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:

GSK-3 modulates cellular responses to a broad spectrum of kinase inhibitors

Abstract

A fundamental challenge in treating disease is identifying molecular states that affect cellular responses to drugs. Here, we focus on glycogen synthase kinase 3 (GSK-3), a key regulator for many of the hallmark behaviors of cancer cells. We alter GSK-3 activity in colon epithelial cells to test its role in modulating drug response. We find that GSK-3 activity broadly affects the cellular sensitivities to a panel of oncology drugs and kinase inhibitors. Specifically, inhibition of GSK-3 activity can strongly desensitize or sensitize cells to kinase inhibitors (for example, mTOR or PLK1 inhibitors, respectively). Additionally, colorectal cancer cell lines, in which GSK-3 function is commonly suppressed, are resistant to mTOR inhibitors and yet highly sensitive to PLK1 inhibitors, and this is further exacerbated by additional GSK-3 inhibition. Finally, by conducting a kinome-wide RNAi screen, we find that GSK-3 modulates the cell proliferative phenotype of a large fraction (35%) of the kinome, which includes 50% of current, clinically relevant kinase-targeted drugs. Our results highlight an underappreciated interplay of GSK-3 with therapeutically important kinases and suggest strategies for identifying disease-specific molecular profiles that can guide optimal selection of drug treatment.

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: Decreased GSK-3 activity alters cellular response to oncology drugs and kinase inhibitors.
Figure 2: GSK-3 inhibition desensitizes cells to mTOR inhibitors.
Figure 3: GSK-3 inhibition sensitizes cells to PLK1 inhibitors.
Figure 4: CRC cell lines are intrinsically more resistant to mTOR inhibitors and sensitive to PLK1 inhibitors.
Figure 5: GSK-3 interacts with the kinome to control cell proliferation.
Figure 6: Summary of drug-targeted kinases affected by GSK-3 activity.

Similar content being viewed by others

References

  1. Doble, B.W. & Woodgett, J.R. GSK-3: tricks of the trade for a multi-tasking kinase. J. Cell Sci. 116, 1175–1186 (2003).

    Article  CAS  Google Scholar 

  2. Kim, L. & Kimmel, A.R. GSK3, a master switch regulating cell-fate specification and tumorigenesis. Curr. Opin. Genet. Dev. 10, 508–514 (2000).

    Article  CAS  Google Scholar 

  3. Pap, M. & Cooper, G.M. Role of glycogen synthase kinase-3 in the phosphatidylinositol 3-kinase/Akt cell survival pathway. J. Biol. Chem. 273, 19929–19932 (1998).

    Article  CAS  Google Scholar 

  4. Sutherland, C. What are the bona fide GSK3 substrates? Int. J. Alzheimers Dis. 2011, 505607 (2011).

    PubMed  PubMed Central  Google Scholar 

  5. Farago, M. et al. Kinase-inactive glycogen synthase kinase 3β promotes Wnt signaling and mammary tumorigenesis. Cancer Res. 65, 5792–5801 (2005).

    Article  CAS  Google Scholar 

  6. Kang, T. et al. GSK-3β targets Cdc25A for ubiquitin-mediated proteolysis, and GSK-3β inactivation correlates with Cdc25A overproduction in human cancers. Cancer Cell 13, 36–47 (2008).

    Article  CAS  Google Scholar 

  7. Leis, H., Segrelles, C., Ruiz, S., Santos, M. & Paramio, J.M. Expression, localization, and activity of glycogen synthase kinase 3β during mouse skin tumorigenesis. Mol. Carcinog. 35, 180–185 (2002).

    Article  CAS  Google Scholar 

  8. Zhang, H.H., Lipovsky, A.I., Dibble, C.C., Sahin, M. & Manning, B.D. S6K1 regulates GSK3 under conditions of mTOR-dependent feedback inhibition of Akt. Mol. Cell 24, 185–197 (2006).

    Article  CAS  Google Scholar 

  9. Zhou, B.P. et al. Dual regulation of Snail by GSK-3β–mediated phosphorylation in control of epithelial-mesenchymal transition. Nat. Cell Biol. 6, 931–940 (2004).

