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:

Engineered allosteric activation of kinases in living cells

Abstract

Studies of cellular and tissue dynamics benefit greatly from tools that can control protein activity with specificity and precise timing in living systems. Here we describe an approach to confer allosteric regulation specifically on the catalytic activity of protein kinases. A highly conserved portion of the kinase catalytic domain is modified with a small protein insert that inactivates catalytic activity but does not affect other protein functions (Fig. 1a). Catalytic activity is restored by addition of rapamycin or non-immunosuppresive rapamycin analogs. Molecular modeling and mutagenesis indicate that the protein insert reduces activity by increasing the flexibility of the catalytic domain. Drug binding restores activity by increasing rigidity. We demonstrate the approach by specifically activating focal adhesion kinase (FAK) within minutes in living cells and show that FAK is involved in the regulation of membrane dynamics. Successful regulation of Src and p38 by insertion of the rapamycin-responsive element at the same conserved site used in FAK suggests that our strategy will be applicable to other kinases.

(a) Schematic representation of the approach used to regulate the catalytic activity of FAK. A fragment of FKBP is inserted at a position in the catalytic domain where it abrogates catalytic activity. Binding to rapamycin and FRB restores activity. (b) The truncated fragment of human FKBP12 (amino acids Thr22 through Glu108) inserted into the kinase domain. Blue and red, full-length FKBP12; red, proposed structure of the inserted fragment. The FKBP12 is shown in complex with rapamycin and FRB (cyan). (c) Immunoblot analysis of iFKBP interaction with rapamycin and FRB. Myc-tagged FKBP12 and iFKBP constructs were immunoprecipitated from cells treated for 1 h with either 200 nM rapamycin or ethanol (solvent control). Co-immunoprecipitation of co-expressed GFP-FRB was detected using anti-GFP antibody. (d) Changes in the molecular dynamics of iFKBP upon binding to rapamycin and FRB. Warmer colors and thicker backbone indicate increasing root mean square fluctuation.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 2: Development and biochemical characterization of RapR-FAK.
Figure 3: Activation of FAK catalytic activity initiates large dorsal ruffles through the activation of Src.
Figure 4: Mechanism of regulation by iFKBP; Src regulation.

Similar content being viewed by others

References

  1. Spencer, D.M., Wandless, T.J., Schreiber, S.L. & Crabtree, G.R. Controlling signal transduction with synthetic ligands. Science 262, 1019–1024 (1993).

    Article  CAS  Google Scholar 

  2. Bishop, A.C. et al. A chemical switch for inhibitor-sensitive alleles of any protein kinase. Nature 407, 395–401 (2000).

    Article  CAS  Google Scholar 

  3. Qiao, Y., Molina, H., Pandey, A., Zhang, J. & Cole, P.A. Chemical rescue of a mutant enzyme in living cells. Science 311, 1293–1297 (2006).

    Article  CAS  Google Scholar 

  4. Banaszynski, L.A., Chen, L.C., Maynard-Smith, L.A., Ooi, A.G. & Wandless, T.J. A rapid, reversible, and tunable method to regulate protein function in living cells using synthetic small molecules. Cell 126, 995–1004 (2006).

    Article  CAS  Google Scholar 

  5. Tucker, C.L. & Fields, S. A yeast sensor of ligand binding. Nat. Biotechnol. 19, 1042–1046 (2001).

    Article  CAS  Google Scholar 

  6. Guntas, G., Mansell, T.J., Kim, J.R. & Ostermeier, M. Directed evolution of protein switches and their application to the creation of ligand-binding proteins. Proc. Natl. Acad. Sci. USA 102, 11224–11229 (2005).

    Article  CAS  Google Scholar 

  7. Radley, T.L., Markowska, A.I., Bettinger, B.T., Ha, J.H. & Loh, S.N. Allosteric switching by mutually exclusive folding of protein domains. J. Mol. Biol. 332, 529–536 (2003).

    Article  CAS  Google Scholar 

  8. Zhao, J. & Guan, J.L. Signal transduction by focal adhesion kinase in cancer. Cancer Metastasis Rev. 28, 35–49 (2009).

    Article  Google Scholar 

  9. Gabarra-Niecko, V., Schaller, M.D. & Dunty, J.M. FAK regulates biological processes important for the pathogenesis of cancer. Cancer Metastasis Rev. 22, 359–374 (2003).

