1932

Abstract

My scientific career started at an extraordinary time, shortly after the discoveries of the helical structure of DNA, the central dogma of DNA to RNA to protein, and the genetic code. Part I of this series emphasizes my education and early studies highlighted by the isolation and characterization of numerous vaccinia virus enzymes, determination of the cap structure of messenger RNA, and development of poxviruses as gene expression vectors for use as recombinant vaccines. Here I describe a shift in my research focus to combine molecular biology and genetics for a comprehensive understanding of poxvirus biology. The dominant paradigm during the early years was to select a function, isolate the responsible proteins, and locate the corresponding gene, whereas later the common paradigm was to select a gene, make a mutation, and determine the altered function. Motivations, behind-the-scenes insights, importance of new technologies, and the vital roles of trainees and coworkers are emphasized.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-virology-021020-100558
2020-09-29
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/virology/7/1/annurev-virology-021020-100558.html?itemId=/content/journals/10.1146/annurev-virology-021020-100558&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Moss B. 2017. Investigating viruses during the transformation of molecular biology. J. Biol. Chem. 292:103958–69
    [Google Scholar]
  2. 2. 
    Moss B, Ingram VM. 1968. Hemoglobin synthesis during amphibian metamorphosis. I. Chemical studies on the hemoglobins from the larval and adult stages of Rana catesbeiana. J. Mol. . Biol 32:481–92
    [Google Scholar]
  3. 3. 
    Moss B, Ingram VM. 1968. Hemoglobin synthesis during amphibian metamorphosis. II. Synthesis of adult hemoglobin following thyroxine administration. J. Mol. Biol. 32:493–502
    [Google Scholar]
  4. 4. 
    Moss B, Rosenblum EN, Paoletti E 1973. Polyadenylate polymerase from vaccinia virions. Nat. New Biol. 245:59–63
    [Google Scholar]
  5. 5. 
    Paoletti E, Moss B. 1974. Two nucleic acid-dependent nucleoside triphosphate phosphohydrolases from vaccinia virus. Nucleotide substrate and polynucleotide cofactor specificities. J. Biol. Chem. 249:3281–86
    [Google Scholar]
  6. 6. 
    Paoletti E, Rosemond-Hornbeak H, Moss B 1974. Two nucleic acid-dependent nucleoside triphosphate phosphohydrolases from vaccinia virus: purification and characterization. J. Biol. Chem. 249:3273–80
    [Google Scholar]
  7. 7. 
    Rosemond-Hornbeak H, Paoletti E, Moss B 1974. Single-stranded deoxyribonucleic acid-specific nuclease from vaccinia virus. Purification and characterization. J. Biol. Chem. 249:3287–91
    [Google Scholar]
  8. 8. 
    Kleiman JH, Moss B. 1975. Purification of a protein kinase and two phosphate acceptor proteins from vaccinia virions. J. Biol. Chem. 250:2420–29
    [Google Scholar]
  9. 9. 
    Wei CM, Moss B. 1974. Methylation of newly synthesized viral messenger RNA by an enzyme in vaccinia virus. PNAS 71:3014–18
    [Google Scholar]
  10. 10. 
    Wei CM, Moss B. 1975. Methylated nucleotides block 5′-terminus of vaccinia virus mRNA. PNAS 72:318–22
    [Google Scholar]
  11. 11. 
    Wei CM, Gershowitz A, Moss B 1975. Methylated nucleotides block 5′ terminus of HeLa cell messenger RNA. Cell 4:379–86
    [Google Scholar]
  12. 12. 
    Wei CM, Gershowitz A, Moss B 1975. N6, O2′-dimethyladenosine a novel methylated ribonucleoside next to the 5′ terminal of animal cell and virus mRNAs. Nature 257:251–53
    [Google Scholar]
  13. 13. 
    Wei CM, Gershowitz A, Moss B 1976. 5′-terminal and internal methylated nucleotide sequences in HeLa cell mRNA. Biochemistry 15:397–401
    [Google Scholar]
  14. 14. 
    Wei CM, Moss B. 1977. Nucleotide sequences at the N6-methyladenosine sites of HeLa cell messenger ribonucleic acid. Biochemistry 16:1672–76
    [Google Scholar]
  15. 15. 
