1. Selgrade MK, Cooper GS, Germolec DR, Heindel JJ. Linking environmental agents and autoimmune disease: an agenda for future research. Environ Health Perspect. 1999;107(Suppl. 5):811–3. [PMC free article] [PubMed] [Google Scholar]
3. Bowles NE, Richardson PJ, Olsen EG, Archard LC. Detection of Coxsackie-B-virus-specific RNA sequences in myocardial biopsy samples from patients with myocarditis and dilated cardiomyopathy. Lancet. 1986;1:1120–3. [PubMed] [Google Scholar]
4. Pauschinger M, Doerner A, Kuehl U, et al. Enteroviral RNA replication in the myocardium of patients with left ventricular dysfunction and clinically suspected myocarditis. Circulation. 1999;99:889–95. [PubMed] [Google Scholar]
5. Fairweather D, Kaya Z, Shellam GR, Lawson CM, Rose NR. From infection to autoimmunity. J Autoimmun. 2001;16:175–86. [PubMed] [Google Scholar]
6. Herzum M, Ruppert V, Kuytz B, Jomaa H, Nakamura I, Maisch B. Coxsackievirus B3 infection leads to cell death of cardiac myocytes. J Mol Cell Cardiol. 1994;26:907–13. [PubMed] [Google Scholar]
7. McManus BM, Chow LH, Wilson JE, et al. Direct myocardial injury by enterovirus: a central role in the evolution of murine myocarditis. Clin Immunol Immunopathol. 1993;68:159–69. [PubMed] [Google Scholar]
8. Chow LH, Beisel KW, McManus BM. Enteroviral infection of mice with severe combined immunodeficiency. Lab Invest. 1992;66:24–31. Evidence for direct viral pathogenesis of myocardial injury. [PubMed] [Google Scholar]
9. Si X, Wang Y, Wong J, Zhang J, McManus BM, Luo H. Dysregulation of the ubiquitin–proteasome system by curcumin suppresses coxsackievirus B3 replication. J Virol. 2007;81:3142–50. [PMC free article] [PubMed] [Google Scholar]
10. Luo H, Zhang J, Cheung C, Suarez A, McManus BM, Yang D. Proteasome inhibition reduces coxsackievirus B3 replication in murine cardiomyocytes. Am J Pathol. 2003;163:381–5. [PMC free article] [PubMed] [Google Scholar]
11. Luo H, Zhang J, Dastvan F, et al. Ubiquitin-dependent proteolysis of cyclin D1 is associated with coxsackievirus-induced cell growth arrest. J Virol. 2003;77:1–9. [PMC free article] [PubMed] [Google Scholar]
12. Woodruff JF, Woodruff JJ. Involvement of T lymphocytes in the pathogenesis of coxsackie virus B3 heart disease. J Immunol. 1974;113:1726–34. [PubMed] [Google Scholar]
14. Guthrie M, Lodge PA, Huber SA. Cardiac injury in myocarditis induced by Coxsackievirus group B, type 3 in Balb/c mice is mediated by Lyt 2 + cytolytic lymphocytes. Cell Immunol. 1984;88:558–67. [PubMed] [Google Scholar]
15. Huber SA. Coxsackievirus-induced myocarditis is dependent on distinct immunopathogenic responses in different strains of mice. Lab Invest. 1997;76:691–701. [PubMed] [Google Scholar]
16. Henke A, Huber S, Stelzner A, Whitton JL. The role of CD8+ T lymphocytes in coxsackievirus B3-induced myocarditis. J Virol. 1995;69:6720–8. [PMC free article] [PubMed] [Google Scholar]
17. Gauntt CJ, Arizpe HM, Higdon AL, et al. Molecular mimicry, anti-coxsackievirus B3 neutralizing monoclonal antibodies, and myocarditis. J Immunol. 1995;154:2983–95. [PubMed] [Google Scholar]
18. Gauntt CJ, Higdon AL, Arizpe HM, et al. Epitopes shared between coxsackievirus B3 (CVB3) and normal heart tissue contribute to CVB3-induced murine myocarditis. Clin Immunol Immunopathol. 1993;68:129–34. [PubMed] [Google Scholar]
19. Huber SA. Autoimmunity in myocarditis: relevance of animal models. Clin Immunol Immunopathol. 1997;83:93–102. [PubMed] [Google Scholar]
20. Huber SA, Moraska A, Cunningham M. Alterations in major histocompatibility complex association of myocarditis induced by coxsackievirus B3 mutants selected with monoclonal antibodies to group A streptococci. Proc Natl Acad Sci USA. 1994;91:5543–7. [PMC free article] [PubMed] [Google Scholar]
21. Rose NR, Hill SL. The pathogenesis of postinfectious myocarditis. Clin Immunol Immunopathol. 1996;80:S92–9. [PubMed] [Google Scholar]
22. Tam PE, Fontana DR, Messner RP. Coxsackievirus B1-induced chronic inflammatory myopathy: differences in induction of autoantibodies to muscle and nuclear antigens by cloned myopathic and amyopathic viruses. J Lab Clin Med. 2003;142:196–204. [PubMed] [Google Scholar]
23. Whitton JL, Feuer R. Myocarditis, microbes and autoimmunity. Autoimmunity. 2004;37:375–86. [PubMed] [Google Scholar]
24. Schwimmbeck PL, Bigalke B, Schulze K, Pauschinger M, Kuhl U, Schultheiss HP. The humoral immune response in viral heart disease: characterization and pathophysiological significance of antibodies. Med Microbiol Immunol. 2004;193:115–9. [PubMed] [Google Scholar]
25. Bengtsson E, Birke G, Wingstrand H. Acute non-specific myocarditis in scarlet fever and acute haemolytic tonsillitis A clinical investigation of 3,069 cases of scarlet fever, 798 cases of acute tonsillitis, and 333 cases of haemolytic streptococcus carriers. Cardiologia. 1951;18:360–74. [PubMed] [Google Scholar]
26. Friedman I, Laufer A, Ron N, Davies AM. Experimental myocarditis: in vitro and in vivo studies of lymphocytes sensitized to heart extracts and group A streptococci. Immunology. 1971;20:225–32. [PMC free article] [PubMed] [Google Scholar]
27. Cunningham MW, McCormack JM, Talaber LR, et al. Human monoclonal antibodies reactive with antigens of the group A Streptococcus and human heart. J Immunol. 1988;141:2760–6. [PubMed] [Google Scholar]
28. Adderson EE, Shikhman AR, Ward KE, Cunningham MW. Molecular analysis of polyreactive monoclonal antibodies from rheumatic carditis: human anti-N-acetylglucosamine/anti-myosin antibody V region genes. J Immunol. 1998;161:2020–31. [PubMed] [Google Scholar]
29. Mertens NM, Galvin JE, Adderson EE, Cunningham MW. Molecular analysis of cross-reactive anti-myosin/anti-streptococcal mouse monoclonal antibodies. Mol Immunol. 2000;37:901–13. [PubMed] [Google Scholar]
30. Cunningham MW, McCormack JM, Fenderson PG, Ho MK, Beachey EH, Dale JB. Human and murine antibodies cross-reactive with streptococcal M protein and myosin recognize the sequence GLN-LYS-SER-LYS-GLN in M protein. J Immunol. 1989;143:2677–83. [PubMed] [Google Scholar]
31. Fenderson PG, Fischetti VA, Cunningham MW. Tropomyosin shares immunologic epitopes with group A streptococcal M proteins. J Immunol. 1989;142:2475–81. [PubMed] [Google Scholar]