    Article  CAS  Google Scholar 

  10. Tilghman, R.W. & Parsons, J.T. Focal adhesion kinase as a regulator of cell tension in the progression of cancer. Semin. Cancer Biol. 18, 45–52 (2008).

    Article  CAS  Google Scholar 

  11. Schlaepfer, D.D., Mitra, S.K. & Ilic, D. Control of motile and invasive cell phenotypes by focal adhesion kinase. Biochim. Biophys. Acta 1692, 77–102 (2004).

    Article  CAS  Google Scholar 

  12. Marquis-Omer, D. et al. Stabilization of the FK506 binding protein by ligand binding. Biochem. Biophys. Res. Commun. 179, 741–748 (1991).

    Article  CAS  Google Scholar 

  13. Lietha, D. et al. Structural basis for the autoinhibition of focal adhesion kinase. Cell 129, 1177–1187 (2007).

    Article  CAS  Google Scholar 

  14. Golubovskaya, V.M., Kweh, F.A. & Cance, W.G. Focal adhesion kinase and cancer. Histol. Histopathol. 24, 503–510 (2009).

    CAS  PubMed  Google Scholar 

  15. Chatzizacharias, N.A., Kouraklis, G.P. & Theocharis, S.E. Clinical significance of FAK expression in human neoplasia. Histol. Histopathol. 23, 629–650 (2008).

    CAS  PubMed  Google Scholar 

  16. Sood, A.K. et al. Biological significance of focal adhesion kinase in ovarian cancer: role in migration and invasion. Am. J. Pathol. 165, 1087–1095 (2004).

    Article  CAS  Google Scholar 

  17. Sieg, D.J. et al. FAK integrates growth-factor and integrin signals to promote cell migration. Nat. Cell Biol. 2, 249–256 (2000).

    Article  CAS  Google Scholar 

  18. Schaller, M.D. et al. Autophosphorylation of the focal adhesion kinase, pp125FAK, directs SH2-dependent binding of pp60src. Mol. Cell. Biol. 14, 1680–1688 (1994).

    Article  CAS  Google Scholar 

  19. Xing, Z. et al. Direct interaction of v-Src with the focal adhesion kinase mediated by the Src SH2 domain. Mol. Biol. Cell 5, 413–421 (1994).

    Article  CAS  Google Scholar 

  20. Veracini, L. et al. Two distinct pools of Src family tyrosine kinases regulate PDGF-induced DNA synthesis and actin dorsal ruffles. J. Cell Sci. 119, 2921–2934 (2006).

    Article  CAS  Google Scholar 

  21. Smart, J.E. et al. Characterization of sites for tyrosine phosphorylation in the transforming protein of Rous sarcoma virus (pp60v-src) and its normal cellular homologue (pp60c-src). Proc. Natl. Acad. Sci. USA 78, 6013–6017 (1981).

    Article  CAS  Google Scholar 

  22. Playford, M.P. & Schaller, M.D. The interplay between Src and integrins in normal and tumor biology. Oncogene 23, 7928–7946 (2004).

    Article  CAS  Google Scholar 

  23. Suetsugu, S., Yamazaki, D., Kurisu, S. & Takenawa, T. Differential roles of WAVE1 and WAVE2 in dorsal and peripheral ruffle formation for fibroblast cell migration. Dev. Cell 5, 595–609 (2003).

    Article  CAS  Google Scholar 

  24. Ding, F. & Dokholyan, N.V. Dynamical roles of metal ions and the disulfide bond in Cu, Zn superoxide dismutase folding and aggregation. Proc. Natl. Acad. Sci. USA 105, 19696–19701 (2008).

    Article  CAS  Google Scholar 

  25. Ding, F., Tsao, D., Nie, H. & Dokholyan, N.V. Ab initio folding of proteins with all-atom discrete molecular dynamics. Structure 16, 1010–1018 (2008).

    Article  CAS  Google Scholar 

  26. Krupa, A., Preethi, G. & Srinivasan, N. Structural modes of stabilization of permissive phosphorylation sites in protein kinases: distinct strategies in Ser/Thr and Tyr kinases. J. Mol. Biol. 339, 1025–1039 (2004).

    Article  CAS  Google Scholar 

  27. Foster, D.A. & Toschi, A. Targeting mTOR with rapamycin: one dose does not fit all. Cell Cycle 8, 1026–1029 (2009).

    Article  CAS  Google Scholar 

  28. Inoue, T., Heo, W.D., Grimley, J.S., Wandless, T.J. & Meyer, T. An inducible translocation strategy to rapidly activate and inhibit small GTPase signaling pathways. Nat. Methods 2, 415–418 (2005).

