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.

  • Protocol
  • Published:

Engineering proteins for allosteric control by light or ligands

Abstract

Control of protein activity in living cells can reveal the role of spatiotemporal dynamics in signaling circuits. Protein analogs with engineered allosteric responses can be particularly effective in the interrogation of protein signaling, as they can replace endogenous proteins with minimal perturbation of native interactions. However, it has been a challenge to identify allosteric sites in target proteins where insertion of responsive domains produces an allosteric response comparable to the activity of native proteins. Here, we describe a detailed protocol to generate genetically encoded analogs of proteins that can be allosterically controlled by either rapamycin or blue light, as well as experimental procedures to produce and test these analogs in vitro and in mammalian cell lines. We describe computational methods, based on crystal structures or homology models, to identify effective sites for insertion of either an engineered rapamycin-responsive (uniRapR) domain or the light-responsive light–oxygen–voltage 2 (LOV2) domain. The inserted domains allosterically regulate the active site, responding to rapamycin with irreversible activation, or to light with reversible inactivation at higher spatial and temporal resolution. These strategies have been successfully applied to catalytic domains of protein kinases, Rho family GTPases, and guanine exchange factors (GEFs), as well as the binding domain of a GEF Vav2. Computational tasks can be completed within a few hours, followed by 1–2 weeks of experimental validation. We provide protocols for computational design, cloning, and experimental testing of the engineered proteins, using Src tyrosine kinase, GEF Vav2, and Rho GTPase Rac1 as examples.

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

Fig. 1: Outline of the procedure.
Fig. 2: Strategies for engineered control of protein activity.
Fig. 3: An approach to the design of allosteric protein switches.
Fig. 4: QuikChange strategy to clone iFKBP, uniRapR, or LOV2 into a target gene.
Fig. 5: An expected result for PI-Vav2 generated using high-content imaging.
Fig. 6: Testing of uniRapR and PI constructs with biochemical and imaging assays.

Similar content being viewed by others

References

  1. Dagliyan, O. et al. Engineering extrinsic disorder to control protein activity in living cells. Science 354, 1441–1444 (2016).

    Article  CAS  Google Scholar 

  2. Karginov, A. V. et al. Dissecting motility signaling through activation of specific Src-effector complexes. Nat. Chem. Biol. 10, 286–290 (2014).

    Article  CAS  Google Scholar 

  3. Chu, P. H. et al. Engineered kinase activation reveals unique morphodynamic phenotypes and associated trafficking for Src family isoforms. Proc. Natl. Acad. Sci. USA 111, 12420–12425 (2014).

    Article  CAS  Google Scholar 

  4. Ostermeier, M. Designing switchable enzymes. Curr. Opin. Struct. Biol. 19, 442–448 (2009).

    Article  CAS  Google Scholar 

  5. Oakes, B. L. et al. Profiling of engineering hotspots identifies an allosteric CRISPR-Cas9 switch. Nat. Biotechnol. 34, 646–651 (2016).

    Article  CAS  Google Scholar 

  6. Edwards, W. R., Busse, K., Allemann, R. K. & Jones, D. D. Linking the functions of unrelated proteins using a novel directed evolution domain insertion method. Nucleic Acids Res. 36, e78 (2008).

    Article  Google Scholar 

  7. Inoue, T., Do Heo, W., 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 

  8. Bayle, J. H. et al. Rapamycin analogs with differential binding specificity permit orthogonal control of protein activity. Chem. Biol. 13, 99–107 (2006).

    Article  CAS  Google Scholar 

  9. 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 

  10. Karginov, A. V., Ding, F., Kota, P., Dokholyan, N. V. & Hahn, K. M. Engineered allosteric activation of kinases in living cells. Nat. Biotechnol. 28, 743–747 (2010).

    Article  CAS  Google Scholar 

  11. Dagliyan, O. et al. Engineering Pak1 allosteric switches. ACS Synth. Biol. 6, 1257–1262 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  13. Dagliyan, O. et al. Rational design of a ligand-controlled protein conformational switch. Proc. Natl. Acad. Sci. USA 110, 6800–6804 (2013).

