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:

Nanoscale polarization of the entry fusion complex of vaccinia virus drives efficient fusion

Abstract

To achieve efficient binding and subsequent fusion, most enveloped viruses encode between one and five proteins1. For many viruses, the clustering of fusion proteins—and their distribution on virus particles—is crucial for fusion activity2,3. Poxviruses, the most complex mammalian viruses, dedicate 15 proteins to binding and membrane fusion4. However, the spatial organization of these proteins and how this influences fusion activity is unknown. Here, we show that the membrane of vaccinia virus is organized into distinct functional domains that are critical for the efficiency of membrane fusion. Using super-resolution microscopy and single-particle analysis, we found that the fusion machinery of vaccinia virus resides exclusively in clusters at virion tips. Repression of individual components of the fusion complex disrupts fusion-machinery polarization, consistent with the reported loss of fusion activity5. Furthermore, we show that displacement of functional fusion complexes from virion tips disrupts the formation of fusion pores and infection kinetics. Our results demonstrate how the protein architecture of poxviruses directly contributes to the efficiency of membrane fusion, and suggest that nanoscale organization may be an intrinsic property of these viruses to assure successful infection.

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: VACV binding and fusion show orientation bias that reflects distinct binding and fusion domains on virions.
Fig. 2: VACV binding and fusion proteins are organized into nanoscale clusters.
Fig. 3: A27 regulates the protein architecture of MV membranes.
Fig. 4: VACV fusion machinery polarization is required for full-fusion efficiency.

Similar content being viewed by others

Data availability

The datasets generated and/or analysed during the current study are available from the corresponding authors on reasonable request.

Code availability

All custom code used for analysis in the current study is available on GitHub at https://github.com/HenriquesLab/fusiontools.

References

  1. Kielian, M. & Rey, F. A. Virus membrane-fusion proteins: more than one way to make a hairpin. Nat. Rev. Microbiol. 4, 67–76 (2006).

    Article  CAS  Google Scholar 

  2. Harrison, S. C. Viral membrane fusion. Nat. Struct. Mol. Biol. 15, 690–698 (2008).

    Article  CAS  Google Scholar 

  3. White, J. M., Delos, S. E., Brecher, M. & Schornberg, K. Structures and mechanisms of viral membrane fusion proteins: multiple variations on a common theme. Crit. Rev. Biochem. Mol. Biol. 43, 189–219 (2008).

    Article  CAS  Google Scholar 

  4. Moss, B. in Fields Virology Vol. 5 (eds Knipe, D. M. & Howley, P. M.) 2906 (Lippincott-Raven, 2007).

  5. Moss, B. Poxvirus cell entry: how many proteins does it take? Viruses 4, 688–707 (2012).

    Article  CAS  Google Scholar 

  6. Hsiao, J. C., Chung, C. S. & Chang, W. Vaccinia virus envelope D8L protein binds to cell surface chondroitin sulfate and mediates the adsorption of intracellular mature virions to cells. J. Virol. 73, 8750–8761 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Lin, C.-L., Chung, C.-S., Heine, H. G. & Chang, W. Vaccinia virus envelope H3L protein binds to cell surface heparan sulfate and is important for intracellular mature virion morphogenesis and virus infection in vitro and in vivo. J. Virol. 74, 3353–3365 (2000).

    Article  CAS  Google Scholar 

  8. Chiu, W.-L., Lin, C.-L., Yang, M.-H., Tzou, D.-L. M. & Chang, W. Vaccinia virus 4c (A26L) protein on intracellular mature virus binds to the extracellular cellular matrix laminin. J. Virol. 81, 2149–2157 (2007).

    Article  CAS  Google Scholar 

  9. Chung, C. S., Hsiao, J. C., Chang, Y. S. & Chang, W. A27L protein mediates vaccinia virus interaction with cell surface heparan sulfate. J. Virol. 72, 1577–1585 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Senkevich, T. G., Ward, B. M. & Moss, B. Vaccinia virus entry into cells is dependent on a virion surface protein encoded by the A28L gene. J. Virol. 78, 2357–2366 (2004).