32. Galvin JE, Hemric ME, Ward K, Cunningham MW. Cytotoxic mAb from rheumatic carditis recognizes heart valves and laminin. J Clin Invest. 2000;106:217–24. [PMC free article] [PubMed] [Google Scholar]
33. Ellis NM, Li Y, Hildebrand W, Fischetti VA, Cunningham MW. T cell mimicry and epitope specificity of cross-reactive T cell clones from rheumatic heart disease. J Immunol. 2005;175:5448–56. [PubMed] [Google Scholar]
34. Fae KC, da Silva DD, Oshiro SE, et al. Mimicry in recognition of cardiac myosin peptides by heart-intralesional T cell clones from rheumatic heart disease. J Immunol. 2006;176:5662–70. [PubMed] [Google Scholar]
35. Raizada V, Williams RC, Jr., Chopra P, et al. Tissue distribution of lymphocytes in rheumatic heart valves as defined by monoclonal anti-T cell antibodies. Am J Med. 1983;74:90–6. [PubMed] [Google Scholar]
36. Guilherme L, Oshiro SE, Fae KC, et al. T-cell reactivity against streptococcal antigens in the periphery mirrors reactivity of heart-infiltrating T lymphocytes in rheumatic heart disease patients. Infect Immun. 2001;69:5345–51. [PMC free article] [PubMed] [Google Scholar]
37. Cunningham MW, Antone SM, Smart M, Liu R, Kosanke S. Molecular analysis of human cardiac myosin-cross-reactive B- and T-cell epitopes of the group A streptococcal M5 protein. Infect Immun. 1997;65:3913–23. [PMC free article] [PubMed] [Google Scholar]
38. Quinn A, Kosanke S, Fischetti VA, Factor SM, Cunningham MW. Induction of autoimmune valvular heart disease by recombinant streptococcal m protein. Infect Immun. 2001;69:4072–8. [PMC free article] [PubMed] [Google Scholar]
39. Murphy GE, Swift HF. The induction of rheumatic-like cardiac lesions in rabbits by repeated focal infections with group A streptococci; comparison with the cardiac lesions of serum disease. J Exp Med. 1950;91:485–98. [PMC free article] [PubMed] [Google Scholar]
40. Cromartie WJ, Craddock JG. Rheumatic-like cardiac lesions in mice. Science. 1966;154:285–7. [PubMed] [Google Scholar]
41. Singer HS, Giuliano JD, Hansen BH, et al. Antibodies against human putamen in children with Tourette syndrome. Neurology. 1998;50:1618–24. [PubMed] [Google Scholar]
42. Pavone P, Bianchini R, Parano E, et al. Anti-brain antibodies in PANDAS versus uncomplicated streptococcal infection. Pediatr Neurol. 2004;30:107–10. [PubMed] [Google Scholar]
43. Kurlan R. Tourette’s syndrome and ‘PANDAS’: will the relation bear out? Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection. Neurology. 1998;50:1530–4. [PubMed] [Google Scholar]
44. Snider LA, Swedo SE. Post-streptococcal autoimmune disorders of the central nervous system. Curr Opin Neurol. 2003;16:359–65. [PubMed] [Google Scholar]
45. Bronze MS, Dale JB. Epitopes of streptococcal M proteins that evoke antibodies that cross-react with human brain. J Immunol. 1993;151:2820–8. [PubMed] [Google Scholar]
46. Husby G, van de Rijn I, Zabriskie JB, Abdin ZH, Williams RC., Jr. Antibodies reacting with cytoplasm of subthalamic and caudate nuclei neurons in chorea and acute rheumatic fever. J Exp Med. 1976;144:1094–110. [PMC free article] [PubMed] [Google Scholar]
47. Kirvan CA, Swedo SE, Heuser JS, Cunningham MW. Mimicry and autoantibody-mediated neuronal cell signaling in Sydenham chorea. Nat Med. 2003;9:914–20. [PubMed] [Google Scholar]
48. Kirvan CA, Swedo SE, Snider LA, Cunningham MW. Antibody-mediated neuronal cell signaling in behavior and movement disorders. J Neuroimmunol. 2006;179:173–9. [PubMed] [Google Scholar]
49. Kirvan CA, Cox CJ, Swedo SE, Cunningham MW. Tubulin is a neuronal target of autoantibodies in Sydenham’s chorea. J Immunol. 2007;178:7412–21. [PubMed] [Google Scholar]
50. Hoffman KL, Hornig M, Yaddanapudi K, Jabado O, Lipkin WI. A murine model for neuropsychiatric disorders associated with group A beta-hemolytic streptococcal infection. J Neurosci. 2004;24:1780–91. [PMC free article] [PubMed] [Google Scholar]
51. Soares MB, Santos RR. Immunopathology of cardiomyopathy in the experimental Chagas disease. Mem Inst Oswaldo Cruz. 1999;94(Suppl. 1):257–62. [PubMed] [Google Scholar]
52. Leon JS, Engman DM. The significance of autoimmunity in the pathogenesis of Chagas heart disease. Front Biosci. 2003;8:e315–22. [PubMed] [Google Scholar]
53. Cunha-Neto E, Bilate AM, Hyland KV, Fonseca SG, Kalil J, Engman DM. Induction of cardiac autoimmunity in Chagas heart disease: a case for molecular mimicry. Autoimmunity. 2006;39:41–54. [PubMed] [Google Scholar]
54. Sartori AM, Shikanai-Yasuda MA, Amato Neto V, Lopes MH. Follow-up of 18 patients with human immunodeficiency virus infection and chronic Chagas’ disease, with reactivation of Chagas’ disease causing cardiac disease in three patients. Clin Infect Dis. 1998;26:177–9. [PubMed] [Google Scholar]
55. Pereira JB, Wilcox HP, Coura JR. The evolution of chronic chagasic cardiopathy. Rev Soc Bras Med Trop. 1992;25:101–8. I. The influence of parasitemia. [PubMed] [Google Scholar]
56. Elias FE, Vigliano CA, Laguens RP, Levin MJ, Berek C. Analysis of the presence of Trypanosoma cruzi in the heart tissue of three patients with chronic Chagas’ heart disease. Am J Trop Med Hyg. 2003;68:242–7. [PubMed] [Google Scholar]
57. Higuchi ML, De Morais CF, Pereira Barreto AC, et al. The role of active myocarditis in the development of heart failure in chronic Chagas’ disease: a study based on endomyocardial biopsies. Clin Cardiol. 1987;10:665–70. [PubMed] [Google Scholar]
58. Abel LC, Rizzo LV, Ianni B, et al. Chronic Chagas’ disease cardiomyopathy patients display an increased IFN-gamma response to Trypanosoma cruzi infection. J Autoimmun. 2001;17:99–107. [PubMed] [Google Scholar]
59. Reis MM, Higuchi Mde L, Benvenuti LA, et al. An in situ quantitative immunohistochemical study of cytokines and IL-2R+ in chronic human chagasic myocarditis: correlation with the presence of myocardial Trypanosoma cruzi antigens. Clin Immunol Immunopathol. 1997;83:165–72. [PubMed] [Google Scholar]
60. Cunha-Neto E, Dzau VJ, Allen PD, et al. Cardiac gene expression profiling provides evidence for cytokinopathy as a molecular mechanism in Chagas’ disease cardiomyopathy. Am J Pathol. 2005;167:305–13. [PMC free article] [PubMed] [Google Scholar]
