1932

Abstract

This odyssey is mine from early junior high school, where my dreams for adventure were shaped by Arthur Conan Doyle's Sherlock Holmes, Percival Christopher Wren's , and best of all the remarkable explorers in Paul de Kruif's . My birth site was in Manhattan (my mother was a Vogue model and my father worked in retail), and I traveled to college at the University of Alabama, Tuscaloosa, where my love of history and English literature was shaped along with a sufficient exposure to biology, chemistry, and genetics to meet requirements for entering medical school. By the second year at the University of Maryland School of Medicine, through expert teachers such as Theodore (Ted) Woodward and Sheldon (Shelly) Greisman in medicine and Charles Weissmann in virology and microbiology, I found that understanding why and how people became ill was more my cup of tea than identifying and treating their illnesses. Although I was becoming competent in diagnosis and treatment, I left medical school at the end of my sophomore year to seek a more basic understanding of biology and chemistry. I achieved this by working toward a PhD in biochemistry at Johns Hopkins McCollum-Pratt Institute combined with study of rickettsial toxin at Maryland. This was a very important time in my life, because it convinced me that addressing biologic and medical questions in a disciplined scientific manner was what my life voyage should be. That voyage led me initially, through Woodward's contact, to work a summer in Joe Smadel's unit at Walter Reed (Smadel being one of the deans of American virology) and to meet several times with Carleton Gajdusek and then John Enders at Harvard, who pointed me to Frank Dixon at Scripps in La Jolla, California, for postdoctoral training. Dixon was among the founders of modern immunology and a pathfinder for immunopathology. Training by and association with Dixon and his other postdoctoral fellows, my independent position at Scripps, early polishing by Karl Habel (a superb senior virologist who left the National Institutes of Health and came to Scripps), and the gifted postdoctoral fellows who joined my laboratory over four decades form the log of my scientific voyage. The strong friendships and collaborations developed with other young but growing experimentalists like Bernie Fields and Abner Notkins are the fabric of the tale I will weave and were pivotal in the establishment of viral pathogenesis as a discipline.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pathol-012615-044107
2016-05-23
2024-04-24
Loading full text...

Full text loading...

/deliver/fulltext/pathol/11/1/annurev-pathol-012615-044107.html?itemId=/content/journals/10.1146/annurev-pathol-012615-044107&mimeType=html&fmt=ahah

Literature Cited

  1. Ivanovski DI. 1.  1899. Ueber die Mosaikkrankheit der Tabakspflanze. Zent. Bakteriol. Parasitenkd. Infekt. 2:250–54 [Google Scholar]
  2. Beijerinck MW. 2.  1899. Bemerkung zu dem Aufsatz von Herrn Iwanowsky über die Mosaikkrankheit der Tabakspflanze. Zent. Bakteriol. Parasitenkd. Infekt. 1:310–11 [Google Scholar]
  3. Loeffler F, Frosch P. 3.  1898. Berichte der Kommission zur Erforschung der Maul und Klauenseuche bei dem Institut für Infektionskrankheiten in Berlin. Zent. Bakteriol. Parasitenkd. Infekt. 1:371–91 [Google Scholar]
  4. Zeliadt N. 4.  2013. Profile of Michael B.A. Oldstone. PNAS 110:4155–57 [Google Scholar]
  5. Oldstone MBA. 5.  1959. Altered reactivity to Escherichia coli endotoxin of mice subjected to sublethal tourniquet treatment. Proc. Soc. Exp. Biol. Med. 102:256–58 [Google Scholar]
  6. Oldstone MBA. 6.  2009. Frank James Dixon (1920–2008): A Biographical Memoir Washington, DC: Natl. Acad. Sci. [Google Scholar]
