Tagging of the vaccinia virus protein F13 with mCherry causes aberrant virion morphogenesis Open Access

Abstract

Vaccinia virus produces two distinct infectious virions; the single-enveloped intracellular mature virus (IMV), which remains in the cell until cell lysis, and the double-enveloped extracellular enveloped virus (EEV), which mediates virus spread. The latter is derived from a triple-enveloped intracellular enveloped virus (IEV) precursor, which is transported to the cell periphery by the kinesin-1 motor complex. This transport involves the viral protein A36 as well as F12 and E2. A36 is an integral membrane protein associated with the outer virus envelope and is the only known direct link between virion and kinesin-1 complex. Yet in the absence of A36 virion egress still occurs on microtubules, albeit at reduced efficiency. In this paper double-fluorescent labelling of the capsid protein A5 and outer-envelope protein F13 was exploited to visualize IEV transport by live-cell imaging in the absence of either A36 or F12. During the generation of recombinant viruses expressing both A5-GFP and F13-mCherry a plaque size defect was identified that was particularly severe in viruses lacking A36. Electron microscopy showed that this phenotype was caused by abnormal wrapping of IMV to form IEV, and this resulted in reduced virus egress to the cell surface. The aberrant wrapping phenotype suggests that the fluorescent fusion protein interferes with an interaction of F13 with the IMV surface that is required for tight association between IMVs and wrapping membranes. The severity of this defect suggests that these viruses are imperfect tools for characterizing virus egress.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.000917
2017-10-01
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jgv/98/10/2543.html?itemId=/content/journal/jgv/10.1099/jgv.0.000917&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, Anderson DA, Arita I, Jezek Z, Ladnyi ID et al. Smallpox and Its Eradication Geneva: World Health Organisation; 1988
    [Google Scholar]
  3. 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]
  4. Ward BM. Visualization and characterization of the intracellular movement of vaccinia virus intracellular mature virions. J Virol 2005; 79:4755–4763 [View Article][PubMed]
    [Google Scholar]
  5. 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]
  6. 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]
  7. 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]
  8. Smith GL, Law M. The exit of vaccinia virus from infected cells. Virus Res 2004; 106:189–197 [View Article][PubMed]
    [Google Scholar]
  9. 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]
  10. 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]
  11. 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]
  12. 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]
  13. 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]
  14. 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]
  15. 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]
  16. 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]
  17. 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]
  18. 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]
  19. 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]
  20. 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]
  21. 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]
  22. Gao WND, Carpentier DCJ, Ewles HA, Lee SA, Smith GL. Vaccinia virus proteins A36 and F12/E2 show strong preferences for different kinesin light chain isoforms. Traffic 2017; 18:505–518 [View Article][PubMed]
    [Google Scholar]
  23. 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]
  24. 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]
  25. 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]
  26. 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]
  27. 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]
  28. Husain M, Moss B. Vaccinia virus F13L protein with a conserved phospholipase catalytic motif induces colocalization of the B5R envelope glycoprotein in post-Golgi vesicles. J Virol 2001; 75:7528–7542 [View Article][PubMed]
    [Google Scholar]
  29. Ward BM, Moss B. Visualization of intracellular movement of vaccinia virus virions containing a green fluorescent protein-B5R membrane protein chimera. J Virol 2001; 75:4802–4813 [View Article][PubMed]
    [Google Scholar]
  30. 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]
  31. 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]
  32. Hiller G, Weber K. Golgi-derived membranes that contain an acylated viral polypeptide are used for vaccinia virus envelopment. J Virol 1985; 55:651–659[PubMed]
    [Google Scholar]
  33. 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[PubMed]
    [Google Scholar]
  34. 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]
  35. Wolffe EJ, Isaacs SN, Moss B. Deletion of the vaccinia virus B5R gene encoding a 42-kilodalton membrane glycoprotein inhibits extracellular virus envelope formation and dissemination. J Virol 1993; 67:4732–4741[PubMed]