61. Teixeira AR, Teixeira G, Macedo V, Prata A. Trypanosoma cruzi-sensitized T-lymphocyte mediated 51CR release from human heart cells in Chagas’ disease. Am J Trop Med Hyg. 1978;27:1097–107. [PubMed] [Google Scholar]
63. Mosca W, Plaja J, Hubsch R, Cedillos R. Longitudinal study of immune response in human Chagas’ disease. J Clin Microbiol. 1985;22:438–41. [PMC free article] [PubMed] [Google Scholar]
64. Gruber A, Zingales B. Trypanosoma cruzi: characterization of two recombinant antigens with potential application in the diagnosis of Chagas’ disease. Exp Parasitol. 1993;76:1–12. [PubMed] [Google Scholar]
65. Cunha-Neto E, Duranti M, Gruber A, et al. Autoimmunity in Chagas disease cardiopathy: biological relevance of a cardiac myosin-specific epitope crossreactive to an immunodominant Trypanosoma cruzi antigen. Proc Natl Acad Sci USA. 1995;92:3541–5. [PMC free article] [PubMed] [Google Scholar]
66. Cunha-Neto E, Coelho V, Guilherme L, Fiorelli A, Stolf N, Kalil J. Autoimmunity in Chagas’ disease. J Clin Invest. 1996;98:1709–12. Identification of cardiac myosin-B13 Trypanosoma cruzi protein crossreactive T cell clones in heart lesions of a chronic Chagas’ cardiomyopathy patient. [PMC free article] [PubMed] [Google Scholar]
67. Abel LC, Kalil J, Cunha Neto E. Molecular mimicry between cardiac myosin and Trypanosoma cruzi antigen B13: identification of a B13-driven human T cell clone that recognizes cardiac myosin. Braz J Med Biol Res. 1997;30:1305–8. [PubMed] [Google Scholar]
68. Lannes-Vieira J. Trypanosoma cruzi-elicited CD8+ T cell-mediated myocarditis: chemokine receptors and adhesion molecules as potential therapeutic targets to control chronic inflammation? Mem Inst Oswaldo Cruz. 2003;98:299–304. [PubMed] [Google Scholar]
69. Rizzo LV, Cunha-Neto E, Teixeira AR. Autoimmunity in Chagas’ disease: specific inhibition of reactivity of CD4+ T cells against myosin in mice chronically infected with Trypanosoma cruzi. Infect Immun. 1989;57:2640–4. [PMC free article] [PubMed] [Google Scholar]
70. dos Santos RR, Rossi MA, Laus JL, Silva JS, Savino W, Mengel J. Anti-CD4 abrogates rejection and reestablishes long-term tolerance to syngeneic newborn hearts grafted in mice chronically infected with Trypanosoma cruzi. J Exp Med. 1992;175:29–39. [PMC free article] [PubMed] [Google Scholar]
71. Ribeiro-Dos-Santos R, Mengel JO, Postol E, et al. A heart-specific CD4+ T-cell line obtained from a chronic chagasic mouse induces carditis in heart-immunized mice and rejection of normal heart transplants in the absence of Trypanosoma cruzi. Parasite Immunol. 2001;23:93–101. [PubMed] [Google Scholar]
72. Girones N, Rodriguez CI, Carrasco-Marin E, Hernaez RF, de Rego JL, Fresno M. Dominant T- and B-cell epitopes in an autoantigen linked to Chagas’ disease. J Clin Invest. 2001;107:985–93. [PMC free article] [PubMed] [Google Scholar]
73. Oliveira MF, Bijovsky AT, Carvalho TU, de Souza W, Alves MJ, Colli W. A monoclonal antibody to Trypanosoma cruzi trypomastigotes recognizes a myosin tail epitope. Parasitol Res. 2001;87:1043–9. [PubMed] [Google Scholar]
74. Giordanengo L, Guinazu N, Stempin C, Fretes R, Cerban F, Gea S. Cruzipain, a major Trypanosoma cruzi antigen, conditions the host immune response in favor of parasite. Eur J Immunol. 2002;32:1003–11. [PubMed] [Google Scholar]
75. McCormick TS, Rowland EC. Trypanosoma cruzi: cross-reactive anti-heart autoantibodies produced during infection in mice. Exp Parasitol. 1989;69:393–401. [PubMed] [Google Scholar]
76. Leon JS, Daniels MD, Toriello KM, Wang K, Engman DM. A cardiac myosin-specific autoimmune response is induced by immunization with Trypanosoma cruzi proteins. Infect Immun. 2004;72:3410–7. [PMC free article] [PubMed] [Google Scholar]
78. Meneghelli UG. Chagasic enteropathy. Rev Soc Bras Med Trop. 2004;37:252–60. [PubMed] [Google Scholar]
79. Van Voorhis WC, Eisen H. FL-160: a surface antigen of Trypanosoma cruzi that mimics mammalian nervous tissue. J Exp Med. 1989;169:641–52. [PMC free article] [PubMed] [Google Scholar]
80. Van Voorhis WC, Barrett L, Koelling R, Farr AG. FL-160 proteins of Trypanosoma cruzi are expressed from a multigene family and contain two distinct epitopes that mimic nervous tissues. J Exp Med. 1993;178:681–94. [PMC free article] [PubMed] [Google Scholar]
81. Van Voorhis WC, Schlekewy L, Trong HL. Molecular mimicry by Trypanosoma cruzi: the F1-160 epitope that mimics mammalian nerve can be mapped to a 12-amino acid peptide. Proc Natl Acad Sci USA. 1991;88:5993–7. [PMC free article] [PubMed] [Google Scholar]
82. Petry K, Eisen H. Chemical characterization of epitopes common to Trypanosoma cruzi and mammalian nervous cells. Mem Inst Oswaldo Cruz. 1988;83(Suppl. 1):498–501. [PubMed] [Google Scholar]
83. Gea S, Ordonez P, Cerban F, Iosa D, Chizzolini C, Vottero-Cima E. Chagas’ disease cardioneuropathy: association of anti-Trypanosoma cruzi and anti-sciatic nerve antibodies. Am J Trop Med Hyg. 1993;49:581–8. [PubMed] [Google Scholar]
84. Steere AC, Schoen RT, Taylor E. The clinical evolution of Lyme arthritis. Ann Intern Med. 1987;107:725–31. [PubMed] [Google Scholar]
85. Nocton JJ, Dressler F, Rutledge BJ, Rys PN, Persing DH, Steere AC. Detection of Borrelia burgdorferi DNA by polymerase chain reaction in synovial fluid from patients with Lyme arthritis. N Engl J Med. 1994;330:229–34. [PubMed] [Google Scholar]
86. Steere AC, Levin RE, Molloy PJ, Kalish RA, Abraham JH, 3rd, Liu NY, Schmid CH. Treatment of Lyme arthritis. Arthritis Rheum. 1994;37:878–88. [PubMed] [Google Scholar]
87. Carlson D, Hernandez J, Bloom BJ, Coburn J, Aversa JM, Steere AC. Lack of Borrelia burgdorferi DNA in synovial samples from patients with antibiotic treatment-resistant Lyme arthritis. Arthritis Rheum. 1999;42:2705–9. [PubMed] [Google Scholar]
88. Malawista SE. Resolution of Lyme arthritis, acute or prolonged: a new look. Inflammation. 2000;24:493–504. [PubMed] [Google Scholar]
