78
Views
9
CrossRef citations to date
0
Altmetric
Review

Understanding orthopoxvirus interference with host immune responses to inform novel vaccine design

Pages 87-95 | Published online: 09 Jan 2014

References

  • Fenner F, Henderson DA, Arita I, Jezek Z, Ladnyi ID. Smallpox and its Eradication. WHO, Geneva, Switzerland (1988).
  • Panicali D, Paoletti E. Construction of poxviruses as cloning vectors: insertion of the thymidine kinase gene from herpes simplex virus into the DNA of infectious vaccinia virus. Proc. Natl Acad. Sci. USA79(16), 4927–4931 (1982).
  • Panicali D, Davis SW, Weinberg RL, Paoletti E. Construction of live vaccines by using genetically engineered poxviruses: biological activity of recombinant vaccinia virus expressing influenza virus hemagglutinin. Proc. Natl Acad. Sci. USA80(17), 5364–5368 (1983).
  • Mackett M, Smith GL, Moss B. Vaccinia virus: a selectable eukaryotic cloning and expression vector. Proc. Natl Acad. Sci. USA79(23), 7415–7419 (1982).
  • Smith GL, Murphy BR, Moss B. Construction and characterization of an infectious vaccinia virus recombinant that expresses the influenza hemagglutinin gene and induces resistance to influenza virus infection in hamsters. Proc. Natl Acad. Sci. USA80(23), 7155–7159 (1983).
  • Breman JG, Henderson DA. Poxvirus dilemmas – monkeypox, smallpox, and biologic terrorism. N. Engl. J. Med.339(8), 556–559 (1998).
  • Stephenson J. Monkeypox outbreak a reminder of emerging infections vulnerabilities. JAMA290(1), 23–24 (2003).
  • Smith GL. Vaccinia virus immune evasion. Immunol. Lett.65(1–2), 55–62 (1999).
  • Moss B, Shisler JL. Immunology 101 at poxvirus U: immune evasion genes. Semin. Immunol.13(1), 59–66 (2001).
  • Seet BT, Johnston JB, Brunetti CR et al. Poxviruses and immune evasion. Annu. Rev. Immunol.21, 377–423 (2003).
  • Haga IR, Bowie AG. Evasion of innate immunity by vaccinia virus. Parasitology130(Suppl.) S11–S25 (2005).
  • Finlay BB, McFadden G. Anti-immunology: evasion of the host immune system by bacterial and viral pathogens. Cell124(4), 767–782 (2006).
  • Ng A, Tscharke DC, Reading PC, Smith GL. The vaccinia virus A41L protein is a soluble 30 kDa glycoprotein that affects virus virulence. J. Gen. Virol.82(Pt 9), 2095–2105 (2001).
  • Bowie A, Kiss-Toth E, Symons JA, Smith GL, Dower SK, O’Neill LA. A46R and A52R from vaccinia virus are antagonists of host IL-1 and Toll-like receptor signaling. Proc. Natl Acad. Sci. USA97(18), 10162–10167 (2000).
  • Xiang Y, Condit RC, Vijaysri S, Jacobs B, Williams BR, Silverman RH. Blockade of interferon induction and action by the E3L double-stranded RNA binding proteins of vaccinia virus. J. Virol.76(10), 5251–5259 (2002).
  • Oie KL, Pickup DJ. Cowpox virus and other members of the orthopoxvirus genus interfere with the regulation of NF-κB activation. Virology288(1), 175–187 (2001).
  • Born TL, Morrison LA, Esteban DJ et al. A poxvirus protein that binds to and inactivates IL-18, and inhibits NK cell response. J. Immunol.164(6), 3246–3254 (2000).
  • Reading PC, Smith GL. Vaccinia virus interleukin-18-binding protein promotes virulence by reducing γ interferon production and natural killer and T-cell activity. J. Virol.77(18), 9960–9968 (2003).
  • Kirwan S, Merriam D, Barsby N, McKinnon A, Burshtyn DN. Vaccinia virus modulation of natural killer cell function by direct infection. Virology347(1), 75–87 (2006).