    Article  CAS  Google Scholar 

  14. Banaszynski, L. A., Chen, L. C., Maynard-Smith, L. A., Ooi, A. G. L. & 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 

  15. Bonger, K. M., Chen, L. C., Liu, C. W. & Wandless, T. J. Small-molecule displacement of a cryptic degron causes conditional protein degradation. Nat. Chem. Biol. 7, 531–537 (2011).

    Article  CAS  Google Scholar 

  16. Kennis, J. T. et al. Primary reactions of the LOV2 domain of phototropin, a plant blue-light photoreceptor. Biochemistry 42, 3385–3392 (2003).

    Article  CAS  Google Scholar 

  17. Peter, E., Dick, B. & Baeurle, S. A. Mechanism of signal transduction of the LOV2-Jα photosensor from Avena sativa. Nat. Commun. 1, 122 (2010).

    Article  Google Scholar 

  18. Harper, S. M., Neil, L. C. & Gardner, K. H. Structural basis of a phototropin light switch. Science 301, 1541–1544 (2003).

    Article  CAS  Google Scholar 

  19. Wu, Y. I. et al. A genetically encoded photoactivatable Rac controls the motility of living cells. Nature 461, 104–108 (2009).

    Article  CAS  Google Scholar 

  20. Wang, X. B., He, L., Wu, Y. I., Hahn, K. M. & Montell, D. J. Light-mediated activation reveals a key role for Rac in collective guidance of cell movement in vivo. Nat. Cell Biol. 12, 591–597 (2010).

    Article  CAS  Google Scholar 

  21. Hayashi-Takagi, A. et al. Labelling and optical erasure of synaptic memory traces in the motor cortex. Nature 525, 333–338 (2015).

    Article  CAS  Google Scholar 

  22. Guo, X. M., Liao, Z. H., Tao, Y. Z., Wang, F. F. & Ma, L. [Optogenetic activation of dorsal hippocampal astrocytic Rac1 blocks the learning of associative memory]. Sheng Li Xue Bao 69, 241–251 (2017).

    PubMed  Google Scholar 

  23. Das, A., Dines, M., Alapin, J. M. & Lamprecht, R. Affecting long-term fear memory formation through optical control of Rac1 GTPase and PAK activity in lateral amygdala. Sci. Rep. 7, 13930 (2017).

    Article  Google Scholar 

  24. Mills, E., Chen, X., Pham, E., Wong, S. & Truong, K. Engineering a photoactivated caspase-7 for rapid induction of apoptosis. ACS Synth. Biol. 1, 75–82 (2012).

    Article  CAS  Google Scholar 

  25. Pham, E., Mills, E. & Truong, K. A synthetic photoactivated protein to generate local or global Ca2+ signals. Chem. Biol. 18, 880–890 (2011).

    Article  CAS  Google Scholar 

  26. Schierling, B. & Pingoud, A. Controlling the DNA cleavage activity of light-inducible chimeric endonucleases by bidirectional photoactivation. Bioconjug. Chem. 23, 1105–1109 (2012).

    Article  CAS  Google Scholar 

  27. Renicke, C., Schuster, D., Usherenko, S., Essen, L. O. & Taxis, C. A LOV2 domain-based optogenetic tool to control protein degradation and cellular function. Chem. Biol. 20, 619–626 (2013).

    Article  CAS  Google Scholar 

  28. Baarlink, C., Wang, H. C. & Grosse, R. Nuclear actin network assembly by formins regulates the SRF coactivator MAL. Science 340, 864–867 (2013).

    Article  CAS  Google Scholar 

  29. Rao, M. V., Chu, P. H., Hahn, K. M. & Zaidel-Bar, R. An optogenetic tool for the activation of endogenous diaphanous-related formins induces thickening of stress fibers without an increase in contractility. Cytoskeleton 70, 394–407 (2013).

    Article  CAS  Google Scholar 

  30. Rivera-Cancel, G., Motta-Mena, L. B. & Gardner, K. H. Identification of natural and artificial DNA substrates for light-activated LOV-HTH transcription factor EL222. Biochemistry 51, 10024–10034 (2012).

    Article  CAS  Google Scholar 

  31. Wang, X., Chen, X. & Yang, Y. Spatiotemporal control of gene expression by a light-switchable transgene system. Nat. Methods 9, 266–269 (2012).