89. Chen J, Field JA, Glickstein L, Molloy PJ, Huber BT, Steere AC. Association of antibiotic treatment-resistant Lyme arthritis with T cell responses to dominant epitopes of outer surface protein A of Borrelia burgdorferi. Arthritis Rheum. 1999;42:1813–22. [PubMed] [Google Scholar]
90. Lengl-Janssen B, Strauss AF, Steere AC, Kamradt T. The T helper cell response in Lyme arthritis: differential recognition of Borrelia burgdorferi outer surface protein A in patients with treatment-resistant or treatment-responsive Lyme arthritis. J Exp Med. 1994;180:2069–78. [PMC free article] [PubMed] [Google Scholar]
91. Kalish RA, Leong JM, Steere AC. Association of treatment-resistant chronic Lyme arthritis with HLA-DR4 and antibody reactivity to OspA and OspB of Borrelia burgdorferi. Infect Immun. 1993;61:2774–9. [PMC free article] [PubMed] [Google Scholar]
92. Kalish RA, Leong JM, Steere AC. Early and late antibody responses to full-length and truncated constructs of outer surface protein A of Borrelia burgdorferi in Lyme disease. Infect Immun. 1995;63:2228–35. [PMC free article] [PubMed] [Google Scholar]
93. Akin E, McHugh GL, Flavell RA, Fikrig E, Steere AC. The immunoglobulin (IgG) antibody response to OspA and OspB correlates with severe and prolonged Lyme arthritis and the IgG response to P35 correlates with mild and brief arthritis. Infect Immun. 1999;67:173–81. [PMC free article] [PubMed] [Google Scholar]
94. Meyer AL, Trollmo C, Crawford F, et al. Direct enumeration of Borrelia-reactive CD4 T cells ex vivo by using MHC class II tetramers. Proc Natl Acad Sci USA. 2000;97:11433–8. [PMC free article] [PubMed] [Google Scholar]
95. Steere AC, Gross D, Meyer AL, Huber BT. Autoimmune mechanisms in antibiotic treatment-resistant lyme arthritis. J Autoimmun. 2001;16:263–8. [PubMed] [Google Scholar]
96. Gross DM, Forsthuber T, Tary-Lehmann M, et al. Identification of LFA-1 as a candidate autoantigen in treatment-resistant Lyme arthritis. Science. 1998;281:703–6. [PubMed] [Google Scholar]
97. Akin E, Aversa J, Steere AC. Expression of adhesion molecules in synovia of patients with treatment-resistant lyme arthritis. Infect Immun. 2001;69:1774–80. [PMC free article] [PubMed] [Google Scholar]
98. Trollmo C, Meyer AL, Steere AC, Hafler DA, Huber BT. Molecular mimicry in Lyme arthritis demonstrated at the single cell level: LFA-1 alpha L is a partial agonist for outer surface protein A-reactive T cells. J Immunol. 2001;166:5286–91. [PubMed] [Google Scholar]
99. Steere AC, Falk B, Drouin EE, Baxter-Lowe LA, Hammer J, Nepom GT. Binding of outer surface protein A and human lymphocyte function-associated antigen 1 peptides to HLA-DR molecules associated with antibiotic treatment-resistant Lyme arthritis. Arthritis Rheum. 2003;48:534–40. [PubMed] [Google Scholar]
100. Kalish RS, Wood JA, Golde W, et al. Human T lymphocyte response to Borrelia burgdorferi infection: no correlation between human leukocyte function antigen type 1 peptide response and clinical status. J Infect Dis. 2003;187:102–8. [PubMed] [Google Scholar]
101. Maier B, Molinger M, Cope AP, et al. Multiple cross-reactive self-ligands for Borrelia burgdorferi-specific HLA-DR4-restricted T cells. Eur J Immunol. 2000;30:448–57. [PubMed] [Google Scholar]
102. Barthold SW, Moody KD, Terwilliger GA, Duray PH, Jacoby RO, Steere AC. Experimental Lyme arthritis in rats infected with Borrelia burgdorferi. J Infect Dis. 1988;157:842–6. [PubMed] [Google Scholar]
103. Barthold SW, Beck DS, Hansen GM, Terwilliger GA, Moody KD. Lyme borreliosis in selected strains and ages of laboratory mice. J Infect Dis. 1990;162:133–8. [PubMed] [Google Scholar]
104. Ma Y, Seiler KP, Eichwald EJ, Weis JH, Teuscher C, Weis JJ. Distinct characteristics of resistance to Borrelia burgdorferi-induced arthritis in C57BL/6N mice. Infect Immun. 1998;66:161–8. [PMC free article] [PubMed] [Google Scholar]
105. Schaible UE, Kramer MD, Museteanu C, Zimmer G, Mossmann H, Simon MM. The severe combined immunodeficiency (scid) mouse. A laboratory model for the analysis of Lyme arthritis and carditis. J Exp Med. 1989;170:1427–32. [PMC free article] [PubMed] [Google Scholar]
106. Barthold SW, de Souza M. Exacerbation of Lyme arthritis in beige mice. J Infect Dis. 1995;172:778–84. [PubMed] [Google Scholar]
107. Anguita J, Rincon M, Samanta S, Barthold SW, Flavell RA, Fikrig E. Borrelia burgdorferi-infected, interleukin-6-deficient mice have decreased Th2 responses and increased lyme arthritis. J Infect Dis. 1998;178:1512–5. [PubMed] [Google Scholar]
108. Brown JP, Zachary JF, Teuscher C, Weis JJ, Wooten RM. Dual role of interleukin-10 in murine Lyme disease: regulation of arthritis severity and host defense. Infect Immun. 1999;67:5142–50. [PMC free article] [PubMed] [Google Scholar]
109. McKisic MD, Redmond WL, Barthold SW. Cutting edge: T cell-mediated pathology in murine Lyme borreliosis. J Immunol. 2000;164:6096–9. [PubMed] [Google Scholar]
110. Sigal LH. Lyme disease: a review of aspects of its immunology and immunopathogenesis. Annu Rev Immunol. 1997;15:63–92. [PubMed] [Google Scholar]
111. Sigal LH, Tatum AH. Lyme disease patients’ serum contains IgM antibodies to Borrelia burgdorferi that cross-react with neuronal antigens. Neurology. 1988;38:1439–42. [PubMed] [Google Scholar]
112. Wang WZ, Fredrikson S, Sun JB, Link H. Lyme neuroborreliosis: evidence for persistent up-regulation of Borrelia burgdorferi-reactive cells secreting interferon-gamma. Scand J Immunol. 1995;42:694–700. [PubMed] [Google Scholar]
113. Garcia-Monco JC, Coleman JL, Benach JL. Antibodies to myelin basic protein in Lyme disease. J Infect Dis. 1988;158:667–8. [PubMed] [Google Scholar]
114. Sigal LH. Cross-reactivity between Borrelia burgdorferi flagellin and a human axonal 64,000 molecular weight protein. J Infect Dis. 1993;167:1372–8. [PubMed] [Google Scholar]
115. Dai Z, Lackland H, Stein S, et al. Molecular mimicry in Lyme disease: monoclonal antibody H9724 to B. Biochim Biophys Acta. 1993;1181:97–100. burgdorferi flagellin specifically detects chaperonin-HSP60. [PubMed] [Google Scholar]
116. Fikrig E, Berland R, Chen M, Williams S, Sigal LH, Flavell RA. Serologic response to the Borrelia burgdorferi flagellin demonstrates an epitope common to a neuroblastoma cell line. Proc Natl Acad Sci USA. 1993;90:183–7. [PMC free article] [PubMed] [Google Scholar]
117. Sigal LH, Williams S. A monoclonal antibody to Borrelia burgdorferi flagellin modifies neuroblastoma cell neuritogenesis in vitro: a possible role for autoimmunity in the neuropathy of Lyme disease. Infect Immun. 1997;65:1722–8. [PMC free article] [PubMed] [Google Scholar]
118. Alaedini A, Latov N. Antibodies against OspA epitopes of Borrelia burgdorferi cross-react with neural tissue. J Neuroimmunol. 2005;159:192–5. [PubMed] [Google Scholar]
119. Hemmer B, Gran B, Zhao Y, et al. Identification of candidate T-cell epitopes and molecular mimics in chronic Lyme disease. Nat Med. 1999;5:1375–82. [PubMed] [Google Scholar]
120. Pachner AR, Cadavid D, Shu G, et al. Central and peripheral nervous system infection, immunity, and inflammation in the NHP model of Lyme borreliosis. Ann Neurol. 2001;50:330–8. [PubMed] [Google Scholar]
121. Ramesh G, Alvarez AL, Roberts ED, et al. Pathogenesis of Lyme neuroborreliosis: Borrelia burgdorferi lipoproteins induce both proliferation and apoptosis in rhesus monkey astrocytes. Eur J Immunol. 2003;33:2539–50. [PubMed] [Google Scholar]
122. Steiner I. Treating post Lyme disease: trying to solve one equation with too many unknowns. Neurology. 2003;60:1888–9. [PubMed] [Google Scholar]
123. Liesegang TJ. Epidemiology of ocular herpes simplex. Arch Ophthalmol. 1989;107:1160–5. Natural history in Rochester, Minn, 1950 through 1982. [PubMed] [Google Scholar]
124. Liesegang TJ. Classification of herpes simplex virus keratitis and anterior uveitis. Cornea. 1999;18:127–43. [PubMed] [Google Scholar]
125. Heiligenhaus A, Steuhl KP. Treatment of HSV-1 stromal keratitis with topical cyclosporin A: a pilot study. Graefes Arch Clin Exp Ophthalmol. 1999;237:435–8. [PubMed] [Google Scholar]
126. Fenton RR, Molesworth-Kenyon S, Oakes JE, Lausch RN. Linkage of IL-6 with neutrophil chemoattractant expression in virus-induced ocular inflammation. Invest Ophthalmol Vis Sci. 2002;43:737–43. [PubMed] [Google Scholar]
127. Lausch RN, Chen SH, Tumpey TM, Su YH, Oakes JE. Early cytokine synthesis in the excised mouse cornea. J Interferon Cytokine Res. 1996;16:35–40. [PubMed] [Google Scholar]
128. Staats HF, Lausch RN. Cytokine expression in vivo during murine herpetic stromal keratitis. Effect of protective antibody therapy. J Immunol. 1993;151:277–83. [PubMed] [Google Scholar]
129. Tumpey TM, Chen SH, Oakes JE, Lausch RN. Neutrophil-mediated suppression of virus replication after herpes simplex virus type 1 infection of the murine cornea. J Virol. 1996;70:898–904. [PMC free article] [PubMed] [Google Scholar]
130. Thomas J, Gangappa S, Kanangat S, Rouse BT. On the essential involvement of neutrophils in the immunopathologic disease: herpetic stromal keratitis. J Immunol. 1997;158:1383–91. [PubMed] [Google Scholar]
131. Hendricks RL, Tumpey TM, Finnegan A. IFN-gamma and IL-2 are protective in the skin but pathologic in the corneas of HSV-1-infected mice. J Immunol. 1992;149:3023–8. [PubMed] [Google Scholar]
132. Bauer D, Mrzyk S, Van Rooijen N, Steuhl KP, Heiligenhaus A. Incidence and severity of herpetic stromal keratitis: impaired by the depletion of lymph node macrophages. Exp Eye Res. 2001;72:261–9. [PubMed] [Google Scholar]
133. Habu S, Akamatsu K, Tamaoki N, Okumura K. In vivo significance of NK cell on resistance against virus (HSV-1) infections in mice. J Immunol. 1984;133:2743–7. [PubMed] [Google Scholar]
134. Zisman B, Hirsch MS, Allison AC. Selective effects of anti-macrophage serum, silica and anti-lymphocyte serum on pathogenesis of herpes virus infection of young adult mice. J Immunol. 1970;104:1155–9. [PubMed] [Google Scholar]
135. Bouley DM, Kanangat S, Wire W, Rouse BT. Characterization of herpes simplex virus type-1 infection and herpetic stromal keratitis development in IFN-gamma knockout mice. J Immunol. 1995;155:3964–71. [PubMed] [Google Scholar]
136. Tang Q, Hendricks RL. Interferon gamma regulates platelet endothelial cell adhesion molecule 1 expression and neutrophil infiltration into herpes simplex virus-infected mouse corneas. J Exp Med. 1996;184:1435–47. [PMC free article] [PubMed] [Google Scholar]
137. Streilein JW, Dana MR, Ksander BR. Immunity causing blindness: five different paths to herpes stromal keratitis. Immunol Today. 1997;18:443–9. [PubMed] [Google Scholar]
138. Halford WP, Gebhardt BM, Carr DJ. Persistent cytokine expression in trigeminal ganglion latently infected with herpes simplex virus type 1. J Immunol. 1996;157:3542–9. [PubMed] [Google Scholar]
139. Gangappa S, Babu JS, Thomas J, Daheshia M, Rouse BT. Virus-induced immunoinflammatory lesions in the absence of viral antigen recognition. J Immunol. 1998;161:4289–300. [PubMed] [Google Scholar]
140. Maggs DJ, Chang E, Nasisse MP, Mitchell WJ. Persistence of herpes simplex virus type 1 DNA in chronic conjunctival and eyelid lesions of mice. J Virol. 1998;72:9166–72. [PMC free article] [PubMed] [Google Scholar]
141. Lundberg P, Welander P, Han X, Cantin E. Herpes simplex virus type 1 DNA is immunostimulatory in vitro and in vivo. J Virol. 2003;77:11158–69. [PMC free article] [PubMed] [Google Scholar]
142. Zhao ZS, Granucci F, Yeh L, Schaffer PA, Cantor H. Molecular mimicry by herpes simplex virus-type 1: autoimmune disease after viral infection. Science. 1998;279:1344–7. [PubMed] [Google Scholar]
143. Panoutsakopoulou V, Sanchirico ME, Huster KM, et al. Analysis of the relationship between viral infection and autoimmune disease. Immun. 2001;15:137–47. [PubMed] [Google Scholar]
144. Verjans GM, Remeijer L, Mooy CM, Osterhaus AD. Herpes simplex virus-specific T cells infiltrate the cornea of patients with herpetic stromal keratitis: no evidence for autoreactive T cells. Invest Ophthalmol Vis Sci. 2000;41:2607–12. [PubMed] [Google Scholar]
145. Koelle DM, Reymond SN, Chen H, et al. Tegument-specific, virus-reactive CD4 T cells localize to the cornea in herpes simplex virus interstitial keratitis in humans. J Virol. 2000;74:10930–8. [PMC free article] [PubMed] [Google Scholar]
146. Babu JS, Thomas J, Kanangat S, Morrison LA, Knipe DM, Rouse BT. Viral replication is required for induction of ocular immunopathology by herpes simplex virus. J Virol. 1996;70:101–7. [PMC free article] [PubMed] [Google Scholar]
