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:

Selective, rapid and optically switchable regulation of protein function in live mammalian cells

Abstract

The rapid and selective regulation of a target protein within living cells that contain closely related family members is an outstanding challenge. Here we introduce genetically directed bioorthogonal ligand tethering (BOLT) and demonstrate selective inhibition (iBOLT) of protein function. In iBOLT, inhibitor–conjugate/target protein pairs are created where the target protein contains a genetically encoded unnatural amino acid with bioorthogonal reactivity and the inhibitor conjugate contains a complementary bioorthogonal group. iBOLT enables the first rapid and specific inhibition of MEK isozymes, and introducing photoisomerizable linkers in the inhibitor conjugate enables reversible, optical regulation of protein activity (photo-BOLT) in live mammalian cells. We demonstrate that a pan kinase inhibitor conjugate allows selective and rapid inhibition of the lymphocyte specific kinase, indicating the modularity and scalability of BOLT. We anticipate that BOLT will enable the rapid and selective regulation of diverse proteins for which no selective small-molecule ligands exist.

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: Pathways for tethering a ligand conjugate to a protein, and the structures of compounds used in the study.
Figure 2: iBOLT of MEK1(XXX-1) variants by 3.
Figure 3: iBOLT is sensitive to inhibitor potency and linker length in inhibitor tetrazine conjugates.
Figure 4: Reversible optical toggling of protein function in live mammalian cells via photo-BOLT.
Figure 5: iBOLT of MEK2(XXX-1) variants.
Figure 6: Inhibition of MEK1(XXX-1) and MEK2(XXX-1) variants with sunitinib tetrazine conjugate 15.
Figure 7: iBOLT of LCK(250-16)-GFP using conjugate 15 inhibits ZAP70-mCherry phosphorylation and TCR phosphorylation and elicits rapid ZAP70-mCherry dissociation from the membrane.

Similar content being viewed by others

References

  1. Shogren-Knaak, M. A., Alaimo, P. J. & Shokat, K. M. Recent advantages in chemical approaches to the study of biological systems. Annu. Rev. Cell Dev. Biol. 17, 405–433 (2001).

    Article  CAS  Google Scholar 

  2. Knight, Z. A. & Shokat, K. M. Chemical genetics: where genetics and pharmacology meet. Cell 128, 425–430 (2007).

    Article  CAS  Google Scholar 

  3. Schreiber, S. L. Organic synthesis toward small-molecule probes and drugs. Proc. Natl Acad. Sci. USA 108, 6699–6702 (2011).

    Article  CAS  Google Scholar 

  4. Belshaw, P. J., Schoepfer, J. G., Liu, K-Q., Morrison, K. L. & Schreiber, S. L. Rational design of orthogonal receptor–ligand combinations. Angew. Chem. Int. Ed. 34, 2129–2132 (1995).

    Article  CAS  Google Scholar 

  5. 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 

  6. Zheng, H., Al-Ayoubi, A. & Eblen, S. T. MAP Kinase Signaling Protocols Vol. 661 (Springer, 2010).

    Google Scholar 

  7. Erlanson, D. A., Wells, J. A. & Braisted, A. C. Tethering: fragment-based drug discovery. Annu. Rev. Biophys. Biomol. Struct. 33, 199–223 (2004).

    Article  CAS  Google Scholar 

  8. Cravatt, B. F., Wright, A. T. & Kozarich, J. W. Activity-based protein profiling: from enzyme chemistry to proteomic chemistry. Annu. Rev. Biochem. 77, 383–414 (2008).

    Article  CAS  Google Scholar 

  9. Takaoka, Y., Ojida, A. & Hamachi, I. Protein organic chemistry and applications for labeling and engineering in live-cell systems. Angew. Chem. Int. Ed. 52, 4088–4106 (2013).

    Article  CAS  Google Scholar 

  10. Chen, Z., Jing, C., Gallagher, S. S., Sheetz, M. P. & Cornish, V. W. Second-generation covalent TMP-Tag for live cell imaging. J. Am. Chem. Soc. 134, 13692–13699 (2012).

    Article  CAS  Google Scholar 

  11. Liu, Q. et al. Developing irreversible inhibitors of the protein kinase cysteinome. Chem. Biol. 20, 146–159 (2013).

    Article  Google Scholar 

  12. Blair, J. A. et al. Structure-guided development of affinity probes for tyrosine kinases using chemical genetics. Nature Chem. Biol. 3, 229–238 (2007).

    Article  CAS  Google Scholar 

  13. Volgraf, M. et al. Allosteric control of an ionotropic glutamate receptor with an optical switch. Nature Chem. Biol. 2, 47–52 (2006).

    Article  CAS  Google Scholar 

  14. Fehrentz, T., Schönberger, M. & Trauner, D. Optochemical genetics. Angew. Chem. Int. Ed. 50, 12156–12182 (2011).

    Article  CAS  Google Scholar 

  15. Szymański, W., Beierle, J. M., Kistemaker, H. A. V., Velema, W. A. & Feringa, B. L. Reversible photocontrol of biological systems by the incorporation of molecular photoswitches. Chem. Rev. 113, 6114–6178 (2013).