  7. Hotchin J. 7.  1962. The biology of lymphocytic choriomeningitis infection: virus-induced immune disease. Cold Spring Harb. Symp. Quant. Biol. 27:479–99 [Google Scholar]
  8. Rowe WP. 8.  1954. Studies on pathogenesis and immunity in lymphocytic choriomeningitis infection of the mouse. Res. Rep. Nav. Med. Res. Inst. 12:167–220 [Google Scholar]
  9. Zinkernagel RM. 9.  2002. Lymphocytic choriomeningitis virus and immunology. Curr. Top. Microbiol. Immunol. 263:1–6 [Google Scholar]
  10. Oldstone MBA. 10.  2002. Biology and pathogenesis of lymphocytic choriomeningitis virus infection. Curr. Top. Microbiol. Immunol. 263:83–117 [Google Scholar]
  11. Riviere Y, Southern PJ, Ahmed R, Oldstone MBA. 11.  1986. Biology of cloned cytotoxic T lymphocytes specific for lymphocytic choriomeningitis virus. V. Recognition is restricted to gene products encoded by the viral S RNA segment. J. Immunol. 136:304–7 [Google Scholar]
  12. Whitton JL, Southern PJ, Oldstone MBA. 12.  1988. Analyses of the cytotoxic T lymphocyte responses to glycoprotein and nucleoprotein components of lymphocytic choriomeningitis virus. Virology 162:321–27 [Google Scholar]
  13. Oldstone MBA, Whitton JL, Lewicki H, Tishon A. 13.  1988. Fine dissection of a nine amino acid glycoprotein epitope, a major determinant recognized by lymphocytic choriomeningitis virus-specific class I-restricted H-2Db cytotoxic T lymphocytes. J. Exp. Med. 168:559–70 [Google Scholar]
  14. Klavinskis LS, Whitton JL, Joly E, Oldstone MBA. 14.  1990. Vaccination and protection from a lethal viral infection: identification, incorporation, and use of a cytotoxic T lymphocyte glycoprotein epitope. Virology 178:393–400 [Google Scholar]
  15. Whitton JL, Tishon A, Lewicki H, Gebhard J, Cook T. 15.  et al. 1989. Molecular analyses of a five-amino-acid cytotoxic T-lymphocyte (CTL) epitope: an immunodominant region which induces nonreciprocal CTL cross-reactivity. J. Virol. 63:4303–10 [Google Scholar]
  16. Anderson J, Byrne JA, Schreiber R, Patterson S, Oldstone MBA. 16.  1985. Biology of cloned cytotoxic T lymphocytes specific for lymphocytic choriomeningitis virus: clearance of virus and in vitro properties. J. Virol. 53:552–60 [Google Scholar]
  17. Lewicki H, Tishon A, Borrow P, Evans CF, Gairin JE. 17.  et al. 1995. CTL escape viral variants. I. Generation and molecular characterization. Virology 210:29–40 [Google Scholar]
  18. Lewicki HA, von Herrath MG, Evans CF, Whitton JL, Oldstone MBA. 18.  1995. CTL escape viral variants. II. Biologic activity in vivo. Virology 211:443–50 [Google Scholar]
  19. Emonet SF, Garidou L, McGavern DB, de la Torre JC. 19.  2009. Generation of recombinant lymphocytic choriomeningitis viruses with trisegmented genomes stably expressing two additional genes of interest. PNAS 106:3473–78 [Google Scholar]
  20. Sullivan BM, Emonet S, Welch MJ, Lee AM, Campbell KP. 20.  et al. 2011. Point mutation in the glycoprotein of lymphocytic choriomeningitis virus is necessary for receptor binding, dendritic cell infection, and long-term persistence. PNAS 108:2969–74 [Google Scholar]
  21. Popkin DL, Teijaro JR, Lee AM, Lewicki H, Emonet S. 21.  et al. 2011. Expanded potential for recombinant trisegmented lymphocytic choriomeningitis viruses: protein production, antibody production, and in vivo assessment of biological function of genes of interest. J. Virol. 85:7928–32 [Google Scholar]
  22. Ahmed R, Salmi A, Butler LD, Chiller JM, Oldstone MBA. 22.  1984. Selection of genetic variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice: role in suppression of cytotoxic T lymphocyte response and viral persistence. J. Exp. Med. 160:521–40 [Google Scholar]
  23. Ahmed R, Oldstone MBA. 23.  1988. Organ-specific selection of viral variants during chronic infection. J. Exp. Med. 167:1719–24 [Google Scholar]