147. Durrani OM, Meads CA, Murray PI. Uveitis: a potentially blinding disease. Ophthalmologica. 2004;218:223–36. [PubMed] [Google Scholar]
148. Martin TM, Kurz DE, Rosenbaum JT. Genetics of uveitis. Ophthalmol Clin North Am. 2003;16:555–65. [PubMed] [Google Scholar]
149. de Smet MD, Yamamoto JH, Mochizuki M, et al. Cellular immune responses of patients with uveitis to retinal antigens and their fragments. Am J Ophthalmol. 1990;110:135–42. [PubMed] [Google Scholar]
150. Doekes G, van der Gaag R, Rothova A, et al. Humoral and cellular immune responsiveness to human S-antigen in uveitis. Curr Eye Res. 1987;6:909–19. [PubMed] [Google Scholar]
151. Singh VK, Yamaki K, Abe T, Shinohara T. Molecular mimicry between uveitopathogenic site of retinal S-antigen and Escherichia coli protein: induction of experimental autoimmune uveitis and lymphocyte cross-reaction. Cell Immunol. 1989;122:262–73. [PubMed] [Google Scholar]
152. Singh VK, Kalra HK, Yamaki K, Abe T, Donoso LA, Shinohara T. Molecular mimicry between a uveitopathogenic site of S-antigen and viral peptides. J Immunol. 1990;144:1282–7. Induction of experimental autoimmune uveitis in Lewis rats. [PubMed] [Google Scholar]
153. Wildner G, Diedrichs-Mohring M. Autoimmune uveitis induced by molecular mimicry of peptides from rotavirus, bovine casein and retinal S-antigen. Eur J Immunol. 2003;33:2577–87. [PubMed] [Google Scholar]
154. Lashkevich VA, Koroleva GA, Lukashev AN, Denisova EV, Katargina LA. Enterovirus uveitis. Rev Med Virol. 2004;14:241–54. [PubMed] [Google Scholar]
155. Kaprio J, Tuomilehto J, Koskenvuo M, et al. Concordance for type 1 (insulin-dependent) and type 2 (non-insulin-dependent) diabetes mellitus in a population-based cohort of twins in Finland. Diabetologia. 1992;35:1060–7. [PubMed] [Google Scholar]
156. Kyvik KO, Green A, Beck-Nielsen H. Concordance rates of insulin dependent diabetes mellitus: a population based study of young Danish twins. BMJ. 1995;311:913–7. [PMC free article] [PubMed] [Google Scholar]
157. Jun HS, Yoon JW. A new look at viruses in type 1 diabetes. Diabetes Metab Res Rev. 2003;19:8–31. [PubMed] [Google Scholar]
158. McIntosh ED, Menser MA. A fifty-year follow-up of congenital rubella. Lancet. 1992;340:414–5. [PubMed] [Google Scholar]
159. Ginsberg-Fellner F, Witt ME, Yagihashi S, et al. Congenital rubella syndrome as a model for type 1 (insulin-dependent) diabetes mellitus: increased prevalence of islet cell surface antibodies. Diabetologia. 1984;27(Suppl.):87–9. [PubMed] [Google Scholar]
160. Pak CY, Eun HM, McArthur RG, Yoon JW. Association of cytomegalovirus infection with autoimmune type 1 diabetes. Lancet. 1988;2:1–4. [PubMed] [Google Scholar]
161. Pak CY, Cha CY, Rajotte RV, McArthur RG, Yoon JW. Human pancreatic islet cell specific 38 kilodalton autoantigen identified by cytomegalovirus-induced monoclonal islet cell autoantibody. Diabetologia. 1990;33:569–72. [PubMed] [Google Scholar]
163. Helmke K, Otten A, Willems W. Islet cell antibodies in children with mumps infection. Lancet. 1980;2:211–2. [PubMed] [Google Scholar]
164. Frisk G, Fohlman J, Kobbah M, et al. High frequency of Coxsackie-B-virus-specific IgM in children developing type I diabetes during a period of high diabetes morbidity. J Med Virol. 1985;17:219–27. [PubMed] [Google Scholar]
165. Hyoty H, Hiltunen M, Knip M, et al. A prospective study of the role of coxsackie B and other enterovirus infections in the pathogenesis of IDDM. Diabetes. 1995;44:652–7. Childhood Diabetes in Finland (DiMe) Study Group. [PubMed] [Google Scholar]
166. Schloot NC, Willemen SJ, Duinkerken G, Drijfhout JW, de Vries RR, Roep BO. Molecular mimicry in type 1 diabetes mellitus revisited: T-cell clones to GAD65 peptides with sequence homology to Coxsackie or proinsulin peptides do not crossreact with homologous counterpart. Hum Immunol. 2001;62:299–309. [PubMed] [Google Scholar]
167. Sarugeri E, Dozio N, Meschi F, Pastore MR, Bonifacio E. T cell responses to type 1 diabetes related peptides sharing homologous regions. J Mol Med. 2001;79:213–20. [PubMed] [Google Scholar]
168. Ou D, Mitchell LA, Metzger DL, Gillam S, Tingle AJ. Cross-reactive rubella virus and glutamic acid decarboxylase (65 and 67) protein determinants recognised by T cells of patients with type I diabetes mellitus. Diabetologia. 2000;43:750–62. [PubMed] [Google Scholar]
169. Karounos DG, Wolinsky JS, Thomas JW. Monoclonal antibody to rubella virus capsid protein recognizes a beta-cell antigen. J Immunol. 1993;150:3080–5. [PubMed] [Google Scholar]
170. Hiemstra HS, Schloot NC, van Veelen PA, et al. Cytomegalovirus in autoimmunity: T cell crossreactivity to viral antigen and autoantigen glutamic acid decarboxylase. Proc Natl Acad Sci USA. 2001;98:3988–91. [PMC free article] [PubMed] [Google Scholar]
172. Prince GA, Jenson AB, Billups LC, Notkins AL. Infection of human pancreatic beta cell cultures with mumps virus. Nature. 1978;271:158–61. [PubMed] [Google Scholar]
173. Parkkonen P, Hyoty H, Koskinen L, Leinikki P. Mumps virus infects beta cells in human fetal islet cell cultures upregulating the expression of HLA class I molecules. Diabetologia. 1992;35:63–9. [PubMed] [Google Scholar]
174. Vuorinen T, Nikolakaros G, Simell O, Hyypia T, Vainionpaa R. Mumps and Coxsackie B3 virus infection of human fetal pancreatic islet-like cell clusters. Pancreas. 1992;7:460–4. [PubMed] [Google Scholar]
175. Tisch R, Yang XD, Singer SM, Liblau RS, Fugger L, McDevitt HO. Immune response to glutamic acid decarboxylase correlates with insulitis in non-obese diabetic mice. Nature. 1993;366:72–5. [PubMed] [Google Scholar]
176. Tian J, Lehmann PV, Kaufman DL. T cell cross-reactivity between coxsackievirus and glutamate decarboxylase is associated with a murine diabetes susceptibility allele. J Exp Med. 1994;180:1979–84. [PMC free article] [PubMed] [Google Scholar]
177. Lawson CM. Evidence for mimicry by viral antigens in animal models of autoimmune disease including myocarditis. Cell Mol Life Sci. 2000;57:552–60. [PubMed] [Google Scholar]
