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.

Fragment-derived inhibitors of human N-myristoyltransferase block capsid assembly and replication of the common cold virus

Abstract

Rhinoviruses (RVs) are the pathogens most often responsible for the common cold, and are a frequent cause of exacerbations in asthma, chronic obstructive pulmonary disease and cystic fibrosis. Here we report the discovery of IMP-1088, a picomolar dual inhibitor of the human N-myristoyltransferases NMT1 and NMT2, and use it to demonstrate that pharmacological inhibition of host-cell N-myristoylation rapidly and completely prevents rhinoviral replication without inducing cytotoxicity. The identification of cooperative binding between weak-binding fragments led to rapid inhibitor optimization through fragment reconstruction, structure-guided fragment linking and conformational control over linker geometry. We show that inhibition of the co-translational myristoylation of a specific virus-encoded protein (VP0) by IMP-1088 potently blocks a key step in viral capsid assembly, to deliver a low nanomolar antiviral activity against multiple RV strains, poliovirus and foot and-mouth disease virus, and protection of cells against virus-induced killing, highlighting the potential of host myristoylation as a drug target in picornaviral infections.

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

Fig. 1: Proposed pathway for the generation of infectious RV particles in an infected host cell.
Fig. 2: Structure-guided discovery of potent human NMT inhibitors.
Fig. 3: Potent inhibitors of human NMTs inhibit RV myristoylation in infected cells.
Fig. 4: Novel human NMT inhibitors potently and efficiently block RV replication without cytotoxicity.
Fig. 5: IMP-1088 inhibits the production of infectious RV particles by blocking virus assembly.

Similar content being viewed by others

References

  1. Ritchie, A. I. et al. Pathogenesis of viral infection in exacerbations of airway disease. Ann. Am. Thorac. Soc. 12, S115–132 (2015).

    PubMed  Google Scholar 

  2. Kieninger, E. et al. High rhinovirus burden in lower airways of children with cystic fibrosis. Chest 143, 782–790 (2013).

    Article  PubMed  Google Scholar 

  3. Flight, W. G. et al. Incidence and clinical impact of respiratory viruses in adults with cystic fibrosis. Thorax 69, 247–253 (2014).

    Article  PubMed  Google Scholar 

  4. Thibaut, H. J., De Palma, A. M. & Neyts, J. Combating enterovirus replication: state-of-the-art on antiviral research. Biochem. Pharmacol. 83, 185–192 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Jiang, P., Liu, Y., Ma, H. C., Paul, A. V. & Wimmer, E. Picornavirus morphogenesis. Microbiol. Mol. Biol. Rev. 78, 418–437 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Liu, Y. et al. Atomic structure of a rhinovirus C, a virus species linked to severe childhood asthma. Proc. Natl Acad. Sci. USA 113, 8997–9002 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Wright, M. H., Heal, W. P., Mann, D. J. & Tate, E. W. Protein myristoylation in health and disease. J. Chem. Biol. 3, 19–35 (2010).

    Article  PubMed  Google Scholar 

  8. Marc, D., Masson, G., Girard, M. & van der Werf, S. Lack of myristoylation of poliovirus capsid polypeptide VP0 prevents the formation of virions or results in the assembly of noninfectious virus particles. J. Virol. 64, 4099–4107 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Marc, D., Drugeon, G., Haenni, A. L., Girard, M. & van der Werf, S. Role of myristoylation of poliovirus capsid protein VP4 as determined by site-directed mutagenesis of its N-terminal sequence. EMBO J. 8, 2661–2668 (1989).

    Article  CAS  Google Scholar 

  10. Marc, D., Girard, M. & van der Werf, S. A Gly1 to Ala substitution in poliovirus capsid protein VP0 blocks its myristoylation and prevents viral assembly. J. Gen. Virol. 72, 1151–1157 (1991).

    Article  CAS  PubMed  Google Scholar 

  11. Moscufo, N., Simons, J. & Chow, M. Myristoylation is important at multiple stages in poliovirus assembly. J. Virol. 65, 2372–2380 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Ritzefeld, M., Wright, M. H. & Tate, E. W. New developments in probing and targeting protein acylation in malaria, leishmaniasis and African sleeping sickness. Parasitology 145, 157–174 (2018).