    Article  CAS  Google Scholar 

  11. Ulaeto, D., Grosenbach, D. & Hruby, D. E. The vaccinia virus 4c and A-type inclusion proteins are specific markers for the intracellular mature virus particle. J. Virol. 70, 3372–3377 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Law, M., Carter, G. C., Roberts, K. L., Hollinshead, M. & Smith, G. L. Ligand-induced and nonfusogenic dissolution of a viral membrane. Proc. Natl Acad. Sci. USA 103, 5989–5994 (2006).

    Article  CAS  Google Scholar 

  13. Schmidt, F. I., Bleck, C. K. E., Helenius, A. & Mercer, J. Vaccinia extracellular virions enter cells by macropinocytosis and acid-activated membrane rupture. EMBO J. 30, 3647–3661 (2011).

    Article  CAS  Google Scholar 

  14. Doms, R. W., Blumenthal, R. & Moss, B. Fusion of intra- and extracellular forms of vaccinia virus with the cell membrane. J. Virol. 64, 4884–4892 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Townsley, A. C., Weisberg, A. S., Wagenaar, T. R. & Moss, B. Vaccinia virus entry into cells via a low-pH-dependent endosomal pathway. J. Virol. 80, 8899–8908 (2006).

    Article  CAS  Google Scholar 

  16. Gustafsson, M. G. L. Surpassing the lateral resolution limit by a factor of two using structured illumination microscopy. J. Microsc. 198, 82–87 (2000).

    Article  CAS  Google Scholar 

  17. Gray, R. D. M. et al. VirusMapper: open-source nanoscale mapping of viral architecture through super-resolution microscopy. Sci. Rep. 6, 29132 (2016).

    Article  CAS  Google Scholar 

  18. Gray, R. D. M., Mercer, J. & Henriques, R. Open-source single-particle analysis for super-resolution microscopy with VirusMapper. J. Vis. Exp. 122, e55471 (2017).

    Google Scholar 

  19. Unger, B. & Traktman, P. Vaccinia virus morphogenesis: A13 phosphoprotein is required for assembly of mature virions. J. Virol. 78, 8885–8901 (2004).

    Article  CAS  Google Scholar 

  20. Senkevich, T. G., Ojeda, S., Townsley, A., Nelson, G. E. & Moss, B. Poxvirus multiprotein entry-fusion complex. Proc. Natl Acad. Sci. USA 102, 18572–18577 (2005).

    Article  CAS  Google Scholar 

  21. Mirzakhanyan, Y. & Gershon, P. The Vaccinia virion: filling the gap between atomic and ultrastructure. PLoS Pathog. 15, e1007508 (2019).

    Article  Google Scholar 

  22. Rust, M. J., Bates, M. & Zhuang, X. Sub-diffraction-limit imaging by stochastic optical reconstruction microscopy (STORM). Nat. Methods 3, 793–796 (2006).

    Article  CAS  Google Scholar 

  23. Su, H.-P., Golden, J. W., Gittis, A. G., Hooper, J. W. & Garboczi, D. N. Structural basis for the binding of the neutralizing antibody, 7D11, to the poxvirus L1 protein. Virology 368, 331–341 (2007).

    Article  CAS  Google Scholar 

  24. Levet, F. et al. SR-Tesseler: a method to segment and quantify localization-based super-resolution microscopy data. Nat. Methods 12, 1065–1071 (2015).

    Article  CAS  Google Scholar 

  25. Laliberte, J. P., Weisberg, A. S. & Moss, B. The membrane fusion step of vaccinia virus entry is cooperatively mediated by multiple viral proteins and host cell components. PLoS Pathog. 7, e1002446 (2011).

    Article  CAS  Google Scholar 

  26. Rodriguez, J. F., Paez, E. & Esteban, M. A 14,000-Mr envelope protein of vaccinia virus is involved in cell fusion and forms covalently linked trimers. J. Virol. 61, 395–404 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Kochan, G., Escors, D., González, J. M., Casasnovas, J. M. & Esteban, M. Membrane cell fusion activity of the vaccinia virus A17-A27 protein complex. Cell. Microbiol. 10, 149–164 (2008).