178. Serreze DV, Ottendorfer EW, Ellis TM, Gauntt CJ, Atkinson MA. Acceleration of type 1 diabetes by a coxsackievirus infection requires a preexisting critical mass of autoreactive T-cells in pancreatic islets. Diabetes. 2000;49:708–11. [PubMed] [Google Scholar]
179. Horwitz MS, Ilic A, Fine C, Rodriguez E, Sarvetnick N. Presented antigen from damaged pancreatic beta cells activates autoreactive T cells in virus-mediated autoimmune diabetes. J Clin Invest. 2002;109:79–87. [PMC free article] [PubMed] [Google Scholar]
180. Guberski DL, Thomas VA, Shek WR, et al. Induction of type I diabetes by Kilham’s rat virus in diabetes-resistant BB/Wor rats. Science. 1991;254:1010–3. [PubMed] [Google Scholar]
181. Chung YH, Jun HS, Kang Y, et al. Role of macrophages and macrophage-derived cytokines in the pathogenesis of Kilham rat virus-induced autoimmune diabetes in diabetes-resistant BioBreeding rats. J Immunol. 1997;159:466–71. [PubMed] [Google Scholar]
182. Ellerman KE, Richards CA, Guberski DL, Shek WR, Like AA. Kilham rat triggers T-cell-dependent autoimmune diabetes in multiple strains of rat. Diabetes. 1996;45:557–62. [PubMed] [Google Scholar]
183. Yoon JW, London WT, Curfman BL, Brown RL, Notkins AL. Coxsackie virus B4 produces transient diabetes in nonhuman primates. Diabetes. 1986;35:712–6. [PubMed] [Google Scholar]
184. Willison HJ, Yuki N. Peripheral neuropathies and anti-glycolipid antibodies. Brain. 2002;125:2591–625. [PubMed] [Google Scholar]
185. Ilyas AA, Willison HJ, Quarles RH, et al. Serum antibodies to gangliosides in Guillain–Barré syndrome. Ann Neurol. 1988;23:440–7. [PubMed] [Google Scholar]
186. Joseph SA, Tsao CY. Guillain–Barré syndrome. Adolesc Med. 2002;13:487–94. [PubMed] [Google Scholar]
187. Aspinall GO, Fujimoto S, McDonald AG, Pang H, Kurjanczyk LA, Penner JL. Lipopolysaccharides from Campylobacter jejuni associated with Guillain–Barré syndrome patients mimic human gangliosides in structure. Infect Immun. 1994;62:2122–5. [PMC free article] [PubMed] [Google Scholar]
188. Yuki N, Taki T, Takahashi M, et al. Penner’s serotype 4 of Campylobacter jejuni has a lipopolysaccharide that bears a GM1 ganglioside epitope as well as one that bears a GD1 a epitope. Infect Immun. 1994;62:2101–3. [PMC free article] [PubMed] [Google Scholar]
189. Ang CW, Endtz HP, Jacobs BC, et al. Campylobacter jejuni lipopolysaccharides from Guillain–Barré syndrome patients induce IgG anti-GM1 antibodies in rabbits. J Neuroimmunol. 2000;104:133–8. [PubMed] [Google Scholar]
190. Goodyear CS, O’Hanlon GM, Plomp JJ, et al. Monoclonal antibodies raised against Guillain–Barré syndrome-associated Campylobacter jejuni lipopolysaccharides react with neuronal gangliosides and paralyze muscle-nerve preparations. J Clin Invest. 1999;104:697–708. [PMC free article] [PubMed] [Google Scholar]
191. Wirguin I, Briani C, Suturkova-Milosevic L, et al. Induction of anti-GM1 ganglioside antibodies by Campylobacter jejuni lipopolysaccharides. J Neuroimmunol. 1997;78:138–42. [PubMed] [Google Scholar]
192. Ang CW, Laman JD, Willison HJ, et al. Structure of Campylobacter jejuni lipopolysaccharides determines antiganglioside specificity and clinical features of Guillain–Barré and Miller Fisher patients. Infect Immun. 2002;70:1202–8. [PMC free article] [PubMed] [Google Scholar]
193. Nachamkin I, Liu J, Li M, et al. Campylobacter jejuni from patients with Guillain–Barré syndrome preferentially expresses a GD(1a)-like epitope. Infect Immun. 2002;70:5299–303. [PMC free article] [PubMed] [Google Scholar]
194. Yuki N, Susuki K, Koga M, et al. Carbohydrate mimicry between human ganglioside GM1 and Campylobacter jejuni lipooligosaccharide causes Guillain–Barré syndrome. Proc Natl Acad Sci USA. 2004;101:11404–9. [PMC free article] [PubMed] [Google Scholar]
195. Kusunoki S, Shiina M, Kanazawa I. Anti-Gal-C antibodies in GBS subsequent to mycoplasma infection: evidence of molecular mimicry. Neurology. 2001;57:736–8. [PubMed] [Google Scholar]
196. Ang CW, Tio-Gillen AP, Groen J, et al. Cross-reactive anti-galactocerebroside antibodies and Mycoplasma pneumoniae infections in Guillain–Barré syndrome. J Neuroimmunol. 2002;130:179–83. [PubMed] [Google Scholar]
197. Susuki K, Odaka M, Mori M, Hirata K, Yuki N. Acute motor axonal neuropathy after Mycoplasma infection: evidence of molecular mimicry. Neurology. 2004;62:949–56. [PubMed] [Google Scholar]
198. Koga M, Yuki N, Tai T, Hirata K. Miller Fisher syndrome and Haemophilus influenzae infection. Neurology. 2001;57:686–91. [PubMed] [Google Scholar]
199. Mori M, Kuwabara S, Miyake M, et al. Haemophilus influenzae infection and Guillain–Barré syndrome. Brain. 2000;123:2171–8. [PubMed] [Google Scholar]
200. Mori M, Kuwabara S, Miyake M, et al. Haemophilus influenzae has a GM1 ganglioside-like structure and elicits Guillain–Barré syndrome. Neurology. 1999;52:1282–4. [PubMed] [Google Scholar]
201. Steinman L. Multiple sclerosis: a coordinated immunological attack against myelin in the central nervous system. Cell. 1996;85:299–302. [PubMed] [Google Scholar]
202. Sun JB, Olsson T, Wang WZ, et al. Autoreactive T and B cells responding to myelin proteolipid protein in multiple sclerosis and controls. Eur J Immunol. 1991;21:1461–8. [PubMed] [Google Scholar]
203. Ota K, Matsui M, Milford EL, Mackin GA, Weiner HL, Hafler DA. T-cell recognition of an immunodominant myelin basic protein epitope in multiple sclerosis. Nature. 1990;346:183–7. [PubMed] [Google Scholar]
204. Bernard CC, de Rosbo NK. Immunopathological recognition of autoantigens in multiple sclerosis. Acta Neurol (Napoli) 1991;13:171–8. [PubMed] [Google Scholar]
205. Olson JK, Croxford JL, Miller SD. Virus-induced autoimmunity: potential role of viruses in initiation, perpetuation, and progression of T-cell-mediated autoimmune disease. Viral Immunol. 2001;14:227–50. [PubMed] [Google Scholar]
206. Fujinami RS. Can virus infections trigger autoimmune disease? J Autoimmun. 2001;16:229–34. [PubMed] [Google Scholar]
207. Challoner PB, Smith KT, Parker JD, et al. Plaque-associated expression of human herpesvirus 6 in multiple sclerosis. Proc Natl Acad Sci USA. 1995;92:7440–4. [PMC free article] [PubMed] [Google Scholar]
