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:

Design of fast proteolysis-based signaling and logic circuits in mammalian cells

Abstract

Cellular signal transduction is predominantly based on protein interactions and their post-translational modifications, which enable a fast response to input signals. Owing to difficulties in designing new unique protein–protein interactions, designed cellular logic has focused on transcriptional regulation; however, that process has a substantially slower response, because it requires transcription and translation. Here, we present de novo design of modular, scalable signaling pathways based on proteolysis and designed coiled coils (CC) and implemented in mammalian cells. A set of split proteases with highly specific orthogonal cleavage motifs was constructed and combined with strategically positioned cleavage sites and designed orthogonal CC dimerizing domains with tunable affinity for competitive displacement after proteolytic cleavage. This framework enabled the implementation of Boolean logic functions and signaling cascades in mammalian cells. The designed split-protease-cleavable orthogonal-CC-based (SPOC) logic circuits enable response to chemical or biological signals within minutes rather than hours and should be useful for diverse medical and nonmedical applications.

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: Design of the proteolysis-based signaling pathways and orthogonal proteases.
Fig. 2: Design of proteolytic-cleavage-responsive CC interaction modules.
Fig. 3: Design of Boolean logic functions implemented by SPOC logic.
Fig. 4: Multilayer design of proteolysis-based signaling pathways.
Fig. 5: Fast kinetics of the proteolysis-mediated signaling pathway.

Similar content being viewed by others

Data availability

The authors declare that the data supporting the findings of this study are available in the paper and its supplementary information files. A set of plasmids for SPOC logic, comprising the split orthogonal proteases, cycLuc reporters and CC-building modules, have been deposited with Addgene under Addgene IDs 118966, 118967, 118968, 118969, 118970, 119182, 119207, 119208, 119209, 119210, 119211, 119212, 119213, 119214, 119299, 119300, 119302 and 119303. The raw data are available from the corresponding author upon reasonable request.

References

  1. Cohen, G. B., Ren, R. & Baltimore, D. Modular binding domains in signal transduction proteins. Cell 80, 237–248 (1995).

    Article  CAS  Google Scholar 

  2. Nandagopal, N. & Elowitz, M. B. Synthetic biology: integrated gene circuits. Science 333, 1244–1248 (2011).

    Article  CAS  Google Scholar 

  3. Good, M. C., Zalatan, J. G. & Lim, W. A. Scaffold proteins: hubs for controlling the flow of cellular information. Science 332, 680–686 (2011).

    Article  CAS  Google Scholar 

  4. Gordley, R. M. et al. Engineering dynamical control of cell fate switching using synthetic phospho-regulons. Proc. Natl Acad. Sci. USA 113, 13528–13533 (2016).

    Article  CAS  Google Scholar 

  5. Dueber, J. E., Yeh, B. J., Chak, K. & Lim, W. A. Reprogramming control of an allosteric signaling switch through modular recombination. Science 301, 1904–1908 (2003).

    Article  CAS  Google Scholar 

  6. Howard, P. L., Chia, M. C., Del Rizzo, S., Liu, F.-F. & Pawson, T. Redirecting tyrosine kinase signaling to an apoptotic caspase pathway through chimeric adaptor proteins. Proc. Natl Acad. Sci. USA 100, 11267–11272 (2003).

    Article  CAS  Google Scholar 

  7. Lonzaric, J., Fink, T. & Jerala, R. Design and applications of synthetic information processing circuits in mammalian cells. in Synthetic Biology Vol. 2, 1–34 (Royal Society of Chemistry, London, 2018).

  8. Haellman, V. & Fussenegger, M. Synthetic biology: engineering cell-based biomedical devices. Curr. Opin. Biomed. Eng. 4, 50–56 (2017).

    Article  Google Scholar 

  9. Kitada, T., DiAndreth, B., Teague, B. & Weiss, R. Programming gene and engineered-cell therapies with synthetic biology. Science 359, eaad1067 (2018).

    Article  Google Scholar 

  10. Neurath, H. & Walsh, K. A. Role of proteolytic enzymes in biological regulation (a review). Proc. Natl Acad. Sci. USA 73, 3825–3832 (1976).

    Article  CAS  Google Scholar 

  11. Strasser, A., O’Connor, L. & Dixit, V. M. Apoptosis signaling. Annu. Rev. Biochem. 69, 217–245 (2000).

    Article  CAS  Google Scholar 

  12. Kovall, R. A., Gebelein, B., Sprinzak, D. & Kopan, R. The canonical notch signaling pathway: structural and biochemical insights into shape, sugar, and force. Dev. Cell. 41, 228–241 (2017).