    Furuichi Y, Morgan M, Muthukrishnan S, Shatkin AJ 1975. Reovirus messenger RNA contains a methylated, blocked 5′-terminal structure: m-7G(5′)ppp(5′)G-MpCp. PNAS 72:362–66
    [Google Scholar]
  16. 16. 
    Martin SA, Moss B. 1975. Modification of RNA by mRNA guanylytransferase and mRNA (guanine-7-)methyl-transferase from vaccinia virions. J. Biol. Chem. 250:9330–35
    [Google Scholar]
  17. 17. 
    Martin SA, Paoletti E, Moss B 1975. Purification of mRNA guanylyltransferase and mRNA (guanine 7-)methyltransferase from vaccinia virus. J. Biol. Chem. 250:9322–29
    [Google Scholar]
  18. 18. 
    Barbosa E, Moss B. 1978. mRNA(nucleoside-2′-)-methyltransferase from vaccinia virus. Purification and physical properties. J. Biol. Chem. 253:7692–97
    [Google Scholar]
  19. 19. 
    Barbosa E, Moss B. 1978. mRNA(nucleoside-2′-)-methyltransferase from vaccinia virus. Characteristics and substrate specificity. J. Biol. Chem. 253:7698–702
    [Google Scholar]
  20. 20. 
    Keith JM, Venkatesan S, Gershowitz A, Moss B 1982. Purification and characterization of the messenger ribonucleic acid capping enzyme GTP:RNA guanylyltransferase from wheat germ. Biochemistry 21:327–33
    [Google Scholar]
  21. 21. 
    Langberg SR, Moss B. 1981. Post-transcriptional modifications of mRNA. Purification and characterization of cap I and cap II RNA (nucleoside-2′-)-methyltransferases from HeLa cells. J. Biol. Chem. 256:10054–60
    [Google Scholar]
  22. 22. 
    Wittek R, Cooper J, Barbosa E, Moss B 1980. Expression of the vaccinia virus genome—analysis and mapping of mRNAs encoded within the inverted terminal repetition. Cell 21:487–93
    [Google Scholar]
  23. 23. 
    Wittek R, Moss B. 1980. Tandem repeats within the inverted terminal repetition of vaccinia virus DNA. Cell 21:277–84
    [Google Scholar]
  24. 24. 
    Venkatesan S, Baroudy BM, Moss B 1981. Distinctive nucleotide sequences adjacent to multiple initiation and termination sites of an early vaccinia virus gene. Cell 125:805–13
    [Google Scholar]
  25. 25. 
    Baroudy BM, Moss B. 1982. Sequence homologies of diverse length tandem repetitions near ends of vaccinia virus genome suggest unequal crossing over. Nucl. Acids Res. 10:5673–79
    [Google Scholar]
  26. 26. 
    Baroudy BM, Venkatesan S, Moss B 1982. Incompletely base-paired flip-flop terminal loops link the two DNA strands of the vaccinia virus genome into one uninterrupted polynucleotide chain. Cell 28:315–24
    [Google Scholar]
  27. 27. 
    Mackett M, Smith GL, Moss B 1982. Vaccinia virus: a selectable eukaryotic cloning and expression vector. PNAS 79:7415–19
    [Google Scholar]
  28. 28. 
    Smith GL, Mackett M, Moss B 1983. Infectious vaccinia virus recombinants that express hepatitis B antigen. Nature 302:490–95
    [Google Scholar]
  29. 29. 
    Mackett M, Smith GL, Moss B 1984. General method for production and selection of infectious vaccinia virus recombinants expressing foreign genes. J. Virol. 49:857–64
    [Google Scholar]
  30. 30. 
    Moss B, Smith GL, Gerin JL, Purcell RH 1984. Live recombinant vaccinia virus protects chimpanzees against hepatitis B. Nature 311:67–69
    [Google Scholar]
  31. 31. 
    Panicali D, Paoletti E. 1982. Construction of poxviruses as cloning vectors: insertion of the thymidine kinase gene from herpes simplex virus into the DNA of infectious vaccinia virus. PNAS 79:4927–31
    [Google Scholar]
  32. 32. 
    Moss B. 2013. Poxviridae. Fields Virology DM Knipe, PM Howley 2129–59 Philadelphia, PA: Wolters Kluwer/Lippincott Williams & Wilkins
    [Google Scholar]
  33. 33. 
    Yuen L, Moss B. 1987. Oligonucleotide sequence signaling transcriptional termination of vaccinia virus early genes. PNAS 84:6417–21
    [Google Scholar]
  34. 34. 