  • Brutkiewicz RR, Klaus SJ, Welsh RM. Window of vulnerability of vaccinia virus-infected cells to natural killer (NK) cell-mediated cytolysis correlates with enhanced NK cell triggering and is concomitant with a decrease in H-2 class I antigen expression. Nat. Immun.11(4), 203–214 (1992).
  • Engelmayer J, Larsson M, Subklewe M et al. Vaccinia virus inhibits the maturation of human dendritic cells: a novel mechanism of immune evasion. J. Immunol.163(12), 6762–6768 (1999).
  • Jenne L, Hauser C, Arrighi JF, Saurat JH, Hugin AW. Poxvirus as a vector to transduce human dendritic cells for immunotherapy: abortive infection but reduced APC function. Gene Ther.7(18), 1575–1583 (2000).
  • Drillien R, Spehner D, Bohbot A, Hanau D. Vaccinia virus-related events and phenotypic changes after infection of dendritic cells derived from human monocytes. Virology268(2), 471–481 (2000).
  • Nemeckova S, Hainz P, Otahal P, Gabriel P, Sroller V, Kutinova L. Early gene expression of vaccinia virus strains replicating (Praha) and non-replicating (modified vaccinia virus strain Ankara, MVA) in mammalian cells. Acta Virol.45(4), 243–247 (2001).
  • Behboudi S, Moore A, Gilbert SC, Nicoll CL, Hill AV. Dendritic cells infected by recombinant modified vaccinia virus Ankara retain immunogenicity in vivo despite in vitro dysfunction. Vaccine22(31–32), 4326–4331 (2004).
  • Drillien R, Spehner D, Hanau D. Modified vaccinia virus Ankara induces moderate activation of human dendritic cells. J. Gen. Virol.85(Pt 8), 2167–2175 (2004).
  • Deng L, Dai P, Ding W, Granstein RD, Shuman S. Vaccinia virus infection attenuates innate immune responses and antigen presentation by epidermal dendritic cells. J. Virol.80(20), 9977–9987 (2006).
  • Townsend A, Bastin J, Gould K et al. Defective presentation to class I-restricted cytotoxic T lymphocytes in vaccinia-infected cells is overcome by enhanced degradation of antigen. J. Exp. Med.168(4), 1211–1224 (1988).
  • Di Nicola M, Napoli S, Anichini A et al. Dendritic cell viability is decreased after phagocytosis of apoptotic tumor cells induced by staurosporine or vaccinia virus infection. Haematologica88(12), 1396–1404 (2003).
  • Li P, Wang N, Zhou D et al. Disruption of MHC class II-restricted antigen presentation by vaccinia virus. J. Immunol.175(10), 6481–6488 (2005).
  • Larsson M, Fonteneau JF, Somersan S et al. Efficiency of cross presentation of vaccinia virus-derived antigens by human dendritic cells. Eur. J. Immunol.31(12), 3432–3442 (2001).
  • Norbury CC, Princiotta MF, Bacik I et al. Multiple antigen-specific processing pathways for activating naive CD8+ T cells in vivo. J. Immunol.166(7), 4355–4362 (2001).
  • Ramirez MC, Sigal LJ. Macrophages and dendritic cells use the cytosolic pathway to rapidly cross-present antigen from live, vaccinia-infected cells. J. Immunol.169(12), 6733–6742 (2002).
  • Shen X, Wong SB, Buck CB, Zhang J, Siliciano RF. Direct priming and cross-priming contribute differentially to the induction of CD8+ CTL following exposure to vaccinia virus via different routes. J. Immunol.169(8), 4222–4229 (2002).
  • Serna A, Ramirez MC, Soukhanova A, Sigal LJ. Cutting edge: efficient MHC class I cross-presentation during early vaccinia infection requires the transfer of proteasomal intermediates between antigen donor and presenting cells. J. Immunol.171(11), 5668–5672 (2003).
  • Fonteneau JF, Kavanagh DG, Lirvall M et al. Characterization of the MHC class I cross-presentation pathway for cell-associated antigens by human dendritic cells. Blood102(13), 4448–4455 (2003).
  • Buller RM, Chakrabarti S, Cooper JA, Twardzik DR, Moss B. Deletion of the vaccinia virus growth factor gene reduces virus virulence. J. Virol.62(3), 866–874 (1988).