    Article  CAS  Google Scholar 

  29. Stankunas, K. et al. Conditional protein alleles using knockin mice and a chemical inducer of dimerization. Mol. Cell 12, 1615–1624 (2003).

    Article  CAS  Google Scholar 

  30. Vogel, R., Mammeri, H. & Mallet, J. Lentiviral vectors mediate nonimmunosuppressive rapamycin analog-induced production of secreted therapeutic factors in the brain: regulation at the level of transcription and exocytosis. Hum. Gene Ther. 19, 167–178 (2008).

    Article  CAS  Google Scholar 

  31. Cai, X. et al. Spatial and temporal regulation of focal adhesion kinase activity in living cells. Mol. Cell. Biol. 28, 201–214 (2008).

    Article  CAS  Google Scholar 

  32. Gerwins, P., Blank, J.L. & Johnson, G.L. Cloning of a novel mitogen-activated protein kinase kinase kinase, MEKK4, that selectively regulates the c-Jun amino terminal kinase pathway. J. Biol. Chem. 272, 8288–8295 (1997).

    Article  CAS  Google Scholar 

  33. Sharma, S., Ding, F. & Dokholyan, N.V. Multiscale modeling of nucleosome dynamics. Biophys. J. 92, 1457–1470 (2007).

    Article  CAS  Google Scholar 

  34. Teotico, D.G. et al. Active nuclear receptors exhibit highly correlated AF-2 domain motions. PLOS Comput. Biol. 4, e1000111 (2008).

    Article  Google Scholar 

Download references

Acknowledgements

We thank J. Edwards, D. Dominguez and V. Rao for help with construction and testing of RapR-Src and RapR-p38α constructs, B. Clarke for her design of figures and are grateful to the UNC Cancer Research Fund and the National Institutes of Health for funding (GM64346 and GM057464 to K.M.H.; GM080742 and GM080742- 03S1 to N.V.D.). We acknowledge the following gifts: anti-paxillin antibodies and the construct expressing the GST-tagged N-terminal fragment of paxillin from M. Schaller, Department of Biochemistry, West Virginia University; iRap from T. Wandless, Molecular Pharmacology Department, Stanford University; the construct for myc-tagged mouse FAK from S.K. Hanks, Vanderbilt University Medical Center; the flag-tagged mouse p38α, human FKBP12 and FRB domain of human FRAP1 DNA constructs from G. Johnson, Department of Pharmacology, University of North Carolina at Chapel Hill; AP21967 compound was provided by ARIAD Pharmaceuticals.

Author information

Authors and Affiliations

Authors

Contributions

A.V.K. initiated the project, developed and validated regulation of RapR-kinases and performed the studies of FAK biological function. F.D. performed molecular modeling of FKBP12 variants, RapR-FAK and RapR-Src. P.K. performed biochemical characterization of RapR-p38. N.V.D. coordinated molecular dynamics studies. K.M.H. coordinated the study and wrote the final version of the manuscript, based on contributions from all authors.

Corresponding authors

Correspondence to Nikolay V Dokholyan or Klaus M Hahn.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figs. 1–19 (PDF 3729 kb)

Supplementary Movie S1

HeLa cells co-transfected with GFP-RapR-FAK-YM and mCherry-FRB were filmed for 30 min before and 60 min after addition of rapamycin. Images were taken at one minute intervals. (MOV 1015 kb)

Supplementary Movie S2

HeLa cells co-transfected with GFP-RapR-FAK-YM and mCherry-FRB were pretreated with rapamycin for 1 hour. Cells generating large dorsal protrusions were filmed for 30 min before and 30 min after addition of PP2 compound (5 μM final concentration). Images were taken at one minute intervals. (MOV 669 kb)

Supplementary Movie S3

Molecular dynamics simulations demonstrating free movement of the G-loop (red) and iFKBP insertion (magenta) within RapR-FAK catalytic domain without the binding ligands (top) and in complex with rapamycin and FRB (bottom). The simulations were performed as described in the Materials and Methods section. (MOV 2154 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Karginov, A., Ding, F., Kota, P. et al. Engineered allosteric activation of kinases in living cells. Nat Biotechnol 28, 743–747 (2010). https://doi.org/10.1038/nbt.1639

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.1639

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research