    Chakrabarti S, Brechling K, Moss B 1985. Vaccinia virus expression vector: Coexpression of b-galactosidase provides visual screening of recombinant virus plaques. Mol. Cell. Biol. 5:3403–9
    [Google Scholar]
  35. 35. 
    Davison AJ, Moss B. 1989. The structure of vaccinia virus early promoters. J. Mol. Biol. 210:749–69
    [Google Scholar]
  36. 36. 
    Davison AJ, Moss B. 1989. The structure of vaccinia virus late promoters. J. Mol. Biol. 210:771–84
    [Google Scholar]
  37. 37. 
    Baldick CJ, Keck JG, Moss B 1992. Mutational analysis of the core, spacer and initiator regions of vaccinia virus intermediate class promoters. J. Virol. 66:4710–19
    [Google Scholar]
  38. 38. 
    Weir JP, Bajszar G, Moss B 1982. Mapping of the vaccinia virus thymidine kinase gene by marker rescue and by cell-free translation of selected mRNA. PNAS 79:1210–14
    [Google Scholar]
  39. 39. 
    Jones EV, Puckett C, Moss B 1987. DNA-dependent RNA polymerase subunits encoded within the vaccinia virus genome. J. Virol. 61:1765–71
    [Google Scholar]
  40. 40. 
    Ahn B-Y, Jones EV, Moss B 1990. Identification of the vaccinia virus gene encoding an 18-kilodalton subunit of RNA polymerase and demonstration of a 5′ poly(A) leader on its early transcript. J. Virol. 64:3019–24
    [Google Scholar]
  41. 41. 
    Ahn B-Y, Gershon PD, Jones EV, Moss B 1990. Identification of rpo30, a vaccinia virus RNA polymerase gene with structural similarity to a eukaryotic transcription factor. Mol. Cell. Biol. 10:5433–41
    [Google Scholar]
  42. 42. 
    Amegadzie BY, Cole N, Ahn BY, Moss B 1991. Identification, sequence and expression of the gene encoding a Mr 35,000 subunit of the vaccinia virus DNA-dependent RNA polymerase. J. Biol. Chem. 266:13712–18
    [Google Scholar]
  43. 43. 
    Amegadzie B, Holmes M, Cole NB, Jones EV, Earl PL, Moss B 1991. Identification, sequence, and expression of the gene encoding the second-largest subunit of the vaccinia virus DNA polymerase. Virology 180:88–98
    [Google Scholar]
  44. 44. 
    Ahn B-Y, Rosel J, Cole NB, Moss B 1992. Identification and expression of rpo19, a vaccinia virus gene encoding a 19-kiloDalton DNA-dependent RNA polymerase subunit. J. Virol. 66:971–82
    [Google Scholar]
  45. 45. 
    Amegadzie BY, Ahn BY, Moss B 1992. Characterization of a 7-kilodalton subunit of vaccinia virus DNA-dependent RNA polymerase with structural similarities to the smallest subunit of eukaryotic RNA polymerase-II. J. Virol. 66:3003–10
    [Google Scholar]
  46. 46. 
    Broyles SS, Moss B. 1987. Identification of the vaccinia virus gene encoding nucleoside triphosphate phosphohydrolase I, a DNA-dependent ATPase. J. Virol. 61:1738–42
    [Google Scholar]
  47. 47. 
    Goebel SJ, Johnson GP, Perkus ME, Davis SW, Winslow JP, Paoletti E 1990. The complete DNA sequence of vaccinia virus. Virology 179:247–66
    [Google Scholar]
  48. 48. 
    Gershon PD, Moss B. 1990. Early transcription factor subunits are encoded by vaccinia virus late genes. PNAS 87:4401–5
    [Google Scholar]
  49. 49. 
    Ahn B-Y, Moss B. 1992. RNA polymerase-associated transcription specificity factor encoded by vaccinia virus. PNAS 89:3536–40
    [Google Scholar]
  50. 50. 
    Schnierle BS, Gershon PD, Moss B 1992. Cap-specific mRNA (nucleoside-O2′-)-methyltransferase and poly(A) polymerase stimulatory activities of vaccinia virus are mediated by a single protein. PNAS 89:2897–901
    [Google Scholar]
  51. 51. 
