Kagohara L, Ravi R, Paz K et al. The TGFbeta-miR200-MIG6 pathway orchestrates the EMT-associated kinase switch that induces resistance to EGFR inhibitors. Cancer Res 2014; 74: 3995–4005.

    Article  CAS  Google Scholar 

  25. Zhang YW, Staal B, Su Y, Swiatek P, Zhao P, Cao B et al. Evidence that MIG-6 is a tumor-suppressor gene. Oncogene 2007; 26: 269–276.

    Article  CAS  Google Scholar 

  26. Lafont JE, Talma S, Hopfgarten C, Murphy CL . Hypoxia promotes the differentiated human articular chondrocyte phenotype through SOX9-dependent and -independent pathways. J Biol Chem 2008; 283: 4778–4786.

    Article  CAS  Google Scholar 

  27. Barker HE, Paget JT, Khan AA, Harrington KJ . The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer 2015; 15: 409–425.

    Article  CAS  Google Scholar 

  28. Crea F, Nur Saidy NR, Collins CC, Wang Y . The epigenetic/noncoding origin of tumor dormancy. Trends Mol Med 2015; 21: 206–211.

    Article  CAS  Google Scholar 

  29. Amin R, Morita-Fujimura Y, Tawarayama H, Semba K, Chiba N, Fukumoto M et al. DeltaNp63alpha induces quiescence and downregulates the BRCA1 pathway in estrogen receptor-positive luminal breast cancer cell line MCF7 but not in other breast cancer cell lines. Mol Oncol 2016; 10: 575–593.

    Article  CAS  Google Scholar 

  30. Peart T, Ramos Valdes Y, Correa RJ, Fazio E, Bertrand M, McGee J et al. Intact LKB1 activity is required for survival of dormant ovarian cancer spheroids. Oncotarget 2015; 6: 22424–22438.

    Article  Google Scholar 

  31. Wang N, Docherty F, Brown HK, Reeves K, Fowles A, Lawson M et al. Mitotic quiescence, but not unique 'stemness,' marks the phenotype of bone metastasis-initiating cells in prostate cancer. FASEB J 2015; 29: 3141–3150.

    Article  CAS  Google Scholar 

  32. Bui AT, Laurent F, Havard M, Dautry F, Tchenio T . SMAD signaling and redox imbalance cooperate to induce prostate cancer cell dormancy. Cell Cycle 2015; 14: 1218–1231.

    Article  CAS  Google Scholar 

  33. Tiram G, Segal E, Krivitsky A, Shreberk-Hassidim R, Ferber S, Ofek P et al. Identification of dormancy-associated microRNAs for the design of osteosarcoma-targeted dendritic polyglycerol nanopolyplexes. ACS Nano 2016; 10: 2028–2045.

    Article  CAS  Google Scholar 

  34. Sosa MS, Parikh F, Maia AG, Estrada Y, Bosch A, Bragado P et al. NR2F1 controls tumour cell dormancy via SOX9- and RARbeta-driven quiescence programmes. Nat Commun 2015; 6: 6170.

    Article  CAS  Google Scholar 

  35. Ameri K, Jahangiri A, Rajah AM, Tormos KV, Nagarajan R, Pekmezci M et al. HIGD1A regulates oxygen consumption, ROS production, and AMPK activity during glucose deprivation to modulate cell survival and tumor growth. Cell Rep 2015, pii S2211–1247.

  36. Aguirre-Ghiso JA, Ossowski L, Rosenbaum SK . Green fluorescent protein tagging of extracellular signal-regulated kinase and p38 pathways reveals novel dynamics of pathway activation during primary and metastatic growth. Cancer Res 2004; 64: 7336–7345.

    Article  CAS  Google Scholar 

  37. Abravanel DL, Belka GK, Pan TC, Pant DK, Collins MA, Sterner CJ et al. Notch promotes recurrence of dormant tumor cells following HER2/neu-targeted therapy. J Clin Invest 2015; 125: 2484–2496.

    Article  Google Scholar 

  38. Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2005; 2: e73.

    Article  Google Scholar 

  39. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007; 316: 1039–1043.

    Article  CAS  Google Scholar 

  40. Yano S, Wang W, Li Q, Matsumoto K, Sakurama H, Nakamura T et al. Hepatocyte growth factor induces gefitinib resistance of lung adenocarcinoma with epidermal growth factor receptor-activating mutations. Cancer Res 2008; 68: 9479–9487.

    Article  CAS  Google Scholar 

  41. Ohashi K, Sequist LV, Arcila ME, Moran T, Chmielecki J, Lin YL et al. Lung cancers with acquired resistance to EGFR inhibitors occasionally harbor BRAF gene mutations but lack mutations in KRAS, NRAS, or MEK1. Proc Natl Acad Sci USA 2012; 109: E2127–E2133.