  • Bloom DC, Edwards KM, Hager C, Moyer RW. Identification and characterization of two nonessential regions of the rabbitpox virus genome involved in virulence. J. Virol.65(3), 1530–1542 (1991).
  • Alcami A, Smith GL. A soluble receptor for interleukin-1 β encoded by vaccinia virus: a novel mechanism of virus modulation of the host response to infection. Cell71(1), 153–167 (1992).
  • Isaacs SN, Kotwal GJ, Moss B. Vaccinia virus complement-control protein prevents antibody-dependent complement-enhanced neutralization of infectivity and contributes to virulence. Proc. Natl Acad. Sci. USA89(2), 628–632 (1992).
  • Thompson JP, Turner PC, Ali AN, Crenshaw BC, Moyer RW. The effects of serpin gene mutations on the distinctive pathobiology of cowpox and rabbitpox virus following intranasal inoculation of Balb/c mice. Virology197(1), 328–338 (1993).
  • Senkevich TG, Koonin EV, Buller RM. A poxvirus protein with a RING zinc finger motif is of crucial importance for virulence. Virology198(1), 118–128 (1994).
  • Sroller V, Kutinova L, Nemeckova S, Simonova V, Vonka V. Effect of 3-β-hydroxysteroid dehydrogenase gene deletion on virulence and immunogenicity of different vaccinia viruses and their recombinants. Arch. Virol.143(7), 1311–1320 (1998).
  • Price N, Tscharke DC, Hollinshead M, Smith GL. Vaccinia virus gene B7R encodes an 18-kDa protein that is resident in the endoplasmic reticulum and affects virus virulence. Virology267(1), 65–79 (2000).
  • Gardner JD, Tscharke DC, Reading PC, Smith GL. Vaccinia virus semaphorin A39R is a 50–55 kDa secreted glycoprotein that affects the outcome of infection in a murine intradermal model. J. Gen. Virol.82(Pt 9), 2083–2093 (2001).
  • Verardi PH, Jones LA, Aziz FH, Ahmad S, Yilma TD. Vaccinia virus vectors with an inactivated γ interferon receptor homolog gene (B8R) are attenuated in vivo without a concomitant reduction in immunogenicity. J. Virol.75(1), 11–18 (2001).
  • Symons JA, Adams E, Tscharke DC, Reading PC, Waldmann H, Smith GL. The vaccinia virus C12L protein inhibits mouse IL-18 and promotes virus virulence in the murine intranasal model. J. Gen. Virol.83(Pt 11), 2833–2844 (2002).
  • Bartlett N, Symons JA, Tscharke DC, Smith GL. The vaccinia virus N1L protein is an intracellular homodimer that promotes virulence. J. Gen. Virol.83(Pt 8), 1965–1976 (2002).
  • Reading PC, Khanna A, Smith GL. Vaccinia virus CrmE encodes a soluble and cell surface tumor necrosis factor receptor that contributes to virus virulence. Virology292(2), 285–298 (2002).
  • Reading PC, Moore JB, Smith GL. Steroid hormone synthesis by vaccinia virus suppresses the inflammatory response to infection. J. Exp. Med.197(10), 1269–1278 (2003).
  • Chantrey J, Meyer H, Baxby D et al. Cowpox: reservoir hosts and geographic range. Epidemiol. Infect.122(3), 455–460 (1999).
  • Rosengard AM, Liu Y, Nie Z, Jimenez R. Variola virus immune evasion design: expression of a highly efficient inhibitor of human complement. Proc. Natl Acad. Sci. USA99(13), 8808–8813 (2002).
  • Altenburger W, Suter CP, Altenburger J. Partial deletion of the human host range gene in the attenuated vaccinia virus MVA. Arch. Virol.105(1–2), 15–27 (1989).
  • Meyer H, Sutter G, Mayr A. Mapping of deletions in the genome of the highly attenuated vaccinia virus MVA and their influence on virulence. J. Gen. Virol.72(Pt 5), 1031–1038 (1991).