    Keck JG, Baldick CJ, Moss B 1990. Role of DNA replication in vaccinia virus gene expression: A naked template is required for transcription of three late transactivator genes. Cell 61:801–9
    [Google Scholar]
  52. 52. 
    Parrish S, Resch W, Moss B 2007. Vaccinia virus D10 protein has mRNA decapping activity, providing a mechanism for control of host and viral gene expression. PNAS 104:2139–44
    [Google Scholar]
  53. 53. 
    Liu SW, Katsafanas GC, Liu R, Wyatt LS, Moss B 2015. Poxvirus decapping enzymes enhance virulence by preventing the accumulation of dsRNA and the induction of innate antiviral responses. Cell Host Microbe 17:320–31
    [Google Scholar]
  54. 54. 
    Liu R, Moss B. 2016. Opposing roles of double-stranded RNA effector pathways and viral defense proteins revealed with CRISPR/Cas9 knock-out cell lines and vaccinia virus mutants. J. Virol. 90:7864–79
    [Google Scholar]
  55. 55. 
    Sanz P, Moss B. 1998. A new vaccinia virus intermediate transcription factor. J. Virol. 72:6880–83
    [Google Scholar]
  56. 56. 
    Sanz P, Moss B. 1999. Identification of a transcription factor, encoded by two vaccinia virus early genes, that regulates the intermediate stage of viral gene expression. PNAS 96:2692–97
    [Google Scholar]
  57. 57. 
    Yang Z, Bruno DP, Martens CA, Porcella SF, Moss B 2010. Simultaneous high-resolution analysis of vaccinia virus and host cell transcriptomes by deep RNA sequencing. PNAS 107:11513–18
    [Google Scholar]
  58. 58. 
    Yang Z, Reynolds SE, Martens CA, Bruno DP, Porcella SF, Moss B 2011. Expression profiling of the intermediate and late stages of poxvirus replication. J. Virol. 85:9899–908
    [Google Scholar]
  59. 59. 
    Yang Z, Cao S, Martens CA, Porcella SF, Xie Z et al. 2015. Deciphering poxvirus gene expression by RNA sequencing and ribosome profiling. J. Virol. 89:6874–86
    [Google Scholar]
  60. 60. 
    Buller RML, Chakrabarti S, Cooper JA, Twardzik DR, Moss B 1988. Deletion of the vaccinia virus growth factor gene reduces virus virulence. J. Virol. 62:866–74
    [Google Scholar]
  61. 61. 
    Holzer G, Falkner FG. 1997. Construction of a vaccinia virus deficient in the essential DNA repair enzyme uracil DNA glycosylase by a complementing cell line. J. Virol. 71:4997–5002
    [Google Scholar]
  62. 62. 
    Kato SEM, Moussatche N, D'Costa SM, Bainbridge TW, Prins C et al. 2008. Marker rescue mapping of the combined Condit/Dales collection of temperature-sensitive vaccinia virus mutants. Virology 375:213–22
    [Google Scholar]
  63. 63. 
    Zhang Y, Moss B. 1991. Inducer-dependent conditional-lethal mutant animal viruses. PNAS 88:1511–15
    [Google Scholar]
  64. 64. 
    Rodriguez JF, Smith GL. 1990. IPTG-dependent vaccinia virus: identification of a virus protein enabling virion envelopment by Golgi membrane and egress. Nucleic Acids Res 18:5347–51
    [Google Scholar]
  65. 65. 
    Fuerst TR, Fernandez MP, Moss B 1989. Transfer of the inducible lac repressor/operator system from Escherichia coli to a vaccinia virus expression vector. PNAS 86:2549–53
    [Google Scholar]
  66. 66. 
    Ward GA, Stover CK, Moss B, Fuerst TR 1995. Stringent chemical and thermal regulation of recombinant gene expression by vaccinia virus vectors in mammalian cells. PNAS 92:6773–77
    [Google Scholar]
  67. 67. 
    Wolffe EJ, Moore DM, Peters PJ, Moss B 1996. Vaccinia virus A17L open reading frame encodes an essential component of nascent viral membranes that is required to initiate morphogenesis. J. Virol. 70:2797–808
    [Google Scholar]
  68. 68. 
    Domi A, Moss B. 2002. Cloning the vaccinia virus genome as a bacterial artificial chromosome in Escherichia coli and recovery of infectious virus in mammalian cells. PNAS 99:12415–20
    [Google Scholar]
  69. 69. 