    Article  Google Scholar 

  16. Sletten, E. M. & Bertozzi, C. R. Bioorthogonal chemistry: fishing for selectivity in a sea of functionality. Angew. Chem. Int. Ed. 48, 6974–6998 (2009).

    Article  CAS  Google Scholar 

  17. Lang, K. & Chin, J. W. Bioorthogonal reactions for labeling proteins. ACS Chem. Biol. 9, 16–20 (2014).

    Article  CAS  Google Scholar 

  18. Lang, K. & Chin, J. W. Cellular incorporation of unnatural amino acids and bioorthogonal labeling of proteins. Chem. Rev. 114, 4764–4806 (2014).

    Article  CAS  Google Scholar 

  19. Lang, K. et al. Genetically encoded norbornene directs site-specific cellular protein labelling via a rapid bioorthogonal reaction. Nature Chem. 4, 298–304 (2012).

    Article  CAS  Google Scholar 

  20. Plass, T. et al. Amino acids for Diels–Alder reactions in living cells. Angew. Chem. Int. Ed. 51, 4166–4170 (2012).

    Article  CAS  Google Scholar 

  21. Kaya, E. et al. A genetically encoded norbornene amino acid for the mild and selective modification of proteins in a copper free click reaction. Angew. Chem. Int. Ed. 51, 4466–4469 (2012).

    Article  CAS  Google Scholar 

  22. Seitchik, J. L. et al. Genetically encoded tetrazine amino acid directs rapid site-specific in vivo bioorthogonal ligation with trans-cyclooctenes. J. Am. Chem. Soc. 134, 2898–2901 (2012).

    Article  CAS  Google Scholar 

  23. Lang, K. et al. Genetic encoding of bicyclononynes and trans-cyclooctenes for site-specific protein labeling in vitro and in live mammalian cells via rapid fluorogenic Diels–Alder reactions. J. Am. Chem. Soc. 134, 10317–10320 (2012).

    Article  CAS  Google Scholar 

  24. Elliott, T. S. et al. Proteome labeling and protein identification in specific tissues and at specific developmental stages in an animal. Nature Biotechnol. 32, 465–472 (2014).

    Article  CAS  Google Scholar 

  25. Bianco, A., Townsley, F. M., Greiss, S., Lang, K. & Chin, J. W. Expanding the genetic code of Drosophila melanogaster. Nature Chem. Biol. 8, 748–750 (2012).

    Article  CAS  Google Scholar 

  26. Krishnamurthy, V. M. et al. Carbonic anhydrase as a model for biophysical and physical–organic studies of proteins and protein–ligand binding. Chem. Rev. 108, 946–1051 (2008).

    Article  CAS  Google Scholar 

  27. Vauquelin, G. Simplified models for heterobivalent ligand binding: when are they applicable and which are the factors that affect their target residence time. Naunyn-Schmiedeberg’s Arch. Pharmacol. 386, 949–962 (2013).

    Article  CAS  Google Scholar 

  28. Gargano, J. M., Ngo, T., Kim, J. Y., Acheson, D. W. K. & Lees, W. J. Multivalent inhibition of AB5 toxins. J. Am. Chem. Soc. 123, 12909–12910 (2001).

    Article  CAS  Google Scholar 

  29. Krishnamurthy, V. M., Semetey, V., Bracher, P. J., Shen, N. & Whitesides, G. M. Dependence of effective molarity on linker length for an intramolecular protein–ligand system. J. Am. Chem. Soc. 129, 1312–1320 (2007).

    Article  CAS  Google Scholar 

  30. Hilger, R. A., Scheulen, M. E. & Strumberg, D. The Ras-Raf-MEK-ERK pathway in the treatment of cancer. Oncol. Res. Treat. 25, 511–518 (2002).

    Article  CAS  Google Scholar 

  31. Sebolt-Leopold, J. S. & Herrera, R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nature Rev. Cancer 4, 937–947 (2004).

    Article  CAS  Google Scholar 

  32. Solit, D. B. et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature 439, 358–362 (2006).

    Article  CAS  Google Scholar 

  33. Brott, B. K. et al. Mek2 is a kinase related to Mek1 and Ts differentially expressed in murine tissues. Cell Growth Differ. 4, 921–929 (1993).

    CAS  PubMed  Google Scholar 

  34. Giroux, S. et al. Embryonic death of Mek1-deficient mice reveals a role for this kinase in angiogenesis in the labyrinthine region of the placenta. Curr. Biol. 9, 369–376 (1999).

    Article  CAS  Google Scholar 

  35. Ussar, S. & Voss, T. MEK1 and MEK2, different regulators of the G1/S transition. J. Biol. Chem. 279, 43861–43869 (2004).