  • Antoine G, Scheiflinger F, Dorner F, Falkner FG. The complete genomic sequence of the modified vaccinia Ankara strain: comparison with other orthopoxviruses. Virology244(2), 365–396 (1998).
  • Kotwal GJ, Moss B. Vaccinia virus encodes a secretory polypeptide structurally related to complement control proteins. Nature335(6186), 176–178 (1988).
  • Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc. Natl Acad. Sci. USA74(12), 5463–5467 (1977).
  • Sanger F, Coulson AR, Barrell BG, Smith AJ, Roe BA. Cloning in single-stranded bacteriophage as an aid to rapid DNA sequencing. J. Mol. Biol.143(2), 161–178 (1980).
  • Buller RM, Smith GL, Cremer K, Notkins AL, Moss B. Decreased virulence of recombinant vaccinia virus expression vectors is associated with a thymidine kinase-negative phenotype. Nature317(6040), 813–815 (1985).
  • Lee MS, Roos JM, McGuigan LC et al. Molecular attenuation of vaccinia virus: mutant generation and animal characterization. J. Virol.66(5), 2617–2630 (1992).
  • Tartaglia J, Perkus ME, Taylor J et al. NYVAC: a highly attenuated strain of vaccinia virus. Virology188(1), 217–232 (1992).
  • Symons JA, Tscharke DC, Price N, Smith GL. A study of the vaccinia virus interferon-γ receptor and its contribution to virus virulence. J. Gen. Virol.83(Pt 8), 1953–1964 (2002).
  • Legrand FA, Verardi PH, Jones LA, Chan KS, Peng Y, Yilma TD. Induction of potent humoral and cell-mediated immune responses by attenuated vaccinia virus vectors with deleted serpin genes. J. Virol.78(6), 2770–2779 (2004).
  • Kettle S, Blake NW, Law KM, Smith GL. Vaccinia virus serpins B13R (SPI-2) and B22R (SPI-1) encode M(r) 38.5 and 40K, intracellular polypeptides that do not affect virus virulence in a murine intranasal model. Virology206(1), 136–147 (1995).
  • Jackson SS, Ilyinskii P, Philippon V et al. Role of genes that modulate host immune responses in the immunogenicity and pathogenicity of vaccinia virus. J. Virol.79(10), 6554–6559 (2005).
  • Buller RM. The BALB/c mouse as a model to study orthopoxviruses. Curr. Top. Microbiol. Immunol.122, 148–153 (1985).
  • Turner PC, Moyer RW. Poxvirus immune modulators: functional insights from animal models. Virus Res.88(1–2), 35–53 (2002).
  • Tscharke DC, Smith GL. A model for vaccinia virus pathogenesis and immunity based on intradermal injection of mouse ear pinnae. J. Gen. Virol.80(Pt 10), 2751–2755 (1999).
  • Tscharke DC, Reading PC, Smith GL. Dermal infection with vaccinia virus reveals roles for virus proteins not seen using other inoculation routes. J. Gen. Virol.83(Pt 8), 1977–1986 (2002).
  • Staib C, Kisling S, Erfle V, Sutter G. Inactivation of the viral interleukin-1β receptor improves CD8+ T-cell memory responses elicited upon immunization with modified vaccinia virus Ankara. J. Gen. Virol.86(Pt 7), 1997–2006 (2005).
  • Clark RH, Kenyon JC, Bartlett NW, Tscharke DC, Smith GL. Deletion of gene A41L enhances vaccinia virus immunogenicity and vaccine efficacy. J. Gen. Virol.87(Pt 1), 29–38 (2006).
  • Poland GA, Grabenstein JD, Neff JM. The US smallpox vaccination program: a review of a large modern era smallpox vaccination implementation program. Vaccine23(17–18), 2078–2081 (2005).
  • Baxby D, Paoletti E. Potential use of non-replicating vectors as recombinant vaccines. Vaccine10(1), 8–9 (1992).
  • Moss B. Replicating and host-restricted non-replicating vaccinia virus vectors for vaccine development. Dev. Biol. Stand.82, 55–63 (1994).
  • Alcami A, Smith GL. A mechanism for the inhibition of fever by a virus. Proc. Natl Acad. Sci. USA93(20), 11029–11034 (1996).