    CAS  PubMed  Google Scholar 

  28. Sanderson, C. M., Hollinshead, M. & Smith, G. L. The vaccinia virus A27L protein is needed for the microtubule-dependent transport of intracellular mature virus particles. J. Gen. Virol. 81, 47–58 (2000).

    Article  CAS  Google Scholar 

  29. Rodriguez, J. F. & Smith, G. L. IPTG-dependent vaccinia virus: identification of a virus protein enabling virion envelopment by golgi membrane and egress. Nucleic Acids Res. 18, 5347–5351 (1990).

    Article  CAS  Google Scholar 

  30. Vázquez, M. I. & Esteban, M. Identification of functional domains in the 14-kilodalton envelope protein (A27L) of vaccinia virus. J. Virol. 73, 9098–9109 (1999).

    PubMed  PubMed Central  Google Scholar 

  31. Kiskowski, M. A., Hancock, J. F. & Kenworthy, A. K. On the use of Ripley’s K-function and its derivatives to analyze domain size. Biophys. J. 97, 1095–1103 (2009).

    Article  CAS  Google Scholar 

  32. Schmidt, F. I., Kuhn, P., Robinson, T., Mercer, J. & Dittrich, P. S. Single-virus fusion experiments reveal proton influx into vaccinia virions and hemifusion lag times. Biophys. J. 105, 420–431 (2013).

    Article  CAS  Google Scholar 

  33. Zawada, K. E., Okamoto, K. & Kasson, P. M. Influenza hemifusion phenotype depends on membrane context: differences in cell–cell and virus–cell fusion. J. Mol. Biol. 430, 594–601 (2018).

    Article  CAS  Google Scholar 

  34. Chojnacki, J. et al. Maturation-dependent HIV-1 surface protein redistribution revealed by fluorescence nanoscopy. Science 338, 524–528 (2012).

    Article  CAS  Google Scholar 

  35. Mercer, J. & Helenius, A. Vaccinia virus uses macropinocytosis and apoptotic mimicry to enter host cells. Science 320, 531–535 (2008).

    Article  CAS  Google Scholar 

  36. Rizopoulos, Z. et al. Vaccinia virus infection requires maturation of macropinosomes. Traffic 16, 814–831 (2015).

    Article  CAS  Google Scholar 

  37. Huang, C.-Y. et al. A novel cellular protein, VPEF, facilitates vaccinia virus penetration into HeLa cells through fluid phase endocytosis. J. Virol. 82, 7988–7999 (2008).

    Article  CAS  Google Scholar 

  38. Vanderplasschen, A., Hollinshead, M. & Smith, G. L. Intracellular and extracellular vaccinia virions enter cells by different mechanisms. J. Gen. Virol. 79, 877–887 (1998).

    Article  CAS  Google Scholar 

  39. Sandgren, K. J. et al. A differential role for macropinocytosis in mediating entry of the two forms of vaccinia virus into dendritic cells. PLoS Pathog. 6, e1000866 (2010).

    Article  Google Scholar 

  40. Dales, S. The uptake and development of vaccinia virus in strain L cells followed with labeled viral deoxyribonucleic acid. J. Cell Biol. 18, 51–72 (1963).

    Article  CAS  Google Scholar 

  41. Schmidt, F. I., Bleck, C. K. E. & Mercer, J. Poxvirus host cell entry. Curr. Opin. Virol. 2, 20–27 (2012).

    Article  CAS  Google Scholar 

  42. Schmidt, F. I. et al. Vaccinia virus entry is followed by core activation and proteasome-mediated release of the immunomodulatory effector VH1 from lateral bodies. Cell Rep. 4, 464–476 (2013).

    Article  CAS  Google Scholar 

  43. da Fonseca, F. G., Wolffe, E. J., Weisberg, A. & Moss, B. Effects of deletion or stringent repression of the H3L envelope gene on vaccinia virus replication. J. Virol. 74, 7518–7528 (2000).

    Article  Google Scholar 

  44. Ojeda, S., Domi, A. & Moss, B. Vaccinia virus G9 protein is an essential component of the poxvirus entry-fusion complex. J. Virol. 80, 9822–9830 (2006).