    Article  CAS  Google Scholar 

  36. Zhao, Y. & Adjei, A. A. The clinical development of MEK inhibitors. Nature Rev. Clin. Oncol. 11, 385–400 (2014).

    Article  CAS  Google Scholar 

  37. Tecle, H. et al. Beyond the MEK-pocket: can current MEK kinase inhibitors be utilized to synthesize novel type III NCKIs? Does the MEK-pocket exist in kinases other than MEK? Bioorg. Med. Chem. Lett. 19, 226–229 (2009).

    Article  CAS  Google Scholar 

  38. Rice, K. D. et al. Novel carboxamide-based allosteric MEK inhibitors: discovery and optimization efforts toward XL518 (GDC-0973). ACS Med. Chem. Lett. 3, 416–421 (2012).

    Article  CAS  Google Scholar 

  39. Yao, J. Z. et al. Fluorophore targeting to cellular proteins via enzyme-mediated azide ligation and strain-promoted cycloaddition. J. Am. Chem. Soc. 134, 3720–3728 (2012).

    Article  CAS  Google Scholar 

  40. Barrett, S. D., Biwersi, C., Kaufman, M., Tecle, H. & Warmus, J. S. Oxygenated esters of 4-iodo phenylamino benzhydroxamic acid. International patent WO 02/06213 (2002).

  41. Hamon, F., Djedaini-Pilard, F., Barbot, F. & Len, C. Azobenzenes—synthesis and carbohydrate applications. Tetrahedron 65, 10105–10123 (2009).

    Article  CAS  Google Scholar 

  42. Renner, C. & Moroder, L. Azobenzene as conformational switch in model peptides. ChemBioChem 7, 868–878 (2006).

    Article  CAS  Google Scholar 

  43. Beharry, A. A., Wong, L., Tropepe, V. & Woolley, G. A. Fluorescence imaging of azobenzene photoswitching in vivo. Angew. Chem. Int. Ed. 50, 1325–1327 (2011).

    Article  CAS  Google Scholar 

  44. Tang, P. C. et al. Pyrrole substituted 2-indolinone protein kinase inhibitors. US patent 6,573,293 (2003).

  45. Gajiwala, K. S. et al. KIT kinase mutants show unique mechanisms of drug resistance to imatinib and sunitinib in gastrointestinal stromal tumor patients. Proc. Natl Acad. Sci. USA 106, 1542–1547 (2009).

    Article  CAS  Google Scholar 

  46. Palacios, E. H. & Weiss, A. Function of the Src-family kinases, Lck and Fyn, in T-cell development and activation. Oncogene 23, 7990–8000 (2004).

    Article  CAS  Google Scholar 

  47. James, J. R. & Vale, R. D. Biophysical mechanism of T-cell receptor triggering in a reconstituted system. Nature 487, 64–69 (2012).

    Article  CAS  Google Scholar 

  48. Au-Yeung, B. B. et al. The structure, regulation, and function of ZAP-70. Immunol. Rev. 228, 41–57 (2009).

    Article  CAS  Google Scholar 

  49. Schmied, W. H., Elsässer, S. J., Uttamapinant, C. & Chin, J. W. Efficient multisite unnatural amino acid incorporation in mammalian cells via optimized pyrrolysyl tRNA synthetase/tRNA expression and engineered eRF1. J. Am. Chem. Soc. 136, 15577–15583 (2014).

    Article  CAS  Google Scholar 

  50. Sander, J. D. & Joung, J. K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nature Biotechnol. 32, 347–355 (2014).

    Article  CAS  Google Scholar 

  51. Mansour, S. et al. Transformation of mammalian cells by constitutively active MAP kinase kinase. Science 265, 966–970 (1994).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the UK Medical Research Council (grants nos. U105181009 and UD99999908) and the European Research Council. Y-H.T. was supported by an EMBO Long-Term Fellowship and S.E. by a DFG Research Fellowship. J.R.J. is a Sir Henry Dale fellow of The Wellcome Trust and The Royal Society. The authors thank K. Wang, L. Davis and M. Mohan for discussions, S. Elsaesser for plasmids and T. Elliot for compound 16.

Author information

Authors and Affiliations

Authors

Contributions

J.W.C. conceived the project. Y-H.T., S.E. and J.R.J. conceived, designed and performed the experiments. S.E. designed and performed the photo-BOLT experiments. K.L. contributed materials. Y-H.T., S.E., J.W.C. and J.R.J. analysed the data. Y-H.T., S.E. and J.W.C. wrote the paper.

Corresponding author

Correspondence to Jason W. Chin.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information

Supplementary information (PDF 5529 kb)

Supplementary movie

Supplementary movie 1 (MP4 475 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tsai, YH., Essig, S., James, J. et al. Selective, rapid and optically switchable regulation of protein function in live mammalian cells. Nature Chem 7, 554–561 (2015). https://doi.org/10.1038/nchem.2253

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchem.2253

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing