1932

Abstract

My great-grandparents were immigrants from Sweden and settled as farmers in Iowa and Illinois. My father, the oldest of six children, was the first in his family to go to college and had careers as a petroleum geologist and an academic. My mother, the youngest of four children, had older siblings in education, and she focused on early childhood education. My childhood in Oklahoma with two younger sisters was happy and comfortable, and public school prepared me well. My career trajectory into virology did not involve much if any advance planning but was characterized by recognizing the fascinating puzzles of virus diseases, being in good places at the right time, taking advantage of opportunities as they presented themselves, and being surrounded by great mentors, colleagues, trainees, and family. Most of my career was spent studying two diseases caused by RNA viruses, alphavirus encephalomyelitis and measles, and was enriched with several leadership opportunities.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-virology-012420-024942
2020-09-29
2024-04-28
Loading full text...

Full text loading...

/deliver/fulltext/virology/7/1/annurev-virology-012420-024942.html?itemId=/content/journals/10.1146/annurev-virology-012420-024942&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Taylor RM, Hurlbut HS, Work TH, Kingston JR, Frothingham TE 1955. Sindbis virus: a newly recognized arthropod-transmitted virus. Am. J. Trop. Med. Hyg. 4:5844–62
    [Google Scholar]
  2. 2. 
    McFarland HF, Griffin DE, Johnson RT 1972. Specificity of the inflammatory response in viral encephalitis. I. Adoptive immunization of immunosuppressed mice infected with Sindbis virus. J. Exp. Med. 136:2216–26
    [Google Scholar]
  3. 3. 
    Johnson RT, McFarland HF, Levy SE 1972. Age-dependent resistance to viral encephalitis: studies of infections due to Sindbis virus in mice. J. Infect. Dis. 125:3257–62
    [Google Scholar]
  4. 4. 
    Griffin DE. 1976. Role of the immune response in age-dependent resistance of mice to encephalitis due to Sindbis virus. J. Infect Dis. 133:4456–64
    [Google Scholar]
  5. 5. 
    Vernon PS, Griffin DE. 2005. Characterization of an in vitro model of alphavirus infection of immature and mature neurons. J. Virol. 79:63438–47
    [Google Scholar]
  6. 6. 
    Schultz KL, Vernon PS, Griffin DE 2015. Differentiation of neurons restricts Arbovirus replication and increases expression of the alpha isoform of IRF-7. J. Virol. 89:148–60
    [Google Scholar]
  7. 7. 
    Yeh JX, Park E, Schultz KLW, Griffin DE 2019. NF-κB activation promotes alphavirus replication in mature neurons. J. Virol. 93:24e01071-19
    [Google Scholar]
  8. 8. 
    Griffin DE, Johnson RT. 1977. Role of the immune response in recovery from Sindbis virus encephalitis in mice. J. Immunol. 118:31070–75
    [Google Scholar]
  9. 9. 
    Jackson AC, Moench TR, Trapp BD, Griffin DE 1988. Basis of neurovirulence in Sindbis virus encephalomyelitis of mice. Lab. Invest. 58:5503–9
    [Google Scholar]
  10. 10. 
    Strauss EG, Rice CM, Strauss JH 1984. Complete nucleotide sequence of the genomic RNA of Sindbis virus. Virology 133:192–110
    [Google Scholar]
  11. 11. 
    Rice CM, Levis R, Strauss JH, Huang HV 1987. Production of infectious RNA transcripts from Sindbis virus cDNA clones: mapping of lethal mutations, rescue of a temperature-sensitive marker, and in vitro mutagenesis to generate defined mutants. J. Virol. 61:123809–19
    [Google Scholar]
  12. 12. 
    Lustig S, Jackson AC, Hahn CS, Griffin DE, Strauss EG, Strauss JH 1988. Molecular basis of Sindbis virus neurovirulence in mice. J. Virol. 62:72329–36
    [Google Scholar]
  13. 13. 
    Park E, Griffin DE. 2009. The nsP3 macro domain is important for Sindbis virus replication in neurons and neurovirulence in mice. Virology 388:2305–14
    [Google Scholar]
  14. 14. 
    Tucker PC, Lee SH, Bui N, Martinie D, Griffin DE 1997. Amino acid changes in the Sindbis virus E2 glycoprotein that increase neurovirulence improve entry into neuroblastoma cells. J. Virol. 71:86106–12
    [Google Scholar]
  15. 15. 
    Tucker PC, Griffin DE. 1991. Mechanism of altered Sindbis virus neurovirulence associated with a single-amino-acid change in the E2 glycoprotein. J. Virol. 65:31551–57
    [Google Scholar]
  16. 16. 
    Tucker PC, Strauss EG, Kuhn RJ, Strauss JH, Griffin DE 1993. Viral determinants of age-dependent virulence of Sindbis virus for mice. J. Virol. 67:84605–10
    [Google Scholar]
  17. 17. 
    Levine B, Griffin DE. 1993. Molecular analysis of neurovirulent strains of Sindbis virus that evolve during persistent infection of SCID mice. J. Virol. 67:116872–75
    [Google Scholar]
  18. 18. 
    Voss JE, Vaney MC, Duquerroy S, Vonrhein C, Girard-Blanc C et al. 2010. Glycoprotein organization of Chikungunya virus particles revealed by X-ray crystallography. Nature 468:7324709–12
    [Google Scholar]
  19. 19. 
    Gorbalenya AE, Koonin EV, Lai MM 1991. Putative papain-related thiol proteases of positive-strand RNA viruses. Identification of rubi- and aphthovirus proteases and delineation of a novel conserved domain associated with proteases of rubi-, alpha- and coronaviruses. FEBS Lett 288:1–2201–5
    [Google Scholar]
  20. 20. 
    McPherson RL, Abraham R, Sreekumar E, Ong SE, Cheng SJ et al. 2017. ADP-ribosylhydrolase activity of Chikungunya virus macrodomain is critical for virus replication and virulence. PNAS 114:71666–71
    [Google Scholar]
  21. 21. 
    Abraham R, Hauer D, McPherson RL, Utt A, Kirby IT et al. 2018. ADP-ribosyl-binding and hydrolase activities of the alphavirus nsP3 macrodomain are critical for initiation of virus replication. PNAS 115:44E10457–10457
    [Google Scholar]
  22. 22. 
    Levine B, Huang Q, Isaacs JT, Reed JC, Griffin DE, Hardwick JM 1993. Conversion of lytic to persistent alphavirus infection by the bcl-2 cellular oncogene. Nature 361:6414739–42
    [Google Scholar]
  23. 23. 
    Ubol S, Tucker PC, Griffin DE, Hardwick JM 1994. Neurovirulent strains of Alphavirus induce apoptosis in bcl-2-expressing cells: role of a single amino acid change in the E2 glycoprotein. PNAS 91:115202–6
    [Google Scholar]
  24. 24. 
    Levine B, Hardwick JM, Trapp BD, Crawford TO, Bollinger RC, Griffin DE 1991. Antibody-mediated clearance of alphavirus infection from neurons. Science 254:5033856–60
    [Google Scholar]
  25. 25. 
    Ubol S, Levine B, Lee SH, Greenspan NS, Griffin DE 1995. Roles of immunoglobulin valency and the heavy-chain constant domain in antibody-mediated downregulation of Sindbis virus replication in persistently infected neurons. J. Virol. 69:31990–93
    [Google Scholar]
  26. 26. 
    Despres P, Griffin JW, Griffin DE 1995. Effects of anti-E2 monoclonal antibody on Sindbis virus replication in AT3 cells expressing bcl-2. J. Virol. 69:117006–14
    [Google Scholar]
  27. 27. 
    Nilaratanakul V, Chen J, Tran O, Baxter VK, Troisi EM et al. 2018. Germ line IgM is sufficient, but not required, for antibody-mediated alphavirus clearance from the central nervous system. J. Virol. 92:7e02081-17
    [Google Scholar]
  28. 28. 
    Baxter VK, Troisi EM, Pate NM, Zhao JN, Griffin DE 2018. Death and gastrointestinal bleeding complicate encephalomyelitis in mice with delayed appearance of CNS IgM after intranasal alphavirus infection. J. Gen. Virol. 99:309–20
    [Google Scholar]
  29. 29. 
    Binder GK, Griffin DE. 2001. Interferon-γ-mediated site-specific clearance of alphavirus from CNS neurons. Science 293:5528303–6
    [Google Scholar]
  30. 30. 
    Burdeinick-Kerr R, Govindarajan D, Griffin DE 2009. Noncytolytic clearance of Sindbis virus infection from neurons by gamma interferon is dependent on Jak/STAT signaling. J. Virol. 83:83429–35
    [Google Scholar]
  31. 31. 
    Burdeinick-Kerr R, Griffin DE. 2005. Gamma interferon-dependent, noncytolytic clearance of Sindbis virus infection from neurons in vitro. J. Virol. 79:95374–85
    [Google Scholar]
  32. 32. 
    Burdeinick-Kerr R, Wind J, Griffin DE 2007. Synergistic roles of antibody and interferon in noncytolytic clearance of Sindbis virus from different regions of the central nervous system. J. Virol. 81:115628–36
    [Google Scholar]
  33. 33. 
    Levine B, Griffin DE. 1992. Persistence of viral RNA in mouse brains after recovery from acute alphavirus encephalitis. J. Virol. 66:116429–35
    [Google Scholar]
  34. 34. 
    Tyor WR, Wesselingh S, Levine B, Griffin DE 1992. Long term intraparenchymal Ig secretion after acute viral encephalitis in mice. J. Immunol. 149:124016–20
    [Google Scholar]
  35. 35. 
    Metcalf TU, Griffin DE. 2011. Alphavirus-induced encephalomyelitis: antibody-secreting cells and viral clearance from the nervous system. J. Virol. 85:2111490–501
    [Google Scholar]
  36. 36. 
    Metcalf TU, Baxter VK, Nilaratanakul V, Griffin DE 2013. Recruitment and retention of B cells in the central nervous system in response to alphavirus encephalomyelitis. J. Virol. 87:52420–29
    [Google Scholar]
  37. 37. 
    Baxter VK, Griffin DE. 2016. Interferon gamma modulation of disease manifestation and the local antibody response to alphavirus encephalomyelitis. J. Gen. Virol. 97:112908–25
    [Google Scholar]
  38. 38. 
    Camenga DL, Nathanson N. 1975. An immunopathologic component in experimental togavirus encephalitis. J. Neuropathol. Exp. Neurol. 34:6492–500
    [Google Scholar]
  39. 39. 
    Kulcsar KA, Baxter VK, Greene IP, Griffin DE 2014. Interleukin 10 modulation of pathogenic Th17 cells during fatal alphavirus encephalomyelitis. PNAS 111:4516053–58
    [Google Scholar]
  40. 40. 
    Kulcsar KA, Baxter VK, Abraham R, Nelson A, Griffin DE 2015. Distinct immune responses in resistant and susceptible strains of mice during neurovirulent alphavirus encephalomyelitis. J. Virol. 89:168280–91
    [Google Scholar]
  41. 41. 
    Griffin DE, Ward BJ, Jauregui E, Johnson RT, Vaisberg A 1989. Immune activation in measles. N. Engl. J. Med. 320:251667–72
    [Google Scholar]
  42. 42. 
    Ward BJ, Johnson RT, Vaisberg A, Jauregui E, Griffin DE 1990. Spontaneous proliferation of peripheral mononuclear cells in natural measles virus infection: identification of dividing cells and correlation with mitogen responsiveness. Clin. Immunol. Immunopathol. 55:2315–26
    [Google Scholar]
  43. 43. 
    Griffin DE, Ward BJ, Jauregui E, Johnson RT, Vaisberg A 1990. Immune activation during measles: interferon-gamma and neopterin in plasma and cerebrospinal fluid in complicated and uncomplicated disease. J. Infect. Dis. 161:3449–53
    [Google Scholar]
  44. 44. 
    Griffin DE, Ward BJ, Juaregui E, Johnson RT, Vaisberg A 1992. Immune activation during measles: β2-microglobulin in plasma and cerebrospinal fluid in complicated and uncomplicated disease. J. Infect. Dis. 166:51170–73
    [Google Scholar]
  45. 45. 
    Esolen LM, Takahashi K, Johnson RT, Vaisberg A, Moench TR et al. 1995. Brain endothelial cell infection in children with acute fatal measles. J. Clin. Invest. 96:52478–81
    [Google Scholar]
  46. 46. 
    Moench TR, Griffin DE, Obriecht CR, Vaisberg AJ, Johnson RT 1988. Acute measles in patients with and without neurological involvement: distribution of measles virus antigen and RNA. J. Infect. Dis. 158:2433–42
    [Google Scholar]
  47. 47. 
    Gendelman HE, Wolinsky JS, Johnson RT, Pressman NJ, Pezeshkpour GH, Boisset GF 1984. Measles encephalomyelitis: lack of evidence of viral invasion of the central nervous system and quantitative study of the nature of demyelination. Ann. Neurol. 15:4353–60
    [Google Scholar]
  48. 48. 
    Johnson RT, Griffin DE, Hirsch RL, Wolinsky JS, Roedenbeck S et al. 1984. Measles encephalomyelitis—clinical and immunologic studies. N. Engl. J. Med. 310:3137–41
    [Google Scholar]
  49. 49. 
    Auwaerter PG, Rota PA, Elkins WR, Adams RJ, DeLozier T et al. 1999. Measles virus infection in rhesus macaques: altered immune responses and comparison of the virulence of six different virus strains. J. Infect. Dis. 180:4950–58
    [Google Scholar]
  50. 50. 
    Polack FP, Auwaerter PG, Lee SH, Nousari HC, Valsamakis A et al. 1999. Production of atypical measles in rhesus macaques: evidence for disease mediated by immune complex formation and eosinophils in the presence of fusion-inhibiting antibody. Nat. Med. 5:6629–34
    [Google Scholar]
  51. 51. 
    Polack FP, Hoffman SJ, Crujeiras G, Griffin DE 2003. A role for nonprotective complement-fixing antibodies with low avidity for measles virus in atypical measles. Nat. Med. 9:91209–13
    [Google Scholar]
  52. 52. 
    Polack FP, Lee SH, Permar S, Manyara E, Nousari HG et al. 2000. Successful DNA immunization against measles: neutralizing antibody against either the hemagglutinin or fusion glycoprotein protects rhesus macaques without evidence of atypical measles. Nat. Med. 6:7776–81
    [Google Scholar]
  53. 53. 
    Pan CH, Valsamakis A, Colella T, Nair N, Adams RJ et al. 2005. Modulation of disease, T cell responses, and measles virus clearance in monkeys vaccinated with H-encoding alphavirus replicon particles. PNAS 102:3311581–88
    [Google Scholar]
  54. 54. 
    Pan CH, Nair N, Adams RJ, Zink MC, Lee EY et al. 2008. Dose-dependent protection against or exacerbation of disease by a polylactide glycolide microparticle-adsorbed, alphavirus-based measles virus DNA vaccine in rhesus macaques. Clin. Vaccine Immunol. 15:4697–706
    [Google Scholar]
  55. 55. 
    Pan CH, Jimenez GS, Nair N, Wei Q, Adams RJ et al. 2008. Use of Vaxfectin adjuvant with DNA vaccine encoding the measles virus hemagglutinin and fusion proteins protects juvenile and infant rhesus macaques against measles virus. Clin. Vaccine Immunol. 15:81214–21
    [Google Scholar]
  56. 56. 