    [Google Scholar]
  36. Blasco R, Moss B. Extracellular vaccinia virus formation and cell-to-cell virus transmission are prevented by deletion of the gene encoding the 37,000-Dalton outer envelope protein. J Virol 1991; 65:5910–5920[PubMed]
    [Google Scholar]
  37. 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]
  38. 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]
  39. Husain M, Moss B. Role of receptor-mediated endocytosis in the formation of vaccinia virus extracellular enveloped particles. J Virol 2005; 79:4080–4089 [View Article][PubMed]
    [Google Scholar]
  40. Schmidt FI, Bleck CK, Helenius A, Mercer J. Vaccinia extracellular virions enter cells by macropinocytosis and acid-activated membrane rupture. EMBO J 2011; 30:3647–3661 [View Article][PubMed]
    [Google Scholar]
  41. Carter GC, Law M, Hollinshead M, Smith GL. Entry of the vaccinia virus intracellular mature virion and its interactions with glycosaminoglycans. J Gen Virol 2005; 86:1279–1290 [View Article][PubMed]
    [Google Scholar]
  42. Carpentier DCJ, van Loggerenberg A, Dieckmann NMG, Smith GL. Vaccinia virus egress mediated by virus protein A36 is reliant on the F12 protein. J Gen Virol 2017; 98:1500–1514 [View Article][PubMed]
    [Google Scholar]
  43. 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]
  44. Hirt P, Hiller G, Wittek R. Localization and fine structure of a vaccinia virus gene encoding an envelope antigen. J Virol 1986; 58:757–764[PubMed]
    [Google Scholar]
  45. 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]
  46. 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]
  47. Schmutz C, Payne LG, Gubser J, Wittek R. A mutation in the gene encoding the vaccinia virus 37,000-Mr protein confers resistance to an inhibitor of virus envelopment and release. J Virol 1991; 65:3435–3442[PubMed]
    [Google Scholar]
  48. Yang G, Pevear DC, Davies MH, Collett MS, Bailey T et al. An orally bioavailable antipoxvirus compound (ST-246) inhibits extracellular virus formation and protects mice from lethal orthopoxvirus challenge. J Virol 2005; 79:13139–13149 [View Article][PubMed]
    [Google Scholar]
  49. Herrera E, Lorenzo MM, Blasco R, Isaacs SN. Functional analysis of vaccinia virus B5R protein: essential role in virus envelopment is independent of a large portion of the extracellular domain. J Virol 1998; 72:294–302[PubMed]
    [Google Scholar]
  50. Lorenzo MM, Herrera E, Blasco R, Isaacs SN. Functional analysis of vaccinia virus B5R protein: role of the cytoplasmic tail. Virology 1998; 252:450–457 [View Article][PubMed]
    [Google Scholar]
  51. Mathew EC, Sanderson CM, Hollinshead R, Smith GL. A mutational analysis of the vaccinia virus B5R protein. J Gen Virol 2001; 82:1199–1213 [View Article][PubMed]
    [Google Scholar]
  52. Mathew E, Sanderson CM, Hollinshead M, Smith GL. The extracellular domain of vaccinia virus protein B5R affects plaque phenotype, extracellular enveloped virus release, and intracellular actin tail formation. J Virol 1998; 72:2429–2438[PubMed]
    [Google Scholar]
  53. Chan WM, Ward BM. There is an A33-dependent mechanism for the incorporation of B5-GFP into vaccinia virus extracellular enveloped virions. Virology 2010; 402:83–93 [View Article][PubMed]
    [Google Scholar]
  54. 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]
  55. 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]
  56. 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]
  57. Husain M, Moss B. Intracellular trafficking of a palmitoylated membrane-associated protein component of enveloped vaccinia virus. J Virol 2003; 77:9008–9019 [View Article][PubMed]
    [Google Scholar]
  58. Marzook NB, Procter DJ, Lynn H, Yamamoto Y, Horsington J et al. Methodology for the efficient generation of fluorescently tagged vaccinia virus proteins. J Vis Exp 2014e51151 [View Article][PubMed]
    [Google Scholar]
  59. Higuchi R, Krummel B, Saiki RK. A general method of in vitro preparation and specific mutagenesis of DNA fragments: study of protein and DNA interactions. Nucleic Acids Res 1988; 16:7351–7367 [View Article][PubMed]
    [Google Scholar]
  60. Hughes SJ, Johnston LH, de Carlos A, Smith GL. Vaccinia virus encodes an active thymidylate kinase that complements a cdc8 mutant of Saccharomyces cerevisiae. J Biol Chem 1991; 266:20103–20109[PubMed]
    [Google Scholar]
  61. Weissman B, Stanbridge EJ. Characterization of ouabain resistant, hypoxanthine phosphoribosyl transferase deficient human cells and their usefulness as a general method for the production of human cell hybrids. Cytogenet Cell Genet 1980; 28:227–239 [View Article][PubMed]
    [Google Scholar]
  62. 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.0.000917
Loading
/content/journal/jgv/10.1099/jgv.0.000917
Loading

Data & Media loading...

Supplements

Supplementary File 1

Supplementary File 2

Supplementary File 3

Supplementary File 4

Supplementary File 5

PDF

Most cited Most Cited RSS feed