    Article  CAS  Google Scholar 

  45. Satheshkumar, P. S. & Moss, B. Characterization of a newly identified 35-amino-acid component of the vaccinia virus entry/fusion complex conserved in all chordopoxviruses. J. Virol. 83, 12822–12832 (2009).

    Article  CAS  Google Scholar 

  46. Aldaz-Carroll, L. et al. Epitope-mapping studies define two major neutralization sites on the vaccinia virus extracellular enveloped virus glycoprotein B5R. J. Virol. 79, 6260–6271 (2005).

    Article  CAS  Google Scholar 

  47. Szymborska, A. et al. Nuclear pore scaffold structure analyzed by super-resolution microscopy and particle averaging. Science 341, 655–658 (2013).

    Article  CAS  Google Scholar 

  48. Heilemann, M. et al. Subdiffraction-resolution fluorescence imaging with conventional fluorescent probes. Angew. Chem. Int. Ed. Engl. 47, 6172–6176 (2008).

    Article  CAS  Google Scholar 

  49. Nahidiazar, L., Agronskaia, A. V., Broertjes, J., van den Broek, B. & Jalink, K. Optimizing imaging conditions for demanding multi-color super resolution localization microscopy. PLoS ONE 11, e0158884 (2016).

    Article  Google Scholar 

  50. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  Google Scholar 

  51. Ovesný, M., Křížek, P., Borkovec, J., Švindrych, Z. & Hagen, G. M. ThunderSTORM: a comprehensive ImageJ plug-in for PALM and STORM data analysis and super-resolution imaging. Bioinformatics 30, 2389–2390 (2014).

    Article  Google Scholar 

  52. Laine, R. F. et al. NanoJ: a high-performance open-source super-resolution microscopy toolbox. J. Phys. D Appl. Phys. 52, 163001 (2019).

    Article  CAS  Google Scholar 

  53. White, J. Cell fusion by Semliki Forest, influenza, and vesicular stomatitis viruses. J. Cell Biol. 89, 674–679 (1981).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Moss and M. Esteban for providing the mutant viruses that were used in this study, and M. Turmaine (UCL Biosciences EM facility) and A. Weston (UCL School of Pharmacy—Electron Microscopy Unit) for use of their coater and SEM, respectively. We also acknowledge Eisenberg members J. C. Whitbeck, C. H. Foo and L. Aldaz-Carrol, of the poxvirus research group, for their help producing and purifying the VACV EFC antibodies. This work was funded by MRC Programme grant (MC_UU_00012/7; to J.M.), the European Research Council (649101, UbiProPox; to J.M.), the MRC (MR/K015826/1; to J.M. and R.H.), Biotechnology and Biological Sciences Research Council (BB/M022374/1, BB/P027431/1 and BB/R000697/1; to R.H.) and the Wellcome Trust (203276/Z/16/Z; to R.H.). R.D.M.G. is funded by the Engineering and Physical Sciences Research Council (EP/M506448/1). D.A. is a Marie Skłodowska-Curie fellow funded by the European Union (750673). C.B. is funded by the MRC LMCB PhD program.

Author information

Authors and Affiliations

Authors

Contributions

R.D.M.G., D.A., R.H. and J.M. conceived the project, designed the experiments and wrote the manuscript. R.D.M.G., D.A., C.B. and M.H. performed the experiments. G.H.C. helped to produce and purify all of the VACV EFC antibodies. I.J.W. and J.J.B. performed the EM. R.D.M.G., D.A. R.H and J.M analysed the data. R.D.M.G., D.A., R.H. and J.M. discussed the results and implications of the findings. All authors discussed the manuscript and provided comments.

Corresponding authors

Correspondence to Ricardo Henriques or Jason Mercer.

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 Information

Supplementary Figs. 1–9, Supplementary Tables 1 and 2.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gray, R.D.M., Albrecht, D., Beerli, C. et al. Nanoscale polarization of the entry fusion complex of vaccinia virus drives efficient fusion. Nat Microbiol 4, 1636–1644 (2019). https://doi.org/10.1038/s41564-019-0488-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0488-4

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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