    Article  CAS  Google Scholar 

  10. Cancer Genome Atlas, N. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

    Article  Google Scholar 

  11. Knox, C. et al. DrugBank 3.0: a comprehensive resource for ′omics′ research on drugs. Nucleic Acids Res. 39, D1035–D1041 (2011).

    Article  CAS  Google Scholar 

  12. Roig, A.I. et al. Immortalized epithelial cells derived from human colon biopsies express stem cell markers and differentiate in vitro. Gastroenterology 138, 1012-21.e1–1012-21.e5 (2010).

    Article  Google Scholar 

  13. Doble, B.W., Patel, S., Wood, G.A., Kockeritz, L.K. & Woodgett, J.R. Functional redundancy of GSK-3α and GSK-3β in Wnt/β-catenin signaling shown by using an allelic series of embryonic stem cell lines. Dev. Cell 12, 957–971 (2007).

    Article  CAS  Google Scholar 

  14. Bennett, C.N. et al. Regulation of Wnt signaling during adipogenesis. J. Biol. Chem. 277, 30998–31004 (2002).

    Article  CAS  Google Scholar 

  15. US National Cancer Institutes–National Institutes of Health. Synthetics and Pure Natural Products. Developmental Therapeutics Program NCI/NIH, http://dtp.nci.nih.gov/branches/dscb/oncology_drugset_explanation.html (2014).

  16. Yost, C. et al. The axis-inducing activity, stability, and subcellular distribution of β-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3. Genes Dev. 10, 1443–1454 (1996).

    Article  CAS  Google Scholar 

  17. Liu, C. et al. Control of β-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 108, 837–847 (2002).

    Article  CAS  Google Scholar 

  18. Jacinto, E. et al. Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat. Cell Biol. 6, 1122–1128 (2004).

    Article  CAS  Google Scholar 

  19. Sarbassov, D.D. et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr. Biol. 14, 1296–1302 (2004).

    Article  CAS  Google Scholar 

  20. Efeyan, A. & Sabatini, D.M. mTOR and cancer: many loops in one pathway. Curr. Opin. Cell Biol. 22, 169–176 (2010).

    Article  CAS  Google Scholar 

  21. Sabatini, D.M. mTOR and cancer: insights into a complex relationship. Nat. Rev. Cancer 6, 729–734 (2006).

    Article  CAS  Google Scholar 

  22. Thoreen, C.C. & Sabatini, D.M. Rapamycin inhibits mTORC1, but not completely. Autophagy 5, 725–726 (2009).

    Article  CAS  Google Scholar 

  23. O′Reilly, K.E. et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 66, 1500–1508 (2006).

    Article  Google Scholar 

  24. Chen, C.H. et al. ER stress inhibits mTORC2 and Akt signaling through GSK-3beta-mediated phosphorylation of rictor. Sci. Signal. 4, ra10 (2011).

    Article  Google Scholar 

  25. Huang, J. & Manning, B.D. A complex interplay between Akt, TSC2 and the two mTOR complexes. Biochem. Soc. Trans. 37, 217–222 (2009).

    Article  CAS  Google Scholar 

  26. Thoreen, C.C. et al. An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1. J. Biol. Chem. 284, 8023–8032 (2009).

    Article  CAS  Google Scholar 

  27. Feldman, M.E. et al. Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol. 7, e38 (2009).

    Article  Google Scholar 

  28. Petronczki, M., Lenart, P. & Peters, J.M. Polo on the rise–from mitotic entry to cytokinesis with Plk1. Dev. Cell 14, 646–659 (2008).

    Article  CAS  Google Scholar 

  29. Rudolph, D. et al. BI 6727, a Polo-like kinase inhibitor with improved pharmacokinetic profile and broad antitumor activity. Clin. Cancer Res. 15, 3094–3102 (2009).

    Article  CAS  Google Scholar 

  30. Olmos, D. et al. Phase I study of GSK461364, a specific and competitive Polo-like kinase 1 inhibitor, in patients with advanced solid malignancies. Clin. Cancer Res. 17, 3420–3430 (2011).

    Article  CAS  Google Scholar 

  31. Schöffski, P. Polo-like kinase (PLK) inhibitors in preclinical and early clinical development in oncology. Oncologist 14, 559–570 (2009).