  • Kim M, Yang H, Kim SK et al. Biochemical and functional analysis of smallpox growth factor (SPGF) and anti-SPGF monoclonal antibodies. J. Biol. Chem.279(24), 25838–25848 (2004).
  • Tscharke DC, Karupiah G, Zhou J et al. Identification of poxvirus CD8+ T cell determinants to enable rational design and characterization of smallpox vaccines. J. Exp. Med.201(1), 95–104 (2005).
  • Terajima M, Cruz J, Leporati AM, Demkowicz WE Jr, Kennedy JS, Ennis FA. Identification of vaccinia CD8+ T-cell epitopes conserved among vaccinia and variola viruses restricted by common MHC class I molecules, HLA-A2 or HLA-B7. Hum. Immunol.67(7), 512–520 (2006).
  • Mathew A, Terajima M, West K et al. Identification of murine poxvirus-specific CD8+ CTL epitopes with distinct functional profiles. J. Immunol.174(4), 2212–2219 (2005).
  • Oseroff C, Kos F, Bui HH et al. HLA class I-restricted responses to vaccinia recognize a broad array of proteins mainly involved in virulence and viral gene regulation. Proc. Natl Acad. Sci. USA102(39), 13980–13985 (2005).
  • Jing L, Chong TM, McClurkan CL, Huang J, Story BT, Koelle DM. Diversity in the acute CD8 T cell response to vaccinia virus in humans. J. Immunol.175(11), 7550–7559 (2005).
  • Snyder JT, Belyakov IM, Dzutsev A, Lemonnier F, Berzofsky JA. Protection against lethal vaccinia virus challenge in HLA-A2 transgenic mice by immunization with a single CD8+ T-cell peptide epitope of vaccinia and variola viruses. J. Virol.78(13), 7052–7060 (2004).
  • Kotwal GJ, Isaacs SN, McKenzie R, Frank MM, Moss B. Inhibition of the complement cascade by the major secretory protein of vaccinia virus. Science250(4982), 827–830 (1990).
  • Blomquist MC, Hunt LT, Barker WC. Vaccinia virus 19-kilodalton protein: relationship to several mammalian proteins, including two growth factors. Proc. Natl Acad. Sci. USA81(23), 7363–7367 (1984).
  • Twardzik DR, Brown JP, Ranchalis JE, Todaro GJ, Moss B. Vaccinia virus-infected cells release a novel polypeptide functionally related to transforming and epidermal growth factors. Proc. Natl Acad. Sci. USA82(16), 5300–5304 (1985).
  • Brown JP, Twardzik DR, Marquardt H, Todaro GJ. Vaccinia virus encodes a polypeptide homologous to epidermal growth factor and transforming growth factor. Nature313(6002), 491–492 (1985).
  • Pickup DJ, Ink BS, Hu W, Ray CA, Joklik WK. Hemorrhage in lesions caused by cowpox virus is induced by a viral protein that is related to plasma protein inhibitors of serine proteases. Proc. Natl Acad. Sci. USA83(20), 7698–7702 (1986).
  • Boursnell ME, Foulds IJ, Campbell JI, Binns MM. Non-essential genes in the vaccinia virus HindIII K fragment: a gene related to serine protease inhibitors and a gene related to the 37K vaccinia virus major envelope antigen. J. Gen. Virol.69(Pt 12), 2995–3003 (1988).
  • Smith GL, Howard ST, Chan YS. Vaccinia virus encodes a family of genes with homology to serine proteinase inhibitors. J. Gen. Virol.70(Pt 9), 2333–2343 (1989).
  • Kotwal GJ, Moss B. Vaccinia virus encodes two proteins that are structurally related to members of the plasma serine protease inhibitor superfamily. J. Virol.63(2), 600–606 (1989).
  • Ray CA, Black RA, Kronheim SR et al. Viral inhibition of inflammation: cowpox virus encodes an inhibitor of the interleukin-1 β converting enzyme. Cell69(4), 597–604 (1992).
  • Komiyama T, Ray CA, Pickup DJ et al. Inhibition of interleukin-1 β converting enzyme by the cowpox virus serpin CrmA. An example of cross-class inhibition. J. Biol. Chem.269(30), 19331–19337 (1994).