    Domi A, Moss B. 2005. Engineering of a vaccinia virus bacterial artificial chromosome in Escherichia coli by bacteriophage λ-based recombination. Nat. Methods 2:95–97
    [Google Scholar]
  70. 70. 
    Yuan M, Zhang WS, Wang J, Al Yaghchi C, Ahmed J et al. 2015. Efficiently editing the vaccinia virus genome by using the CRISPR-Cas9 system. J. Virol. 89:5176–79
    [Google Scholar]
  71. 71. 
    Cairns J. 1960. The initiation of vaccinia infection. Virology 11:603–23
    [Google Scholar]
  72. 72. 
    Katsafanas GC, Moss B. 2007. Colocalization of transcription and translation within cytoplasmic poxvirus factories coordinates viral expression and subjugates host functions. Cell Host Microbe 2:221–28
    [Google Scholar]
  73. 73. 
    Paszkowski P, Noyce RS, Evans DH 2016. Live-cell imaging of vaccinia virus recombination. PLOS Pathog 12:e1005824
    [Google Scholar]
  74. 74. 
    Jones EV, Moss B. 1984. Mapping of the vaccinia virus DNA polymerase gene by marker rescue and cell-free translation of selected mRNA. J. Virol. 49:72–77
    [Google Scholar]
  75. 75. 
    Earl PL, Jones EV, Moss B 1986. Homology between DNA polymerases of poxviruses, herpesviruses, and adenoviruses: nucleotide sequence of the vaccinia virus DNA polymerase gene. PNAS 83:3659–63
    [Google Scholar]
  76. 76. 
    Merchlinsky M, Moss B. 1986. Resolution of linear minichromosomes with hairpin ends from circular plasmids containing vaccinia virus concatemer junctions. Cell 45:879–84
    [Google Scholar]
  77. 77. 
    Delange AM, Reddy M, Scraba D, Upton C, McFadden G 1986. Replication and resolution of cloned poxvirus telomeres in vivo generates linear minichromsomes with intact viral hairpin termini. J. Virol. 59:249–59
    [Google Scholar]
  78. 78. 
    Garcia AD, Aravind L, Koonin EV, Moss B 2000. Bacterial-type DNA Holliday junction resolvases in eukaryotic viruses. PNAS 97:8926–31
    [Google Scholar]
  79. 79. 
    De Silva FS, Lewis W, Berglund P, Koonin EV, Moss B 2007. Poxvirus DNA primase. PNAS 104:18724–29
    [Google Scholar]
  80. 80. 
    Senkevich TG, Koonin EV, Moss B 2009. Predicted poxvirus FEN1-like nuclease required for homologous recombination, double-strand break repair and full-size genome formation. PNAS 106:17921–26
    [Google Scholar]
  81. 81. 
    Colinas RJ, Goebel SJ, Davis SW, Johnson GP, Norton EK, Paoletti E 1990. A DNA ligase gene in the Copenhagen strain of vaccinia virus is nonessential for viral replication and recombination. Virology 179:267–75
    [Google Scholar]
  82. 82. 
    Kerr SM, Smith GL. 1991. Vaccinia virus DNA ligase is nonessential for virus replication: recovery of plasmids from virus-infected cells. Virology 180:625–32
    [Google Scholar]
  83. 83. 
    Paran N, De Silva FS, Senkevich TG, Moss B 2009. Cellular DNA ligase I is recruited to cytoplasmic vaccinia virus factories and masks the role of the vaccinia ligase in viral DNA replication. Cell Host Microbe 6:563–69
    [Google Scholar]
  84. 84. 
    Senkevich TG, Katsafanas G, Weisberg A, Olano LR, Moss B 2017. Identification of vaccinia virus replisome and transcriptome proteins by iPOND coupled with mass spectrometry. J. Virol. 91:e01015-17
    [Google Scholar]
  85. 85. 
    Postigo A, Ramsden AE, Howell M, Way M 2017. Cytoplasmic ATR activation promotes vaccinia virus genome replication. Cell Rep 19:1022–32
    [Google Scholar]
  86. 86. 
    Senkevich TG, Ward BM, Moss B 2004. Vaccinia virus A28L gene encodes an essential protein component of the virion membrane with intramolecular disulfide bonds formed by the viral cytoplasmic redox pathway. J. Virol. 78:2348–56
    [Google Scholar]
  87. 87. 