  24. Sevilla N, Kunz S, Holz A, Lewicki H, Homann D. 24.  et al. 2000. Immunosuppression and resultant viral persistence by specific viral targeting of dendritic cells. J. Exp. Med. 192:1249–60 [Google Scholar]
  25. Salvato M, Borrow P, Shimomaye E, Oldstone MBA. 25.  1991. Molecular basis of viral persistence: a single amino acid change in the glycoprotein of lymphocytic choriomeningitis virus is associated with suppression of the antiviral cytotoxic T-lymphocyte response and establishment of persistence. J. Virol. 65:1863–69 [Google Scholar]
  26. Oldstone MBA, Campbell KP. 26.  2011. Decoding arenavirus pathogenesis: essential roles for alpha-dystroglycan-virus interactions and the immune response. Virology 411:170–79 [Google Scholar]
  27. Bergthaler A, Flatz L, Hegazy AN, Johnson S, Horvath E. 27.  et al. 2010. Viral replicative capacity is the primary determinant of lymphocytic choriomeningitis virus persistence and immunosuppression. PNAS 107:21641–46 [Google Scholar]
  28. Lee AM, Cruite J, Welch MJ, Sullivan B, Oldstone MBA. 28.  2013. Pathogenesis of Lassa fever virus infection. I. Susceptibility of mice to recombinant Lassa Gp/LCMV chimeric virus. Virology 442:114–21 [Google Scholar]
  29. Teijaro JR, Ng C, Lee AM, Sullivan BM, Sheehan KC. 29.  et al. 2013. Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 340:207–11 [Google Scholar]
  30. Wilson EB, Yamada DH, Elsaesser H, Herskovitz J, Deng J. 30.  et al. 2013. Blockade of chronic type I interferon signaling to control persistent LCMV infection. Science 340:202–7 [Google Scholar]
  31. Wherry EJ. 31.  2011. T cell exhaustion. Nat. Immunol. 12:492–99 [Google Scholar]
  32. Brooks DG, Trifilo MJ, Edelmann KH, Teyton L, McGavern DB, Oldstone MBA. 32.  2006. Interleukin-10 determines viral clearance or persistence in vivo. Nat. Med. 12:1301–9 [Google Scholar]
  33. Ejrnaes M, Filippi CM, Martinic MM, Ling EM, Togher LM. 33.  et al. 2006. Resolution of a chronic viral infection after interleukin-10 receptor blockade. J. Exp. Med. 203:2461–72 [Google Scholar]
  34. Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJ, Suresh M. 34.  et al. 1998. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188:2205–13 [Google Scholar]
  35. Oldstone MBA. 35.  1994. The role of cytotoxic T lymphocytes in infectious disease: history, criteria and state of the art. Curr. Top. Microbiol. Immunol. 189:1–8 [Google Scholar]
  36. Zinkernagel RM, Doherty PC. 36.  1974. Restriction of in vitro T cell–mediated cytotoxicity in lymphocytic choriomeningitis within a syngeneic or semi-allogeneic system. Nature 248:701–2 [Google Scholar]
  37. Burnet FM, Fenner F. 37.  1949. The Production of Antibodies New York: MacMillan
  38. Oldstone MBA, Dixon FJ. 38.  1967. Lymphocytic choriomeningitis: production of anti-LCM antibody by ‘tolerant’ LCM-infected mice. Science 158:1193–95 [Google Scholar]
  39. Oldstone MBA, Dixon FJ. 39.  1969. Pathogenesis of chronic disease associated with persistent lymphocytic choriomeningitis viral infection. I. Relationship of antibody production to disease in neonatally infected mice. J. Exp. Med. 129:483–505 [Google Scholar]
  40. Oldstone MBA, Dixon FJ. 40.  1970. Pathogenesis of chronic disease associated with persistent lymphocytic choriomeningitis viral infection. II. Relationship of the anti-lymphocytic choriomeningitis immune response to tissue injury in chronic lymphocytic choriomeningitis disease. J. Exp. Med. 131:1–19 [Google Scholar]
  41. Lampert PW, Oldstone MBA. 41.  1973. Host immunoglobulin G and complement deposits in the choroid plexus during spontaneous immune complex disease. Science 180:408–10 [Google Scholar]
  42. Oldstone MBA. 42.  1975. Virus neutralization and virus-induced immune complex disease: virus-antibody union resulting in immunoprotection or immunologic injury—two sides of the same coin. Prog. Med. Virol. 19:84–119 [Google Scholar]
