1887

Abstract

Egress of vaccinia virus from its host cell is mediated by the microtubule-associated motor kinesin-1, and three viral proteins, A36 and the F12/E2 complex, have been implicated in this process. Deletion of F12 expression causes a more severe reduction in egress than deletion of A36 but whether these proteins are involved in the same or different mechanisms of kinesin-1 recruitment is unknown. Here it is shown that a virus lacking both proteins forms a smaller plaque than mutants lacking either gene alone, indicating non-redundant functions. A36 not only links virions directly to kinesin-1 but also nucleates actin polymerization to propel surface virions away from the host cell. To address the relative importance of these functions for virus spread, a panel of recombinant viruses was constructed in which the ability of A36 to bind kinesin-1 or to nucleate actin polymerization was abrogated individually or together, in the presence or absence of F12 expression. Analysis of these viruses revealed that in the presence of the F12 protein, loss of kinesin-1 interaction made a greater contribution to plaque size than did the formation of actin tails. However in the absence of F12, the ability of A36 to promote egress was abrogated. Therefore, the ability of A36 to promote egress by kinesin-1 is reliant on the F12 protein.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.000816
2017-06-01
2024-04-25
Loading full text...

Full text loading...

/deliver/fulltext/jgv/98/6/1500.html?itemId=/content/journal/jgv/10.1099/jgv.0.000816&mimeType=html&fmt=ahah

References

  1. Moss B. Poxviridae. In Knipe DM, Howley PM, Cohen JI, Griffin DE, Lamb RA. et al. (editors) Fields Virology Philadelphia, Baltimore, New York, London, Buenos Aires, Hong Kong, Sydney, Tokyo: Wolters Kluwer/Lippincott Williams & Wilkins; 2013
    [Google Scholar]
  2. Fenner F, Wittek R, Dumbell KR. The Orthopoxviruses London: Academic Press Limited; 1989
    [Google Scholar]
  3. Newsome TP, Marzook NB. Viruses that ride on the coat-tails of actin nucleation. Semin Cell Dev Biol 2015; 46:155–163 [View Article][PubMed]
    [Google Scholar]
  4. Roberts KL, Smith GL. Vaccinia virus morphogenesis and dissemination. Trends Microbiol 2008; 16:472–479 [View Article][PubMed]
    [Google Scholar]
  5. Carter GC, Rodger G, Murphy BJ, Law M, Krauss O et al. Vaccinia virus cores are transported on microtubules. J Gen Virol 2003; 84:2443–2458 [View Article][PubMed]
    [Google Scholar]
  6. Liu L, Cooper T, Howley PM, Hayball JD. From crescent to mature virion: vaccinia virus assembly and maturation. Viruses 2014; 6:3787–3808 [View Article][PubMed]
    [Google Scholar]
  7. Condit RC, Moussatche N, Traktman P. In a nutshell: structure and assembly of the vaccinia virion. Adv Virus Res 2006; 66:31–124 [View Article][PubMed]
    [Google Scholar]
  8. Sanderson CM, Hollinshead M, Smith GL. The vaccinia virus A27L protein is needed for the microtubule-dependent transport of intracellular mature virus particles. J Gen Virol 2000; 81:47–58 [View Article][PubMed]
    [Google Scholar]
  9. Tooze J, Hollinshead M, Reis B, Radsak K, Kern H. Progeny vaccinia and human cytomegalovirus particles utilize early endosomal cisternae for their envelopes. Eur J Cell Biol 1993; 60:163–178[PubMed]
    [Google Scholar]
  10. Schmelz M, Sodeik B, Ericsson M, Wolffe EJ, Shida H et al. Assembly of vaccinia virus: the second wrapping cisterna is derived from the trans Golgi network. J Virol 1994; 68:130–147[PubMed]
    [Google Scholar]
  11. Geada MM, Galindo I, Lorenzo MM, Perdiguero B, Blasco R. Movements of vaccinia virus intracellular enveloped virions with GFP tagged to the F13L envelope protein. J Gen Virol 2001; 82:2747–2760 [View Article][PubMed]
    [Google Scholar]