    Pan CH, Greer CE, Hauer D, Legg HS, Lee EY et al. 2010. A chimeric alphavirus replicon particle vaccine expressing the hemagglutinin and fusion proteins protects juvenile and infant rhesus macaques from measles. J. Virol. 84:83798–807
    [Google Scholar]
  57. 57. 
    Lin WH, Vilalta A, Adams RJ, Rolland A, Sullivan SM, Griffin DE 2013. Vaxfectin adjuvant improves antibody responses of juvenile rhesus macaques to a DNA vaccine encoding the measles virus hemagglutinin and fusion proteins. J. Virol. 87:126560–68
    [Google Scholar]
  58. 58. 
    Lin WH, Griffin DE, Rota PA, Papania M, Cape SP et al. 2011. Successful respiratory immunization with dry powder live-attenuated measles virus vaccine in rhesus macaques. PNAS 108:72987–92
    [Google Scholar]
  59. 59. 
    Lin WH, Pan CH, Adams RJ, Laube BL, Griffin DE 2014. Vaccine-induced measles virus-specific T cells do not prevent infection or disease but facilitate subsequent clearance of viral RNA. mBio 5:2e01047
    [Google Scholar]
  60. 60. 
    Moss WJ, Ryon JJ, Monze M, Griffin DE 2002. Differential regulation of interleukin (IL)-4, IL-5, and IL-10 during measles in Zambian children. J. Infect. Dis. 186:7879–87
    [Google Scholar]
  61. 61. 
    Moss WJ, Monze M, Ryon JJ, Quinn TC, Griffin DE, Cutts F 2002. Prospective study of measles in hospitalized, human immunodeficiency virus (HIV)-infected and HIV-uninfected children in Zambia. Clin. Infect. Dis. 35:2189–96
    [Google Scholar]
  62. 62. 
    Moss WJ, Fisher C, Scott S, Monze M, Ryon JJ et al. 2008. HIV type 1 infection is a risk factor for mortality in hospitalized Zambian children with measles. Clin. Infect. Dis. 46:4523–27
    [Google Scholar]
  63. 63. 
    Nair N, Moss WJ, Scott S, Mugala N, Ndhlovu ZM et al. 2009. HIV-1 infection in Zambian children impairs the development and avidity maturation of measles virus-specific immunoglobulin G after vaccination and infection. J. Infect. Dis. 200:71031–38
    [Google Scholar]
  64. 64. 
    Moss WJ, Ryon JJ, Monze M, Cutts F, Quinn TC, Griffin DE 2002. Suppression of human immunodeficiency virus replication during acute measles. J. Infect. Dis. 185:81035–42
    [Google Scholar]
  65. 65. 
    Permar SR, Moss WJ, Ryon JJ, Monze M, Cutts F et al. 2001. Prolonged measles virus shedding in human immunodeficiency virus-infected children, detected by reverse transcriptase-polymerase chain reaction. J. Infect. Dis. 183:4532–38
    [Google Scholar]
  66. 66. 
    Riddell MA, Moss WJ, Hauer D, Monze M, Griffin DE 2007. Slow clearance of measles virus RNA after acute infection. J. Clin. Virol. 39:4312–17
    [Google Scholar]
  67. 67. 
    Lin WH, Kouyos RD, Adams RJ, Grenfell BT, Griffin DE 2012. Prolonged persistence of measles virus RNA is characteristic of primary infection dynamics. PNAS 109:3714989–94
    [Google Scholar]
  68. 68. 
    Nelson AN, Putnam N, Hauer D, Baxter VK, Adams RJ, Griffin DE 2017. Evolution of T cell responses during measles virus infection and RNA clearance. Sci. Rep. 7:111474
    [Google Scholar]
  69. 69. 
    Nelson AN, Lin WW, Shivakoti R, Putnam NE, Mangus LM et al. 2020. Association of persistent wild-type measles virus RNA with long-term humoral immunity in rhesus macaques. JCI Insight 5:3e134992
    [Google Scholar]
/content/journals/10.1146/annurev-virology-012420-024942
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