  43. Oldstone MBA, Aoki T, Dixon FJ. 43.  1972. The antibody response of mice to murine leukemia virus in spontaneous infection: absence of classical immunologic tolerance. PNAS 69:134–38 [Google Scholar]
  44. Oldstone MBA, Del Villano BC, Dixon FJ. 44.  1976. Autologous immune responses to the major oncornavirus polypeptides in unmanipulated AKR/J mice. J. Virol. 18:176–81 [Google Scholar]
  45. Jamieson BD, Ahmed R. 45.  1988. T-cell tolerance: Exposure to virus in utero does not cause a permanent deletion of specific T cells. PNAS 85:2265–68 [Google Scholar]
  46. Notkins AL, Mahar S, Scheele C, Goffman J. 46.  1966. Infectious virus-antibody complex in the blood of chronically infected mice. J. Exp. Med. 124:81–97 [Google Scholar]
  47. Fields BN. 47.  1972. Genetic manipulation of reovirus—a model for modification of disease. N. Engl. J. Med. 287:1026–33 [Google Scholar]
  48. Sharpe AH, Fields BN. 48.  1985. Pathogenesis of viral infections—basic concepts derived from the reovirus model. N. Engl. J. Med. 312:486–97 [Google Scholar]
  49. Nibert M, Furlong D, Fields BN. 49.  1991. Mechanisms of viral pathogenesis. J. Clin. Investig. 88:727–34 [Google Scholar]
  50. Oldstone MBA, Sinha YN, Blount P, Tishon A, Rodriguez M. 50.  et al. 1982. Virus-induced alterations in homeostasis: alterations in differentiated functions of infected cells in vivo. Science 218:1125–27 [Google Scholar]
  51. Oldstone MBA, Rodriguez M, Daughaday WH, Lampert PW. 51.  1984. Viral perturbation of endocrine function: disordered cell function leads to disturbed homeostasis and disease. Nature 307:278–81 [Google Scholar]
  52. Valsamakis A, Riviere Y, Oldstone MBA. 52.  1987. Perturbation of differentiated functions in vivo during persistent viral infection. III. Decreased growth hormone mRNA. Virology 156:214–20 [Google Scholar]
  53. de la Torre JC, Oldstone MBA. 53.  1992. Selective disruption of growth hormone transcription machinery by viral infection. PNAS 89:9939–43 [Google Scholar]
  54. de la Torre JC, Mallory M, Brot M, Gold L, Koob G. 54.  et al. 1996. Viral persistence in neurons alters synaptic plasticity and cognitive functions without destruction of brain cells. Virology 220:508–15 [Google Scholar]
  55. Oldstone MBA. 55.  1998. Molecular mimicry and immune-mediated diseases. FASEB J. 12:1255–65 [Google Scholar]
  56. Köhler G, Milstein C. 56.  1975. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256:495–97 [Google Scholar]
  57. Fujinami RS, Oldstone MBA, Wroblewska Z, Frankel ME, Koprowski H. 57.  1983. Molecular mimicry in virus infection: cross-reaction of measles virus phosphoprotein or of herpes simplex virus protein with human intermediate filaments. PNAS 80:2346–50 [Google Scholar]
  58. Fujinami RS, Oldstone MBA. 58.  1985. Amino acid homology between the encephalitogenic site of myelin basic protein and virus: mechanism for autoimmunity. Science 230:1043–45 [Google Scholar]
  59. Oldstone MBA, Nerenberg M, Southern P, Price J, Lewicki H. 59.  1991. Virus infection triggers insulin-dependent diabetes mellitus in a transgenic model: role of anti-self (virus) immune response. Cell 65:319–31 [Google Scholar]
  60. von Herrath MG, Dockter J, Oldstone MBA. 60.  1994. How virus induces a rapid or slow onset insulin-dependent diabetes mellitus in a transgenic model. Immunity 1:231–42 [Google Scholar]
  61. Sevilla N, Homann D, von Herrath M, Rodriguez F, Harkins S. 61.  et al. 2000. Virus-induced diabetes in a transgenic model: role of cross-reacting viruses and quantitation of effector T cells needed to cause disease. J. Virol. 74:3284–92 [Google Scholar]