  12. Hollinshead M, Rodger G, van Eijl H, Law M, Hollinshead R et al. Vaccinia virus utilizes microtubules for movement to the cell surface. J Cell Biol 2001; 154:389–402 [View Article][PubMed]
    [Google Scholar]
  13. Ward BM, Moss B. Vaccinia virus intracellular movement is associated with microtubules and independent of actin tails. J Virol 2001; 75:11651–11663 [View Article][PubMed]
    [Google Scholar]
  14. Rietdorf J, Ploubidou A, Reckmann I, Holmström A, Frischknecht F et al. Kinesin-dependent movement on microtubules precedes actin-based motility of vaccinia virus. Nat Cell Biol 2001; 3:992–1000 [View Article][PubMed]
    [Google Scholar]
  15. Leite F, Way M. The role of signalling and the cytoskeleton during vaccinia virus egress. Virus Res 2015; 209:87–99 [View Article][PubMed]
    [Google Scholar]
  16. Payne LG. Significance of extracellular enveloped virus in the in vitro and in vivo dissemination of vaccinia. J Gen Virol 1980; 50:89–100 [View Article][PubMed]
    [Google Scholar]
  17. Vanderplasschen A, Mathew E, Hollinshead M, Sim RB, Smith GL. Extracellular enveloped vaccinia virus is resistant to complement because of incorporation of host complement control proteins into its envelope. Proc Natl Acad Sci USA 1998; 95:7544–7549 [View Article][PubMed]
    [Google Scholar]
  18. Engelstad M, Howard ST, Smith GL. A constitutively expressed vaccinia gene encodes a 42-kDa glycoprotein related to complement control factors that forms part of the extracellular virus envelope. Virology 1992; 188:801–810 [View Article][PubMed]
    [Google Scholar]
  19. Isaacs SN, Wolffe EJ, Payne LG, Moss B. Characterization of a vaccinia virus-encoded 42-kilodalton class I membrane glycoprotein component of the extracellular virus envelope. J Virol 1992; 66:7217–7224[PubMed]
    [Google Scholar]
  20. Roper RL, Wolffe EJ, Weisberg A, Moss B. The envelope protein encoded by the A33R gene is required for formation of actin-containing microvilli and efficient cell-to-cell spread of vaccinia virus. J Virol 1998; 72:4192–4204[PubMed]
    [Google Scholar]
  21. Duncan SA, Smith GL. Identification and characterization of an extracellular envelope glycoprotein affecting vaccinia virus egress. J Virol 1992; 66:1610–1621[PubMed]
    [Google Scholar]
  22. Parkinson JE, Smith GL. Vaccinia virus gene A36R encodes a Mr 43-50 K protein on the surface of extracellular enveloped virus. Virology 1994; 204:376–390 [View Article][PubMed]
    [Google Scholar]
  23. van Eijl H, Hollinshead M, Smith GL. The vaccinia virus A36R protein is a type Ib membrane protein present on intracellular but not extracellular enveloped virus particles. Virology 2000; 271:26–36 [View Article][PubMed]
    [Google Scholar]
  24. Shida H. Nucleotide sequence of the vaccinia virus hemagglutinin gene. Virology 1986; 150:451–462 [View Article][PubMed]
    [Google Scholar]
  25. Grosenbach DW, Ulaeto DO, Hruby DE. Palmitylation of the vaccinia virus 37-kDa major envelope antigen. identification of a conserved acceptor motif and biological relevance. J Biol Chem 1997; 272:1956–1964 [View Article][PubMed]
    [Google Scholar]
  26. Zhang WH, Wilcock D, Smith GL. Vaccinia virus F12L protein is required for actin tail formation, normal plaque size, and virulence. J Virol 2000; 74:11654–11662 [View Article][PubMed]
    [Google Scholar]
  27. Domi A, Weisberg AS, Moss B. Vaccinia virus E2L null mutants exhibit a major reduction in extracellular virion formation and virus spread. J Virol 2008; 82:4215–4226 [View Article][PubMed]
    [Google Scholar]
  28. Dodding MP, Newsome TP, Collinson LM, Edwards C, Way M. An E2-F12 complex is required for intracellular enveloped virus morphogenesis during vaccinia infection. Cell Microbiol 2009; 11:808–824 [View Article][PubMed]
    [Google Scholar]
  29. Van Vliet K, Mohamed MR, Zhang L, Villa NY, Werden SJ et al. Poxvirus proteomics and virus-host protein interactions. Microbiol Mol Biol Rev 2009; 73:730–749 [View Article][PubMed]