    Article  CAS  Google Scholar 

  32. Liang, F. S., Ho, W. Q. & Crabtree, G. R. Engineering the ABA plant stress pathway for regulation of induced proximity. Sci. Signal. 4, rs2 (2011).

  33. Miyamoto, T. et al. Rapid and orthogonal logic gating with a gibberellin-induced dimerization system. Nat, Chem. Biol. 8, 465–470 (2012).

    Article  CAS  Google Scholar 

  34. Yazawa, M., Sadaghiani, A. M., Hsueh, B. & Dolmetsch, R. E. Induction of protein-protein interactions in live cells using light. Nat. Biotechnol. 27, 941–945 (2009).

    Article  CAS  Google Scholar 

  35. Guntas, G. et al. Engineering an improved light-induced dimer (iLID) for controlling the localization and activity of signaling proteins. Proc. Natl Acad. Sci. 112, 112–117 (2015).

    Article  CAS  Google Scholar 

  36. Kennedy, M. J. et al. Rapid blue-light-mediated induction of protein interactions in living cells. Nat. Methods 7, 973–975 (2010).

    Article  CAS  Google Scholar 

  37. Levskaya, A., Weiner, O. D., Lim, W. A. & Voigt, C. A. Spatiotemporal control of cell signalling using a light-switchable protein interaction. Nature 461, 997–1001 (2009).

    Article  CAS  Google Scholar 

  38. Wang, H. et al. LOVTRAP: an optogenetic system for photoinduced protein dissociation. Nat. Methods 13, 755-758 (2016).

    Article  CAS  Google Scholar 

  39. Lee, S. et al. Reversible protein inactivation by optogenetic trapping in cells. Nat. Methods 11, 633–636 (2014).

    Article  CAS  Google Scholar 

  40. Taslimi, A. et al. An optimized optogenetic clustering tool for probing protein interaction and function. Nat. Commun. 5, 4925 (2014).

    Article  CAS  Google Scholar 

  41. Zhou, X. X., Chung, H. K., Lam, A. J. & Lin, M. Z. Optical control of protein activity by fluorescent protein domains. Science 338, 810–814 (2012).

    Article  CAS  Google Scholar 

  42. Zayner, J. P. & Sosnick, T. R. Factors that control the chemistry of the LOV domain photocycle. PLoS ONE 9, e87074 (2014).

    Article  Google Scholar 

  43. Kanwar, M., Wright, R. C., Date, A., Tullman, J. & Ostermeier, M. Protein switch engineering by domain insertion. Methods Enzymol. 523, 369–388 (2013).

    Article  Google Scholar 

  44. Aydin, Z., Altunbasak, Y. & Borodovsky, M. Protein secondary structure prediction for a single-sequence using hidden semi-Markov models. BMC Bioinformatics 7, 178 (2006).

    Article  Google Scholar 

  45. Yang, J. et al. The I-TASSER Suite: protein structure and function prediction. Nat. Methods 12, 7–8 (2015).

    Article  CAS  Google Scholar 

  46. Webb, B. & Sali, A. Comparative protein structure modeling using MODELLER. Curr. Protoc. Protein Sci. 86, 2.9.1–2.9.37 (2016).

    Article  Google Scholar 

  47. Alford, R. F. et al. The Rosetta all-atom energy function for macromolecular modeling and design. J. Chem. Theory Comput. 13, 3031–3048 (2017).

    Article  CAS  Google Scholar 

  48. Heinig, M. & Frishman, D. STRIDE: a web server for secondary structure assignment from known atomic coordinates of proteins. Nucleic Acids Res. 32, W500–W502 (2004).

    Article  CAS  Google Scholar 

  49. Finn, R. D. et al. Pfam: the protein families database. Nucleic Acids Res. 42, D222–D230 (2014).

    Article  CAS  Google Scholar 

  50. Simonetti, F. L., Teppa, E., Chernomoretz, A., Nielsen, M. & Marino Buslje, C. MISTIC: mutual information server to infer coevolution. Nucleic Acids Res. 41, W8–W14 (2013).