  • Kettle S, Alcami A, Khanna A, Ehret R, Jassoy C, Smith GL. Vaccinia virus serpin B13R (SPI-2) inhibits interleukin-1β-converting enzyme and protects virus-infected cells from TNF- and Fas-mediated apoptosis, but does not prevent IL-1β-induced fever. J. Gen. Virol.78(Pt 3), 677–685 (1997).
  • Moon KB, Turner PC, Moyer RW. SPI-1-dependent host range of rabbitpox virus and complex formation with cathepsin G is associated with serpin motifs. J. Virol.73(11), 8999–9010 (1999).
  • Chang HW, Watson JC, Jacobs BL. The E3L gene of vaccinia virus encodes an inhibitor of the interferon-induced, double-stranded RNA-dependent protein kinase. Proc. Natl Acad. Sci. USA89(11), 4825–4829 (1992).
  • Kim YG, Muralinath M, Brandt T et al. A role for Z-DNA binding in vaccinia virus pathogenesis. Proc. Natl Acad. Sci. USA100(12), 6974–6979 (2003).
  • Beattie E, Tartaglia J, Paoletti E. Vaccinia virus-encoded eIF-2 α homolog abrogates the antiviral effect of interferon. Virology183(1), 419–422 (1991).
  • Moore JB, Smith GL. Steroid hormone synthesis by a vaccinia enzyme: a new type of virus virulence factor. EMBO J.11(9), 3490 (1992).
  • Spriggs MK, Hruby DE, Maliszewski CR et al. Vaccinia and cowpox viruses encode a novel secreted interleukin-1-binding protein. Cell71(1), 145–152 (1992).
  • Smith CA, Davis T, Wignall JM et al. T2 open reading frame from the Shope fibroma virus encodes a soluble form of the TNF receptor. Biochem. Biophys. Res. Commun.176(1), 335–342 (1991).
  • Hu FQ, Smith CA, Pickup DJ. Cowpox virus contains two copies of an early gene encoding a soluble secreted form of the type II TNF receptor. Virology204(1), 343–356 (1994).
  • Smith CA, Hu FQ, Smith TD et al. Cowpox virus genome encodes a second soluble homologue of cellular TNF receptors, distinct from CrmB, that binds TNF but not LT α. Virology223(1), 132–147 (1996).
  • Loparev VN, Parsons JM, Knight JC et al. A third distinct tumor necrosis factor receptor of orthopoxviruses. Proc. Natl Acad. Sci. USA95(7), 3786–3791 (1998).
  • Saraiva M, Alcami A. CrmE, a novel soluble tumor necrosis factor receptor encoded by poxviruses. J. Virol.75(1), 226–233 (2001).
  • Upton C, Mossman K, McFadden G. Encoding of a homolog of the IFN-γ receptor by myxoma virus. Science258(5086), 1369–1372 (1992).
  • Alcami A, Smith GL. Vaccinia, cowpox, and camelpox viruses encode soluble γ interferon receptors with novel broad species specificity. J. Virol.69(8), 4633–4639 (1995).
  • Colamonici OR, Domanski P, Sweitzer SM, Larner A, Buller RM. Vaccinia virus B18R gene encodes a type I interferon-binding protein that blocks interferon α transmembrane signaling. J. Biol. Chem.270(27), 15974–15978 (1995).
  • Symons JA, Alcami A, Smith GL. Vaccinia virus encodes a soluble type I interferon receptor of novel structure and broad species specificity. Cell81(4), 551–560 (1995).
  • Novick D, Kim SH, Fantuzzi G, Reznikov LL, Dinarello CA, Rubinstein M. Interleukin-18 binding protein: a novel modulator of the Th1 cytokine response. Immunity10(1), 127–136 (1999).
  • Xiang Y, Moss B. IL-18 binding and inhibition of interferon γ induction by human poxvirus-encoded proteins. Proc. Natl Acad. Sci. USA96(20), 11537–11542 (1999).
  • Smith VP, Bryant NA, Alcami A. Ectromelia, vaccinia and cowpox viruses encode secreted interleukin-18-binding proteins. J. Gen. Virol.81(Pt 5), 1223–1230 (2000).