    [Google Scholar]
  30. Smith GL, Vanderplasschen A, Law M. The formation and function of extracellular enveloped vaccinia virus. J Gen Virol 2002; 83:2915–2931 [View Article][PubMed]
    [Google Scholar]
  31. Ward BM, Moss B. Vaccinia virus A36R membrane protein provides a direct link between intracellular enveloped virions and the microtubule motor kinesin. J Virol 2004; 78:2486–2493 [View Article][PubMed]
    [Google Scholar]
  32. van Eijl H, Hollinshead M, Rodger G, Zhang WH, Smith GL. The vaccinia virus F12L protein is associated with intracellular enveloped virus particles and is required for their egress to the cell surface. J Gen Virol 2002; 83:195–207 [View Article][PubMed]
    [Google Scholar]
  33. Morgan GW, Hollinshead M, Ferguson BJ, Murphy BJ, Carpentier DC et al. Vaccinia protein F12 has structural similarity to kinesin light chain and contains a motor binding motif required for virion export. PLoS Pathog 2010; 6:e1000785 [View Article][PubMed]
    [Google Scholar]
  34. Hirokawa N, Noda Y, Tanaka Y, Niwa S. Kinesin superfamily motor proteins and intracellular transport. Nat Rev Mol Cell Biol 2009; 10:682–696 [View Article][PubMed]
    [Google Scholar]
  35. Dodding MP, Mitter R, Humphries AC, Way M. A kinesin-1 binding motif in vaccinia virus that is widespread throughout the human genome. EMBO J 2011; 30:4523–4538 [View Article][PubMed]
    [Google Scholar]
  36. Aoyama T, Hata S, Nakao T, Tanigawa Y, Oka C et al. Cayman ataxia protein caytaxin is transported by kinesin along neurites through binding to kinesin light chains. J Cell Sci 2009; 122:4177–4185 [View Article][PubMed]
    [Google Scholar]
  37. Pernigo S, Lamprecht A, Steiner RA, Dodding MP. Structural basis for kinesin-1: cargo recognition. Science 2013; 340:356–359 [View Article][PubMed]
    [Google Scholar]
  38. Donnelly SK, Weisswange I, Zettl M, Way M. WIP provides an essential link between Nck and N-WASP during Arp2/3-dependent actin polymerization. Curr Biol 2013; 23:999–1006 [View Article][PubMed]
    [Google Scholar]
  39. Humphries AC, Donnelly SK, Way M. Cdc42 and the Rho GEF intersectin-1 collaborate with Nck to promote N-WASP-dependent actin polymerisation. J Cell Sci 2014; 127:673–685 [View Article][PubMed]
    [Google Scholar]
  40. Doceul V, Hollinshead M, van der Linden L, Smith GL. Repulsion of superinfecting virions: a mechanism for rapid virus spread. Science 2010; 327:873–876 [View Article][PubMed]
    [Google Scholar]
  41. Doceul V, Hollinshead M, Breiman A, Laval K, Smith GL. Protein B5 is required on extracellular enveloped vaccinia virus for repulsion of superinfecting virions. J Gen Virol 2012; 93:1876–1886 [View Article][PubMed]
    [Google Scholar]
  42. Johnston SC, Ward BM. Vaccinia virus protein F12 associates with intracellular enveloped virions through an interaction with A36. J Virol 2009; 83:1708–1717 [View Article][PubMed]
    [Google Scholar]
  43. Carpentier DC, Gao WN, Ewles H, Morgan GW, Smith GL. Vaccinia virus protein complex F12/E2 interacts with kinesin light chain isoform 2 to engage the kinesin-1 motor complex. PLoS Pathog 2015; 11:e1004723 [View Article][PubMed]
    [Google Scholar]
  44. Herrero-Martínez E, Roberts KL, Hollinshead M, Smith GL. Vaccinia virus intracellular enveloped virions move to the cell periphery on microtubules in the absence of the A36R protein. J Gen Virol 2005; 86:2961–2968 [View Article][PubMed]
    [Google Scholar]
  45. Law M, Hollinshead R, Smith GL. Antibody-sensitive and antibody-resistant cell-to-cell spread by vaccinia virus: role of the A33R protein in antibody-resistant spread. J Gen Virol 2002; 83:209–222 [View Article][PubMed]
    [Google Scholar]
  46. Frischknecht F, Moreau V, Röttger S, Gonfloni S, Reckmann I et al. Actin-based motility of vaccinia virus mimics receptor tyrosine kinase signalling. Nature 1999; 401:926–929 [View Article][PubMed]