    Article  CAS  Google Scholar 

  13. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nat. Methods 6, 917–922 (2009).

    Article  CAS  Google Scholar 

  14. Fernandez-Rodriguez, J. & Voigt, C. A. Post-translational control of genetic circuits using Potyvirus proteases. Nucleic Acids Res. 44, 6493–6502 (2016).

    Article  CAS  Google Scholar 

  15. Majerle, A., Gaber, R., Benčina, M. & Jerala, R. Function-based mutation-resistant synthetic signaling device activated by HIV-1 proteolysis. ACS Synth. Biol. 4, 667–672 (2015).

    Article  CAS  Google Scholar 

  16. Schwarz, K. A., Daringer, N. M., Dolberg, T. B. & Leonard, J. N. Rewiring human cellular input–output using modular extracellular sensors. Nat. Chem. Biol. 13, 202–209 (2017).

    Article  CAS  Google Scholar 

  17. Shekhawat, S. S., Porter, J. R., Sriprasad, A. & Ghosh, I. An autoinhibited coiled-coil design strategy for split-protein protease sensors. J. Am. Chem. Soc. 131, 15284–15290 (2009).

    Article  CAS  Google Scholar 

  18. Stein, V. & Alexandrov, K. Protease-based synthetic sensing and signal amplification. Proc. Natl Acad. Sci. USA 111, 15934–15939 (2014).

    Article  CAS  Google Scholar 

  19. Zheng, N. et al. Specific and efficient cleavage of fusion proteins by recombinant plum pox virus NIa protease. Protein Expr. Purif. 57, 153–162 (2008).

    Article  CAS  Google Scholar 

  20. Yi, L. et al. Engineering of TEV protease variants by yeast ER sequestration screening (YESS) of combinatorial libraries. Proc. Natl Acad. Sci. USA 110, 7229–7234 (2013).

    Article  CAS  Google Scholar 

  21. Seo, J.-K., Choi, H.-S. & Kim, K.-H. Engineering of soybean mosaic virus as a versatile tool for studying protein-protein interactions in soybean. Sci. Rep. 6, 22436 (2016).

    Article  CAS  Google Scholar 

  22. Kanno, A., Yamanaka, Y., Hirano, H., Umezawa, Y. & Ozawa, T. Cyclic luciferase for real-time sensing of caspase-3 activities in living mammals. Angew. Chem. Int. Ed. Engl. 46, 7595–7599 (2007).

    Article  CAS  Google Scholar 

  23. Nunn, C. M. et al. Crystal structure of the active enzyme and its inactive mutant. Russ. J. Bioorganic Chem. 29, 415–418 (2003).

    Article  Google Scholar 

  24. Wehr, M. C. et al. Monitoring regulated protein-protein interactions using split TEV. Nat. Methods 3, 985–993 (2006).

    Article  CAS  Google Scholar 

  25. 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).

    Article  Google Scholar 

  26. Gradišar, H. & Jerala, R. De novo design of orthogonal peptide pairs forming parallel coiled-coil heterodimers. J. Pept. Sci. 17, 100–106 (2011).

    Article  Google Scholar 

  27. Woolfson, D. N. The design of coiled-coil structures and assemblies. Adv. Protein Chem. 70, 79–112 (2005).

    Article  CAS  Google Scholar 

  28. Grigoryan, G. & Keating, A. E. Structural specificity in coiled-coil interactions. Curr. Opin. Struct. Biol. 18, 477–483 (2008).

    Article  CAS  Google Scholar 

  29. Reinke, A. W., Grant, R. A. & Keating, A. E. A synthetic coiled-coil interactome provides heterospecific modules for molecular engineering. J. Am. Chem. Soc. 132, 6025–6031 (2010).

    Article  CAS  Google Scholar 

  30. Gradišar, H. et al. Design of a single-chain polypeptide tetrahedron assembled from coiled-coil segments. Nat. Chem. Biol. 9, 362–366 (2013).

    Article  Google Scholar 

  31. Ljubetič, A. et al. Design of coiled-coil protein-origami cages that self-assemble in vitro and in vivo. Nat. Biotechnol. 35, 1094–1101 (2017).