    Article  Google Scholar 

  51. Karginov, A. V. et al. Light regulation of protein dimerization and kinase activity in living cells using photocaged rapamycin and engineered FKBP. J. Am. Chem. Soc. 133, 420–423 (2011).

    Article  CAS  Google Scholar 

  52. Garcia-Mata, R. et al. Analysis of activated GAPs and GEFs in cell lysates. Methods Enzymol. 406, 425–437 (2006).

    Article  CAS  Google Scholar 

  53. Slattery, S. D. & Hahn, K. M. A high-content assay for biosensor validation and for examining stimuli that affect biosensor activity. Curr. Protoc. Cell Biol. 65, 14.15.1–14.15.31 (2014).

    Article  Google Scholar 

  54. Tsygankov, D. et al. CellGeo: a computational platform for the analysis of shape changes in cells with complex geometries. J. Cell Biol. 204, 443–460 (2014).

    Article  CAS  Google Scholar 

  55. Tsygankov, D., Chu, P. H., Chen, H., Elston, T. C. & Hahn, K. M. User-friendly tools for quantifying the dynamics of cellular morphology and intracellular protein clusters. Methods Cell Biol. 123, 409–427 (2014).

    Article  Google Scholar 

  56. Barry, D. J., Durkin, C. H., Abella, J. V. & Way, M. Open source software for quantification of cell migration, protrusions, and fluorescence intensities. J. Cell Biol. 209, 163–180 (2015).

    Article  CAS  Google Scholar 

  57. Roy, A., Kucukural, A. & Zhang, Y. I-TASSER: a unified platform for automated protein structure and function prediction. Nat. Protoc. 5, 725–738 (2010).

    Article  CAS  Google Scholar 

  58. Lyons, P. D., Dunty, J. M., Schaefer, E. M. & Schaller, M. D. Inhibition of the catalytic activity of cell adhesion kinase beta by protein-tyrosine phosphatase-PEST-mediated dephosphorylation. J. Biol. Chem. 276, 24422–24431 (2001).

    Article  CAS  Google Scholar 

  59. Ramachandran, S., Kota, P., Ding, F. & Dokholyan, N. V. Automated minimization of steric clashes in protein structures. Proteins 79, 261–270 (2011).

    Article  CAS  Google Scholar 

  60. Machacek, M. et al. Coordination of Rho GTPase activities during cell protrusion. Nature 461, 99–103 (2009).

    Article  CAS  Google Scholar 

  61. Yan, R., Xu, D., Yang, J., Walker, S. & Zhang, Y. A comparative assessment and analysis of 20 representative sequence alignment methods for protein structure prediction. Sci. Rep. 3, 2619 (2013).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants R35GM122596 (K.M.H.), and R01-GM114015, R01-GM064803, and R01-GM123247 (N.V.D).

Author information

Authors and Affiliations

Authors

Contributions

O.D. developed and optimized the protocol with input from N.V.D and K.M.H. O.D. wrote the protocol with input from K.M.H.

Corresponding author

Correspondence to Klaus M. Hahn.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Dagliyan, O. et al. Science 354, 1441–1444 (2016): https://doi.org/10.1126/science.aah3404

Karginov, A. V. et al. Nat. Chem. Biol. 10, 286–290 (2014): https://doi.org/10.1038/nchembio.1477

Dagliyan, O. et al. Proc. Natl Acad. Sci. USA 110, 6800–6804 (2013): https://doi.org/10.1073/pnas.1218319110

Dagliyan, O. et al. ACS Synth. Biol. 6, 1257–1262 (2017): https://doi.org/10.1021/acssynbio.6b00359

Karginov, A. V., Ding, F., Kota, P., Dokholyan, N. V. & Hahn, K. M. Nat. Biotechnol. 28, 743–747 (2010): https://doi.org/10.1038/nbt.1639

Chu, P.-H. et al. PNAS 111, 12420–12425 (2014): https://doi.org/10.1073/pnas.1404487111

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dagliyan, O., Dokholyan, N.V. & Hahn, K.M. Engineering proteins for allosteric control by light or ligands. Nat Protoc 14, 1863–1883 (2019). https://doi.org/10.1038/s41596-019-0165-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-019-0165-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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