  • Smith CA, Smith TD, Smolak PJ et al. Poxvirus genomes encode a secreted, soluble protein that preferentially inhibits β chemokine activity yet lacks sequence homology to known chemokine receptors. Virology236(2), 316–327 (1997).
  • Graham KA, Lalani AS, Macen JL et al. The T1/35kDa family of poxvirus-secreted proteins bind chemokines and modulate leukocyte influx into virus-infected tissues. Virology229(1), 12–24 (1997).
  • Alcami A, Symons JA, Collins PD, Williams TJ, Smith GL. Blockade of chemokine activity by a soluble chemokine binding protein from vaccinia virus. J. Immunol.160(2), 624–633 (1998).
  • Saraiva M, Smith P, Fallon PG, Alcami A. Inhibition of type 1 cytokine-mediated inflammation by a soluble CD30 homologue encoded by ectromelia (mousepox) virus. J. Exp. Med.196(6), 829–839 (2002).
  • Panus JF, Smith CA, Ray CA, Smith TD, Patel DD, Pickup DJ. Cowpox virus encodes a fifth member of the tumor necrosis factor receptor family: a soluble, secreted CD30 homologue. Proc. Natl Acad. Sci. USA99(12), 8348–8353 (2002).
  • Brooks MA, Ali AN, Turner PC, Moyer RW. A rabbitpox virus serpin gene controls host range by inhibiting apoptosis in restrictive cells. J. Virol.69(12), 7688–7698 (1995).
  • Tewari M, Telford WG, Miller RA, Dixit VM. CrmA, a poxvirus-encoded serpin, inhibits cytotoxic T-lymphocyte-mediated apoptosis. J. Biol. Chem.270(39), 22705–22708 (1995).
  • Dobbelstein M, Shenk T. Protection against apoptosis by the vaccinia virus SPI-2 (B13R) gene product. J. Virol.70(9), 6479–6485 (1996).
  • Brick DJ, Burke RD, Minkley AA, Upton C. Ectromelia virus virulence factor p28 acts upstream of caspase-3 in response to UV light-induced apoptosis. J. Gen. Virol.81(Pt 4), 1087–1097 (2000).
  • Wasilenko ST, Meyers AF, Van der Helm K, Barry M. Vaccinia virus infection disarms the mitochondrion-mediated pathway of the apoptotic cascade by modulating the permeability transition pore. J. Virol.75(23), 11437–11448 (2001).
  • Wasilenko ST, Stewart TL, Meyers AF, Barry M. Vaccinia virus encodes a previously uncharacterized mitochondrial-associated inhibitor of apoptosis. Proc. Natl Acad. Sci. USA100(24), 14345–14350 (2003).
  • DiPerna G, Stack J, Bowie AG et al. Poxvirus protein N1L targets the I-κB kinase complex, inhibits signaling to NF-κB by the tumor necrosis factor superfamily of receptors, and inhibits NF-κB and IRF3 signaling by Toll-like receptors. J. Biol. Chem.279(35), 36570–36578 (2004).
  • Shisler JL, Jin XL. The vaccinia virus K1L gene product inhibits host NF-κB activation by preventing IκBα degradation. J. Virol.78(7), 3553–3560 (2004).
  • Najarro P, Traktman P, Lewis JA. Vaccinia virus blocks γ interferon signal transduction: viral VH1 phosphatase reverses Stat1 activation. J. Virol.75(7), 3185–3196 (2001).
  • Harte MT, Haga IR, Maloney G et al. The poxvirus protein A52R targets Toll-like receptor signaling complexes to suppress host defense. J. Exp. Med.197(3), 343–351 (2003).
  • Comeau MR, Johnson R, DuBose RF et al. A poxvirus-encoded semaphorin induces cytokine production from monocytes and binds to a novel cellular semaphorin receptor, VESPR. Immunity8(4), 473–482 (1998).
  • Webb TJ, Litavecz RA, Khan MA et al. Inhibition of CD1d1-mediated antigen presentation by the vaccinia virus B1R and H5R molecules. Eur. J. Immunol.36(10) 2595–2600 (2006).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.