  62. Oldstone MBA, Edelmann KH, McGavern DB, Cruite JT, Welch MJ. 62.  2012. Molecular anatomy and number of antigen-specific CD8 T cells required to cause type 1 diabetes. PLOS Pathog. 8:e1003044 [Google Scholar]
  63. von Herrath MG, Dyrberg T, Oldstone MBA. 63.  1996. Oral insulin treatment suppresses virus-induced antigen-specific destruction of beta cells and prevents autoimmune diabetes in transgenic mice. J. Clin. Investig. 98:1324–31 [Google Scholar]
  64. Oldstone MBA, Blount P, Southern PJ, Lampert PW. 64.  1986. Cytoimmunotherapy for persistent virus infection: unique clearance pattern from the central nervous system. Nature 321:239–43 [Google Scholar]
  65. Brooks DG, McGavern DB, Oldstone MBA. 65.  2006. Reprogramming of antiviral T cells prevents inactivation and restores T cell activity during persistent viral infection. J. Clin. Investig. 116:1675–85 [Google Scholar]
  66. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP. 66.  et al. 2006. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439:682–87 [Google Scholar]
  67. Ng CT, Snell LM, Brooks DG, Oldstone MBA. 67.  2013. Networking at the level of host immunity: immune cell interactions during persistent viral infections. Cell Host Microbe 13:652–64 [Google Scholar]
  68. Wolchok JD, Hodi FS, Weber JS, Allison JP, Urba WJ. 68.  et al. 2013. Development of ipilimumab: a novel immunotherapeutic approach for the treatment of advanced melanoma. Ann. N.Y. Acad. Sci. 1291:1–13 [Google Scholar]
  69. Harshman LC, Choueiri TK, Drake C, Hodi FS. 69.  2014. Subverting the B7-H1/PD-1 pathway in advanced melanoma and kidney cancer. Cancer J. 20:272–80 [Google Scholar]
  70. Ott PA, Hodi FS, Robert C. 70.  2013. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefits in melanoma patients. Clin. Cancer Res. 19:5300–9 [Google Scholar]
  71. Lu J, Lee-Gabel L, Nadeau MC, Ferencz TM, Soefje SA. 71.  2014. Clinical evaluation of compounds targeting PD-1/PD-L1 pathway for cancer immunotherapy. J. Oncol. Pharm. Pract. In press. doi: 10.1177/1078155214538087
  72. Wilson EB, Brooks DG. 72.  2010. Translating insights from persistent LCMV infection into anti-HIV immunity. Immunol. Res. 48:3–13 [Google Scholar]
  73. Cao W, Henry MD, Borrow P, Yamada H, Elder JH. 73.  et al. 1998. Identification of α-dystroglycan as a receptor for lymphocytic choriomeningitis virus and Lassa fever virus. Science 282:2079–81 [Google Scholar]
  74. Kunz S, Sevilla N, McGavern DB, Campbell KP, Oldstone MBA. 74.  2001. Molecular analysis of the interaction of LCMV with its cellular receptor α-dystroglycan. J. Cell Biol. 155:301–10 [Google Scholar]
  75. Sevilla N, McGavern DB, Teng C, Kunz S, Oldstone MBA. 75.  2004. Viral targeting of hematopoietic progenitors and inhibition of DC maturation as a dual strategy for immune subversion. J. Clin. Investig. 113:737–45 [Google Scholar]
  76. Ng CT, Oldstone MBA. 76.  2012. Infected CD8α dendritic cells are the predominant source of IL-10 during establishment of persistent viral infection. PNAS 109:14116–21 [Google Scholar]
  77. Oldstone MBA, Teijaro JR, Walsh KB, Rosen H. 77.  2013. Dissecting influenza virus pathogenesis uncovers a novel chemical approach to combat the infection. Virology 435:92–101 [Google Scholar]
  78. Teijaro JR, Walsh KB, Cahalan S, Fremgen DM, Roberts E. 78.  et al. 2011. Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection. Cell 146:980–91 [Google Scholar]
  79. Oldstone MBA, Rosen H. 79.  2014. Cytokine storm plays a direct role in the morbidity and mortality from influenza virus infection and is chemically treatable with a single sphingosine-1-phosphate agonist molecule. Curr. Top. Microbiol. Immunol. 378:129–47 [Google Scholar]
/content/journals/10.1146/annurev-pathol-012615-044107
Loading
/content/journals/10.1146/annurev-pathol-012615-044107
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