    Article  Google Scholar 

  32. Zheng, H., Saito, H., Masuda, S., Yang, X. & Takano, Y. Phosphorylated GSK3β-Ser9 and EGFR are good prognostic factors for lung carcinomas. Anticancer Res. 27, 3561–3569 (2007).

    PubMed  Google Scholar 

  33. Fiol, C.J. et al. Phosphoserine as a recognition determinant for glycogen synthase kinase-3: phosphorylation of a synthetic peptide based on the G-component of protein phosphatase-1. Arch. Biochem. Biophys. 267, 797–802 (1988).

    Article  CAS  Google Scholar 

  34. Frame, S., Cohen, P. & Biondi, R.M. A common phosphate binding site explains the unique substrate specificity of GSK3 and its inactivation by phosphorylation. Mol. Cell 7, 1321–1327 (2001).

    Article  CAS  Google Scholar 

  35. Thomas, G.M. et al. A GSK3-binding peptide from FRAT1 selectively inhibits the GSK3-catalysed phosphorylation of axin and β-catenin. FEBS Lett. 458, 247–251 (1999).

    Article  CAS  Google Scholar 

  36. Tighe, A., Ray-Sinha, A., Staples, O.D. & Taylor, S.S. GSK-3 inhibitors induce chromosome instability. BMC Cell Biol. 8, 34 (2007).

    Article  Google Scholar 

  37. Hertz, N.T. et al. A neo-substrate that amplifies catalytic activity of Parkinson′s-disease–related kinase PINK1. Cell 154, 737–747 (2013).

    Article  CAS  Google Scholar 

  38. Simpson, G.L., Hughes, J.A., Washio, Y. & Bertrand, S.M. Direct small-molecule kinase activation: novel approaches for a new era of drug discovery. Curr. Opin. Drug Discov. Devel. 12, 585–596 (2009).

    CAS  PubMed  Google Scholar 

  39. Thorne, C.A. et al. Small-molecule inhibition of Wnt signaling through activation of casein kinase 1α. Nat. Chem. Biol. 6, 829–836 (2010).

    Article  CAS  Google Scholar 

  40. Coster, A.D., Wichaidit, C., Rajaram, S., Altschuler, S.J. & Wu, L.F. A simple image correction method for high-throughput microscopy. Nat. Methods 11, 602 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Cobb and members of the Altschuler and Wu laboratories for critical reading of the manuscript; J. Shay (University of Texas Southwestern) for providing the HCEC cell line and guidance on culture techniques, M. Ramirez for experimental support and J. Life for help with high-throughput screening experiments. This research was supported by a Cancer Biology Training Grant T32 CA124334 (C.A.T.), an American Cancer Society–Lakeshore Division Postdoctoral Fellowship (C.A.T.), the National Institute of Health grants K99 DK103126-01 (C.A.T.), CA133253 (S.J.A.), CA184984 (L.F.W.), R01 GM081549 (L.F.W.), CPRIT RP10900 (L.F.W.), and the Welch Foundation I-1619 (S.J.A.) and I-1644 (L.F.W.).

Author information

Authors and Affiliations

Authors

Contributions

C.A.T. designed, performed and analyzed experiments. C.W. designed experiments and performed analysis of data. A.D.C. aided in analysis of data. B.A.P. designed HTS experiments and provided critical reagents. C.A.T., C.W., L.F.W. and S.J.A. wrote the manuscript with advice from all authors. L.F.W. and S.J.A. guided all aspects of this study.

Corresponding authors

Correspondence to Lani F Wu or Steven J Altschuler.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Figures 1–10, Supplementary Table 1 and Supplementary Notes 1–4 (PDF 2419 kb)

Supplementary Data Set 1

DrugBank gene in kinome list (XLSX 118 kb)

Supplementary Data Set 2

Oncology set (XLSX 27 kb)

Supplementary Data Set 3

PKIS (XLS 234 kb)

Supplementary Data Set 4

RNAi (XLSX 26 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Thorne, C., Wichaidit, C., Coster, A. et al. GSK-3 modulates cellular responses to a broad spectrum of kinase inhibitors. Nat Chem Biol 11, 58–63 (2015). https://doi.org/10.1038/nchembio.1690

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1690

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