    Google Scholar 

  13. Thinon, E. et al. Global profiling of co- and post-translationally N-myristoylated proteomes in human cells. Nat. Commun. 5, 4919 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bell, A. S. et al. Selective inhibitors of protozoan protein N-myristoyltransferases as starting points for tropical disease medicinal chemistry programs. PLoS Negl. Trop. Dis. 6, e1625 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Brannigan, J. A. et al. N-myristoyltransferase from Leishmania donovani: structural and functional characterisation of a potential drug target for visceral leishmaniasis. J. Mol. Biol. 396, 985–999 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Goncalves, V. et al. Discovery of Plasmodium vivax N-myristoyltransferase inhibitors: screening, synthesis, and structural characterization of their binding mode. J. Med. Chem. 55, 3578–3582 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Schärfer, C. et al. Torsion angle preferences in druglike chemical space: a comprehensive guide. J. Med. Chem. 56, 2016–2028 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Goncalves, V. et al. A fluorescence-based assay for N-myristoyltransferase activity. Anal. Biochem. 421, 342–344 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Wright, M. H. et al. Validation of N-myristoyltransferase as an antimalarial drug target using an integrated chemical biology approach. Nat. Chem. 6, 112–121 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Thinon, E., Morales-Sanfrutos, J., Mann, D. J. & Tate, E. W. N-myristoyltransferase inhibition induces ER-stress, cell cycle arrest, and apoptosis in cancer cells. ACS Chem. Biol. 11, 2165–2176 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Broncel, M. et al. Multifunctional reagents for quantitative proteome-wide analysis of protein modification in human cells and dynamic profiling of protein lipidation during vertebrate development. Angew. Chem. Int. Ed. 54, 5948–5951 (2015).

    Article  CAS  Google Scholar 

  22. Broncel, M. et al. Myristoylation profiling in human cells and zebrafish. Data Brief. 4, 379–383 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protocols 11, 2301–2319 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Reddel, H. K. et al. A summary of the new GINA strategy: a roadmap to asthma control. Eur. Respir. J. 46, 622–639 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lee, W. M., Monroe, S. S. & Rueckert, R. R. Role of maturation cleavage in infectivity of picornaviruses: activation of an infectosome. J. Virol. 67, 2110–2122 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Koh, C. et al. Oral prenylation inhibition with lonafarnib in chronic hepatitis D infection: a proof-of-concept randomised, double-blind, placebo-controlled phase 2A trial. Lancet Infect. Dis. 15, 1167–1174 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Vizcaino, J. A. et al. ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat. Biotechnol. 32, 223–226 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors gratefully acknowledge financial support from the Medical Research Council (MRC) of the UK (grant G0900278 and a confidence-in-concept award to Imperial College, MC-PC-14100), the EPSRC (grant EP/F500416/1), Cancer Research UK (grant C29637/A20183), MRC Asthma UK Centre in Allergic Mechanisms of Asthma (grant G1000758) and Imperial Innovations. A.M. is supported by an MRF/Asthma UK Research Grant (MRFAUK-2015-311). S.L.J. is supported by a Chair from Asthma UK (CH11SJ) and is a National Institute of Health Research (NIHR) Senior Investigator. The Pirbright Institute receives strategic funding from the Biotechnology and Biological Research Council of the UK. We thank Diamond Light Source for access to beamlines I04 and I04-1 (proposal nos mx12579, mx7864 and mx9948). We thank E. Cota (Imperial College London) for advice on structure determination, and S. Roberts for expert crystal handling. The authors thank K.-K. Conzelmann (Max von Pettenkofer-Institut) for permission to use the BSR-T7/5 cell line.

Author information

Authors and Affiliations

Authors

Contributions

A.M. and D.P.S. designed and executed the RV infection experiments, with assistance from A.G. and R.S.; A.S.B. designed and synthesized the NMT inhibitors, with E.W.T., J.A.H. and R.J.L.; J.N., A.S.A. and T.J.T. undertook the PV and FMDV infections, and the sucrose gradient fractionation. J.M.-S. undertook chemical proteomics experiments and data analysis, with E.W.T.; I.P.-D., J.A.B. and A.J.W. undertook protein purification, X-ray crystallography and structure refinement. M.R. undertook cell viability and tagging experiments. S.W.R. and I.H.N. undertook SPR experiments and data analysis. S.L.J. provided advice on virus experiments, and access to essential facilities. E.W.T. and R.S. conceived the project and, with A.M. and A.B., directed the project. All of the authors contributed to the writing of the manuscript.

Corresponding authors

Correspondence to Roberto Solari or Edward W. Tate.

Ethics declarations

Competing interests

A.B., E.W.T., R.J.L., J.A.H. and J.A.B. are inventors on a patent application that describes NMT inhibitors that include IMP-1031 and IMP-1088 (Bell, A.S.; Tate, E.W.; Leatherbarrow, R.J.; Hutton, J.A.; Brannigan, J.A., Compounds and their use as inhibitors of N-myristoyl transferase, Patent Cooperation Treaty International Application (2017) WO 2017001812).

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figures 1–15, Table 1, Methods, NMR Spectra and References

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mousnier, A., Bell, A.S., Swieboda, D.P. et al. Fragment-derived inhibitors of human N-myristoyltransferase block capsid assembly and replication of the common cold virus. Nature Chem 10, 599–606 (2018). https://doi.org/10.1038/s41557-018-0039-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41557-018-0039-2

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