    [Google Scholar]
  47. Payne LG, Kristenson K. Mechanism of vaccinia virus release and its specific inhibition by N1-isonicotinoyl-N2-3-methyl-4-chlorobenzoylhydrazine. J Virol 1979; 32:614–622[PubMed]
    [Google Scholar]
  48. Kato N, Eggers HJ, Rolly H. Inhibition of release of vaccinia virus by N1-isonicotinoyl-N2-3-methyl-4-chlorobenzoylhydrazine. J Exp Med 1969; 129:795–808 [View Article][PubMed]
    [Google Scholar]
  49. Horsington J, Lynn H, Turnbull L, Cheng D, Braet F et al. A36-dependent actin filament nucleation promotes release of vaccinia virus. PLoS Pathog 2013; 9:e1003239 [View Article][PubMed]
    [Google Scholar]
  50. Sanderson CM, Frischknecht F, Way M, Hollinshead M, Smith GL. Roles of vaccinia virus EEV-specific proteins in intracellular actin tail formation and low pH-induced cell–cell fusion. J Gen Virol 1998; 79:1415–1425 [View Article][PubMed]
    [Google Scholar]
  51. Wolffe EJ, Weisberg AS, Moss B. Role for the vaccinia virus A36R outer envelope protein in the formation of virus-tipped actin-containing microvilli and cell-to-cell virus spread. Virology 1998; 244:20–26 [View Article][PubMed]
    [Google Scholar]
  52. Sivan G, Weisberg AS, Americo JL, Moss B. Retrograde transport from early endosomes to the trans-Golgi network enables membrane wrapping and egress of vaccinia virus virions. J Virol 2016; 90:8891–8905 [View Article][PubMed]
    [Google Scholar]
  53. Engelstad M, Smith GL. The vaccinia virus 42-kDa envelope protein is required for the envelopment and egress of extracellular virus and for virus virulence. Virology 1993; 194:627–637 [View Article][PubMed]
    [Google Scholar]
  54. Ward BM, Moss B. Golgi network targeting and plasma membrane internalization signals in vaccinia virus B5R envelope protein. J Virol 2000; 74:3771–3780 [View Article][PubMed]
    [Google Scholar]
  55. Hollinshead M, Johns HL, Sayers CL, Gonzalez-Lopez C, Smith GL et al. Endocytic tubules regulated by Rab GTPases 5 and 11 are used for envelopment of herpes simplex virus. EMBO J 2012; 31:4204–4220 [View Article][PubMed]
    [Google Scholar]
  56. Krauss O, Hollinshead R, Hollinshead M, Smith GL. An investigation of incorporation of cellular antigens into vaccinia virus particles. J Gen Virol 2002; 83:2347–2359 [View Article][PubMed]
    [Google Scholar]
  57. Hunt SD, Stephens DJ. The role of motor proteins in endosomal sorting. Biochem Soc Trans 2011; 39:1179–1184 [View Article][PubMed]
    [Google Scholar]
  58. Law M, Hollinshead M, Lee HJ, Smith GL. Yaba-like disease virus protein Y144R, a member of the complement control protein family, is present on enveloped virions that are associated with virus-induced actin tails. J Gen Virol 2004; 85:1279–1290 [View Article][PubMed]
    [Google Scholar]
  59. Boulanger D, Smith T, Skinner MA. Morphogenesis and release of fowlpox virus. J Gen Virol 2000; 81:675–687 [View Article][PubMed]
    [Google Scholar]
  60. Duteyrat JL, Gelfi J, Bertagnoli S. Ultrastructural study of myxoma virus morphogenesis. Arch Virol 2006; 151:2161–2180 [View Article][PubMed]
    [Google Scholar]
  61. Dodding MP, Way M. Nck- and N-WASP-dependent actin-based motility is conserved in divergent vertebrate poxviruses. Cell Host Microbe 2009; 6:536–550 [View Article][PubMed]
    [Google Scholar]
  62. Lorenzo MM, Galindo I, Blasco R. Construction and isolation of recombinant vaccinia virus using genetic markers. Methods Mol Biol 2004; 269:15–30 [View Article][PubMed]
    [Google Scholar]
  63. Falkner FG, Moss B. Transient dominant selection of recombinant vaccinia viruses. J Virol 1990; 64:3108–3111[PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.000816
Loading
/content/journal/jgv/10.1099/jgv.0.000816
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error