    Senkevich TG, Ward BM, Moss B 2004. Vaccinia virus entry into cells is dependent on a virion surface protein encoded by the A28L gene. J. Virol. 78:2357–66
    [Google Scholar]
  88. 88. 
    Townsley A, Senkevich TG, Moss B 2005. Vaccinia virus A21 virion membrane protein is required for cell entry and fusion. J. Virol. 79:9458–69
    [Google Scholar]
  89. 89. 
    Townsley A, Senkevich TG, Moss B 2005. The product of the vaccinia virus L5R gene is a fourth membrane protein encoded by all poxviruses that is required for cell entry and cell-cell fusion. J. Virol. 79:10988–98
    [Google Scholar]
  90. 90. 
    Senkevich TG, Ojeda S, Townsley A, Nelson GE, Moss B 2005. Poxvirus multiprotein entry-fusion complex. PNAS 102:18572–77
    [Google Scholar]
  91. 91. 
    Ojeda S, Senkevich TG, Moss B 2006. Entry of vaccinia virus and cell-cell fusion require a highly conserved cysteine-rich membrane protein encoded by the A16L gene. J. Virol. 80:51–61
    [Google Scholar]
  92. 92. 
    Ojeda S, Domi A, Moss B 2006. Vaccinia virus G9 protein is an essential component of the poxvirus entry-fusion complex. J. Virol. 80:9822–30
    [Google Scholar]
  93. 93. 
    Bisht H, Weisberg AS, Moss B 2008. Vaccinia virus L1 protein is required for cell entry and membrane fusion. J. Virol. 82:8687–94
    [Google Scholar]
  94. 94. 
    Satheshkumar PS, Moss B. 2009. Characterization of a newly identified 35 amino acid component of the vaccinia virus entry/fusion complex conserved in all chordopoxviruses. J. Virol. 83:12822–32
    [Google Scholar]
  95. 95. 
    Moss B. 2012. Poxvirus cell entry: How many proteins does it take. ? Viruses 4:688–707
    [Google Scholar]
  96. 96. 
    Senkevich TG, White CL, Koonin EV, Moss B 2002. Complete pathway for protein disulfide bond formation encoded by poxviruses. PNAS 99:6667–72
    [Google Scholar]
  97. 97. 
    Laliberte JP, Weisberg AS, Moss B 2011. The membrane fusion step of vaccinia virus entry is cooperatively mediated by multiple viral proteins and host cell components. PLOS Pathog 7:e1002446
    [Google Scholar]
  98. 98. 
    Townsley AC, Weisberg AS, Wagenaar TR, Moss B 2006. Vaccinia virus entry into cells via a low pH-dependent endosomal pathway. J. Virol. 80:8899–908
    [Google Scholar]
  99. 99. 
    Su HP, Garman SC, Allison TJ, Fogg C, Moss B, Garboczi DN 2005. The 1.51-Å structure of the poxvirus L1 protein, a target of potent neutralizing antibodies. PNAS 102:4240–45
    [Google Scholar]
  100. 100. 
    Diesterbeck US, Gittis AG, Garboczi DN, Moss B 2018. The 2.1 Å structure of protein F9 and its comparison to L1, two components of the conserved poxvirus entry-fusion complex. Sci. Rep. 8:16807
    [Google Scholar]
  101. 101. 
    Wagenaar TR, Ojeda S, Moss B 2008. Vaccinia virus A56/K2 fusion regulatory protein interacts with the A16 and G9 subunits of the entry fusion complex. J. Virol. 82:5153–60
    [Google Scholar]
  102. 102. 
    Wagenaar TR, Moss B. 2009. Expression of the A56 and K2 proteins is sufficient to inhibit vaccinia virus entry and cell fusion. J. Virol. 83:1546–54
    [Google Scholar]
  103. 103. 
    Chang HW, Yang CH, Luo YC, Su BG, Cheng HY et al. 2019. Vaccinia viral A26 protein is a fusion suppressor of mature virus and triggers membrane fusion through conformational change at low pH. PLOS Pathog 15:e1007826
    [Google Scholar]
  104. 104. 
    Moss B. 2018. Origin of the poxviral membrane: a 50-year-old riddle. PLOS Pathog 14:e1007002
    [Google Scholar]
  105. 105. 