    Article  CAS  Google Scholar 

  42. Takezawa K, Pirazzoli V, Arcila ME, Nebhan CA, Song X, de Stanchina E et al. HER2 amplification: a potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov 2012; 2: 922–933.

    Article  CAS  Google Scholar 

  43. Wang W, Li Q, Yamada T, Matsumoto K, Matsumoto I, Oda M et al. Crosstalk to stromal fibroblasts induces resistance of lung cancer to epidermal growth factor receptor tyrosine kinase inhibitors. Clin Cancer Res 2009; 15: 6630–6638.

    Article  CAS  Google Scholar 

  44. Minakata K, Takahashi F, Nara T, Hashimoto M, Tajima K, Murakami A et al. Hypoxia induces gefitinib resistance in non-small-cell lung cancer with both mutant and wild-type epidermal growth factor receptors. Cancer Sci 2012; 103: 1946–1954.

    Article  CAS  Google Scholar 

  45. Makkinje A, Quinn DA, Chen A, Cadilla CL, Force T, Bonventre JV et al. Gene 33/Mig-6, a transcriptionally inducible adapter protein that binds GTP-Cdc42 and activates SAPK/JNK. A potential marker transcript for chronic pathologic conditions, such as diabetic nephropathy. Possible role in the response to persistent stress. J Biol Chem 2000; 275: 17838–17847.

    Article  CAS  Google Scholar 

  46. Xu D, Patten RD, Force T, Kyriakis JM . Gene 33/RALT is induced by hypoxia in cardiomyocytes, where it promotes cell death by suppressing phosphatidylinositol 3-kinase and extracellular signal-regulated kinase survival signaling. Mol Cell Biol 2006; 26: 5043–5054.

    Article  CAS  Google Scholar 

  47. Anastasi S, Lamberti D, Alema S, Segatto O . Regulation of the ErbB network by the MIG6 feedback loop in physiology, tumor suppression and responses to oncogene-targeted therapeutics. Semin Cell Dev Biol 2016; 50: 115–124.

    Article  CAS  Google Scholar 

  48. Carroll VA, Ashcroft M . Role of hypoxia-inducible factor (HIF)-1alpha versus HIF-2alpha in the regulation of HIF target genes in response to hypoxia, insulin-like growth factor-I, or loss of von Hippel-Lindau function: implications for targeting the HIF pathway. Cancer Res 2006; 66: 6264–6270.

    Article  CAS  Google Scholar 

  49. Ferby I, Reschke M, Kudlacek O, Knyazev P, Pante G, Amann K et al. Mig6 is a negative regulator of EGF receptor-mediated skin morphogenesis and tumor formation. Nat Med 2006; 12: 568–573.

    Article  CAS  Google Scholar 

  50. Maity TK, Venugopalan A, Linnoila I, Cultraro CM, Giannakou A, Nemati R et al. Loss of MIG6 accelerates initiation and progression of mutant epidermal growth factor receptor-driven lung adenocarcinoma. Cancer Discov 2015; 5: 534–549.

    Article  CAS  Google Scholar 

  51. Chang X, Izumchenko E, Solis LM, Kim MS, Chatterjee A, Ling S et al. The relative expression of Mig6 and EGFR is associated with resistance to EGFR kinase inhibitors. PLoS One 2013; 8: e68966.

    Article  CAS  Google Scholar 

  52. Sato Y, Tateno H, Adachi J, Okuyama H, Endo H, Tomonaga T et al. Generation of a monoclonal antibody recognizing the CEACAM glycan structure and inhibiting adhesion using cancer tissue-originated spheroid as an antigen. Sci Rep 2016; 6: 24823.

    Article  CAS  Google Scholar 

  53. Nakajima A, Endo H, Okuyama H, Kiyohara Y, Kimura T, Kamiura S et al. Radiation sensitivity assay with a panel of patient-derived spheroids of small cell carcinoma of the cervix. Int J Cancer 2015; 136: 2949–2960.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Dr T Tabuchi and Dr K Fukui for statistical counseling, N Kanto and T Yasuda for technical assistance, and M Izutsu for secretarial assistance. This work was supported by JSPS Grant-in-Aid for Scientific Research on Innovative Areas (26111005) (to MI) and KAKENHI (25461937) (to HE).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to M Inoue.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Endo, H., Okami, J., Okuyama, H. et al. The induction of MIG6 under hypoxic conditions is critical for dormancy in primary cultured lung cancer cells with activating EGFR mutations. Oncogene 36, 2824–2834 (2017). https://doi.org/10.1038/onc.2016.431

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2016.431

This article is cited by

Search

Quick links