    PubMed  Google Scholar 

  32. Crick, F. H. C. Is α-keratin a coiled coil? Nature 170, 882–883 (1952).

    Article  CAS  Google Scholar 

  33. Lupas, A. Coiled coils: new structures and new functions. Trends. Biochem. Sci. 21, 375–382 (1996).

    Article  CAS  Google Scholar 

  34. Berrington, W. R., Smith, K. D., Skerrett, S. J. & Hawn, T. R. Nucleotide-binding oligomerization domain containing-like receptor family, caspase recruitment domain (CARD) containing 4 (NLRC4) regulates intrapulmonary replication of aerosolized Legionella pneumophila. BMC Infect. Dis. 13, 371 (2013).

  35. Oakley, M. G. & Hollenbeck, J. J. The design of antiparallel coiled coils. Curr. Opin. Struct. Biol. 11, 450–457 (2001).

    Article  CAS  Google Scholar 

  36. Mittl, P. R. E. et al. The retro-GCN4 leucine zipper sequence forms a stable three-dimensional structure. Proc. Natl Acad. Sci. USA 97, 2562–2566 (2000).

    Article  CAS  Google Scholar 

  37. Drobnak, I., Gradišar, H., Ljubetič, A., Merljak, E. & Jerala, R. Modulation of coiled-coil dimer stability through surface residues while preserving pairing specificity. J. Am. Chem. Soc. 139, 8229–8236 (2017).

    Article  CAS  Google Scholar 

  38. Pace, C. N. & Scholtz, J. M. A helix propensity scale based on experimental studies of peptides and proteins. Biophys. J. 75, 422–427 (1998).

    Article  CAS  Google Scholar 

  39. Phan, J. et al. Structural basis for the substrate specificity of tobacco etch virus protease. J. Biol. Chem. 277, 50564–50572 (2002).

    Article  CAS  Google Scholar 

  40. Riechmann, J. L., Laín, S. & García, J. A. Highlights and prospects of potyvirus molecular biology. J. Gen. Virol. 73, 1–16 (1992).

    Article  CAS  Google Scholar 

  41. Gao, X. J., Chong, L. S., Kim, M. S. & Elowitz, M. B. Programmable protein circuits in living cells. Science 361, 1252–1258 (2018).

    Article  CAS  Google Scholar 

  42. Gibson, D. G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 6, 343–345 (2009).

    Article  CAS  Google Scholar 

  43. Gao, Y. et al. Complex transcriptional modulation with orthogonal and inducible dCas9 regulators. Nat. Methods 13, 1043–1049 (2016).

    Article  CAS  Google Scholar 

  44. Iwai, H., Züger, S., Jin, J. & Tam, P.-H. Highly efficient protein trans-splicing by a naturally split DnaE intein from Nostoc punctiforme. FEBS Lett. 580, 1853–1858 (2006).

    Article  CAS  Google Scholar 

  45. Lacroix, E., Viguera, A. R. & Serrano, L. Elucidating the folding problem of alpha-helices: local motifs, long-range electrostatics, ionic-strength dependence and prediction of NMR parameters. J. Mol. Biol. 284, 173–191 (1998).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The idea and proof of principle for this work were conceived as part of the Slovenian iGEM 2016 project, and members of the team who are not among the authors—M. Meško, M. Mraz, M. Moškon, D. Križaj, R. Krese, N. Franko, L. Magdevska, M. Gradišek, Ž. Pušnik, S. Roškar and K. Cerović—are acknowledged for their contribution. The project was funded by the Slovenian Research Agency (P4-0176 and J3-7034) and an ERC project MaCChines (to R.J.). We thank N. Landau (Division of AIDS, NIAID)) for providing materials.

Author information

Authors and Affiliations

Authors

Contributions

T.F., J.L., A.P., T.P., K.L., N.J. and E.M. performed the experiments and analyzed the results; T.F., J.L. and T.L. designed SPOC logic gates; Ž.S. and F.L. designed antiparallel CCs; M.B., T.F., J.L. and R.J. wrote and edited the manuscript; M.B. supervised the experimental work; and R.J. conceptualized the study and acquired funding.

Corresponding author

Correspondence to Roman Jerala.

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.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–8 and Supplementary Figures 1–15

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fink, T., Lonzarić, J., Praznik, A. et al. Design of fast proteolysis-based signaling and logic circuits in mammalian cells. Nat Chem Biol 15, 115–122 (2019). https://doi.org/10.1038/s41589-018-0181-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-018-0181-6

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