    Dales S, Mosbach EH. 1968. Vaccinia as a model for membrane biogenesis. Virology 35:564–83
    [Google Scholar]
  106. 106. 
    Maruri-Avidal L, Weisberg AS, Moss B 2013. Direct formation of vaccinia virus membranes from the endoplasmic reticulum in the absence of the newly characterized L2-interacting protein A30.5. J. Virol. 87:12313–26
    [Google Scholar]
  107. 107. 
    Resch W, Weisberg AS, Moss B 2005. Vaccinia virus nonstructural protein encoded by the A11R gene is required for formation of the virion membrane. J. Virol. 79:6598–609
    [Google Scholar]
  108. 108. 
    Maruri-Avidal L, Weisberg AS, Moss B 2013. Association of the vaccinia virus A11 protein with the endoplasmic reticulum and crescent precursors of immature virions. J. Virol. 87:10195–1206
    [Google Scholar]
  109. 109. 
    Weisberg AS, Maruri-Avidal L, Bisht H, Hansen BT, Schwartz CL et al. 2017. Enigmatic origin of the poxvirus membrane from the endoplasmic reticulum shown by 3D imaging of vaccinia virus assembly mutants. PNAS 114:E11001–11001
    [Google Scholar]
  110. 110. 
    Moss B, Rosenblum EN, Katz E, Grimley PM 1969. Rifampicin: a specific inhibitor of vaccinia virus assembly. Nature 224:1280–84
    [Google Scholar]
  111. 111. 
    Nagayama A, Pogo BGT, Dales S 1970. Biogenesis of vaccinia: separation of early stages from maturation by means of rifampicin. Virology 40:1039–51
    [Google Scholar]
  112. 112. 
    Tartaglia J, Piccini A, Paoletti E 1986. Vaccinia virus rifampicin-resistance locus specifies a late 63,000 Da gene product. Virology 150:45–54
    [Google Scholar]
  113. 113. 
    Baldick CJ, Moss B. 1987. Resistance of vaccinia virus to rifampicin conferred by a single nucleotide substitution near the predicted NH2 terminus of a gene encoding an Mr 62,000 polypeptide. Virology 156:138–45
    [Google Scholar]
  114. 114. 
    Szajner P, Weisberg AS, Lebowitz J, Heuser J, Moss B 2005. External scaffold of spherical immature poxvirus particles is made of protein trimers, forming a honeycomb lattice. J. Cell. Biol. 170:971–81
    [Google Scholar]
  115. 115. 
    Erlandson KJ, Bisht H, Weisberg AS, Hyun SI, Hansen BT et al. 2016. Poxviruses encode a reticulon-like protein that promotes membrane curvature. Cell Rep 14:2084–91
    [Google Scholar]
  116. 116. 
    Bisht H, Weisberg AS, Szajner P, Moss B 2009. Assembly and disassembly of the capsid-like external scaffold of immature virions during vaccinia virus morphogenesis. J. Virol. 83:9140–50
    [Google Scholar]
  117. 117. 
    Cassetti MC, Merchlinsky M, Wolffe EJ, Weisberg AS, Moss B 1998. DNA packaging mutant: Repression of the vaccinia virus A32 gene results in noninfectious, DNA-deficient, spherical, enveloped particles. J. Virol. 72:5769–80
    [Google Scholar]
  118. 118. 
    Grubisha O, Traktman P. 2003. Genetic analysis of the vaccinia virus I6 telomere-binding protein uncovers a key role in genome encapsidation. J. Virol. 77:10929–42
    [Google Scholar]
  119. 119. 
    Ahn B-Y, Gershon PD, Moss B 1994. RNA-polymerase associated protein RAP94 confers promoter specificity for initiating transcription of vaccinia virus early stage genes. J. Biol. Chem. 269:7552–57
    [Google Scholar]
  120. 120. 
    Zhang Y, Ahn B-Y, Moss B 1994. Targeting of a multicomponent transcription apparatus into assembling vaccinia virus particles requires RAP94, an RNA polymerase-associated protein. J. Virol. 68:1360–70
    [Google Scholar]
  121. 121. 
    Yang Z, Moss B. 2009. Interaction of the vaccinia virus RNA polymerase-associated 94-kilodalton protein with the early transcription factor. J. Virol. 83:12018–26
    [Google Scholar]
  122. 122. 
    Grimm C, Hillen HS, Bedenk K, Bartuli J, Neyer S et al. 2019. Structural basis of poxvirus transcription: vaccinia RNA polymerase complexes. Cell 179:1537–50
    [Google Scholar]
  123. 123. 
    Dobzhansky T. 1973. Nothing in biology makes sense except in the light of evolution. Am. Biol. Teacher 35:125–29
    [Google Scholar]
  124. 124. 
    Kotwal GJ, Moss B. 1988. Vaccinia virus encodes a secretory polypeptide structurally related to complement control proteins. Nature 335:176–78
    [Google Scholar]
  125. 125. 
    Kotwal GJ, Isaacs SN, Mckenzie R, Frank MM, Moss B 1990. Inhibition of the complement cascade by the major secretory protein of vaccinia virus. Science 250:827–30
    [Google Scholar]
  126. 126. 
    Twardzik DR, Brown JP, Ranchalis JE, Todaro GJ, Moss B 1985. Vaccinia virus-infected cells release a novel polypeptide functionally related to transforming and epidermal growth factors. PNAS 82:5300–4
    [Google Scholar]
  127. 127. 
    Sivan G, Martin SE, Myers TG, Buehler E, Szymczyk KH et al. 2013. Human genome-wide RNAi screen reveals a role for nuclear pore proteins in poxvirus morphogenesis. PNAS 110:3519–24
    [Google Scholar]
  128. 128. 
    Sivan G, Weisberg AS, Americo JL, Moss B 2016. Retrograde transport from early endosomes to the trans-Golgi network enables membrane wrapping and egress of vaccinia virions. J. Virol. 90:8891–905
    [Google Scholar]
  129. 129. 
    Sivan G, Ormanoglu P, Buehler EC, Martin SE, Moss B 2015. Identification of restriction factors by human genome-wide RNA interference screening of viral host range mutants exemplified by discovery of SAMD9 and WDR6 as inhibitors of the vaccinia virus K1LC7L mutant. mBio 6:e01122
    [Google Scholar]
  130. 130. 
    Sivan G, Glushakow-Smith SG, Katsafanas GC, Americo JL, Moss B 2018. Human host range restriction of the vaccinia virus C7/K1 double deletion mutant is mediated by an atypical mode of translation inhibition. J. Virol. 92:e01329-18
    [Google Scholar]
  131. 131. 
    Panda D, Fernandez DJ, Lal M, Buehler E, Moss B 2017. Triad of human cellular proteins, IRF2, FAM111A, and RFC3, restrict replication of orthopoxvirus SPI-1 host-range mutants. PNAS 114:3720–25
    [Google Scholar]
  132. 132. 
    Liu R, Moss B. 2018. Vaccinia virus C9 ankyrin repeat/F-box protein is a newly identified antagonist of the type I interferon-induced antiviral state. J. Virol. 92:e00053-18
    [Google Scholar]
  133. 133. 
    Liu R, Olano LR, Mirzakhanyan Y, Gershon PD, Moss B 2019. Vaccinia virus ankyrin-repeat/F-box protein targets interferon-induced IFITs for proteasomal degradation. Cell Rep 29:816–28
    [Google Scholar]
  134. 134. 
    Shchelkunov SN, Totmenin AV, Safronov PF, Mikheev MV, Gutorov VV et al. 2002. Analysis of the monkeypox virus genome. Virology 297:172–94
    [Google Scholar]
  135. 135. 
    Earl PL, Americo JL, Wyatt LS, Espenshade O, Bassler J et al. 2008. Rapid protection in a monkeypox model by a single injection of a replication-deficient vaccinia virus. PNAS 105:10889–94
    [Google Scholar]
  136. 136. 
    Earl PL, Americo JL, Moss B 2017. Insufficient innate immunity contributes to the susceptibility of the castaneous mouse to orthopoxvirus infection. J. Virol. 91:e01042-17
    [Google Scholar]
  137. 137. 
    Earl PL, Americo JL, Moss B 2020. Natural killer cells expanded in vivo or ex vivo with IL-15 overcomes the inherent susceptibility of CAST mice to lethal infection with orthopoxviruses. PLOS Pathog 16:4e1008505
    [Google Scholar]
/content/journals/10.1146/annurev-virology-021020-100558
Loading
/content/journals/10.1146/annurev-virology-021020-100558
Loading

Data & Media loading...

  • Article Type: Review Article
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