208. Sriram S, Stratton CW, Yao S, et al. Chlamydia pneumoniae infection of the central nervous system in multiple sclerosis. Ann Neurol. 1999;46:6–14. [PubMed] [Google Scholar]
209. Zhang J, Raus J. Myelin basic protein-reactive T cells in multiple sclerosis: pathologic relevance and therapeutic targeting. Cytotechnology. 1994;16:181–7. [PubMed] [Google Scholar]
210. Wucherpfennig KW, Zhang J, Witek C, et al. Clonal expansion and persistence of human T cells specific for an immunodominant myelin basic protein peptide. J Immunol. 1994;152:5581–92. [PubMed] [Google Scholar]
211. Allegretta M, Nicklas JA, Sriram S, Albertini RJ. T cells responsive to myelin basic protein in patients with multiple sclerosis. Science. 1990;247:718–21. [PubMed] [Google Scholar]
212. Wucherpfennig KW, Strominger JL. Molecular mimicry in T cell-mediated autoimmunity: viral peptides activate human T cell clones specific for myelin basic protein. Cell. 1995;80:695–705. [PMC free article] [PubMed] [Google Scholar]
214. Kurtzke JF. Multiple sclerosis in time and space – geographic clues to cause. J Neurovirol. 2000;6(Suppl. 2):S134–40. [PubMed] [Google Scholar]
215. Kurtzke JF, Gudmundsson KR, Bergmann S. Multiple sclerosis in Iceland: 1. Evidence of a postwar epidemic. Neurology. 1982;32:143–50. [PubMed] [Google Scholar]
216. Kurtzke JF, Hyllested K. Multiple sclerosis in the Faroe Islands: I. Clinical and epidemiological features. Ann Neurol. 1979;5:6–21. [PubMed] [Google Scholar]
217. Islam T, Gauderman WJ, Cozen W, Hamilton AS, Burnett ME, Mack TM. Differential twin concordance for multiple sclerosis by latitude of birthplace. Ann Neurol. 2006;60:56–64. [PubMed] [Google Scholar]
218. Oleszak EL, Chang JR, Friedman H, Katsetos CD, Platsoucas CD. Theiler’s virus infection: a model for multiple sclerosis. Clin Microbiol Rev. 2004;17:174–207. [PMC free article] [PubMed] [Google Scholar]
220. Dal Canto MC, Lipton HL. Ultrastructural immunohistochemical localization of virus in acute and chronic demyelinating Theiler’s virus infection. Am J Pathol. 1982;106:20–9. [PMC free article] [PubMed] [Google Scholar]
221. Lipton HL, Twaddle G, Jelachich ML. The predominant virus antigen burden is present in macrophages in Theiler’s murine encephalomyelitis virus-induced demyelinating disease. J Virol. 1995;69:2525–33. [PMC free article] [PubMed] [Google Scholar]
222. Trottier M, Kallio P, Wang W, Lipton HL. High numbers of viral RNA copies in the central nervous system of mice during persistent infection with Theiler’s virus. J Virol. 2001;75:7420–8. [PMC free article] [PubMed] [Google Scholar]
223. Barbano RL, Dal Canto MC. Serum and cells from Theiler’s virus-infected mice fail to injure myelinating cultures or to produce in vivo transfer of disease. J Neurol Sci. 1984;66:283–93. The pathogenesis of Theiler’s virus-induced demyelination appears to differ from that of EAE. [PubMed] [Google Scholar]
224. Miller SD, Clatch RJ, Pevear DC, Trotter JL, Lipton HL. Class II-restricted T cell responses in Theiler’s murine encephalomyelitis virus (TMEV)-induced demyelinating disease. J Immunol. 1987;138:3776–84. I. Cross-specificity among TMEV substrains and related picornaviruses, but not myelin proteins. [PubMed] [Google Scholar]
225. Clatch RJ, Lipton HL, Miller SD. Characterization of Theiler’s murine encephalomyelitis virus (TMEV)-specific delayed-type hypersensitivity responses in TMEV-induced demyelinating disease: correlation with clinical signs. J Immunol. 1986;136:920–7. [PubMed] [Google Scholar]
226. Miller SD, Vanderlugt CL, Begolka WS, et al. Persistent infection with Theiler’s virus leads to CNS autoimmunity via epitope spreading. Nat Med. 1997;3:1133–6. [PubMed] [Google Scholar]
227. Katz-Levy Y, Neville KL, Padilla J, et al. Temporal development of autoreactive Th1 responses and endogenous presentation of self myelin epitopes by central nervous system-resident APCs in Theiler’s virus-infected mice. J Immunol. 2000;165:5304–14. [PubMed] [Google Scholar]
228. Lavi E, Gilden DH, Wroblewska Z, Rorke LB, Weiss SR. Experimental demyelination produced by the A59 strain of mouse hepatitis virus. Neurology. 1984;34:597–603. [PubMed] [Google Scholar]
229. Knobler RL, Lampert PW, Oldstone MB. Virus persistence and recurring demyelination produced by a temperature-sensitive mutant of MHV-4. Nature. 1982;298:279–80. [PMC free article] [PubMed] [Google Scholar]
230. Wang FI, Hinton DR, Gilmore W, Trousdale MD, Fleming JO. Sequential infection of glial cells by the murine hepatitis virus JHM strain (MHV-4) leads to a characteristic distribution of demyelination. Lab Invest. 1992;66:744–54. [PubMed] [Google Scholar]
231. Takahashi K, Goto N, Matsubara Y, Fujiwara K. Postinflammatory remyelination in the spinal cord of mice infected with mouse hepatitis virus, JHM strain. Jpn J Exp Med. 1987;57:145–51. [PubMed] [Google Scholar]
232. Erlich SS, Fleming JO, Stohlman SA, Weiner LP. Experimental neuropathology of chronic demyelination induced by a JHM virus variant (DS) Arch Neurol. 1987;44:839–42. [PubMed] [Google Scholar]
234. Wu GF, Perlman S. Macrophage infiltration, but not apoptosis, is correlated with immune-mediated demyelination following murine infection with a neurotropic coronavirus. J Virol. 1999;73:8771–80. [PMC free article] [PubMed] [Google Scholar]
235. Glass WG, Liu MT, Kuziel WA, Lane TE. Reduced macrophage infiltration and demyelination in mice lacking the chemokine receptor CCR5 following infection with a neurotropic coronavirus. Virology. 2001;288:8–17. [PMC free article] [PubMed] [Google Scholar]
236. Wu GF, Dandekar AA, Pewe L, Perlman S. CD4 and CD8 T cells have redundant but not identical roles in virus-induced demyelination. J Immunol. 2000;165:2278–86. [PubMed] [Google Scholar]
237. Lane TE, Liu MT, Chen BP, et al. A central role for CD4(+) T cells and RANTES in virus-induced central nervous system inflammation and demyelination. J Virol. 2000;74:1415–24. [PMC free article] [PubMed] [Google Scholar]
238. Fazakerley JK, Pathak S, Scallan M, Amor S, Dyson H. Replication of the A7(74) strain of Semliki Forest virus is restricted in neurons. Virology. 1993;195(7):627–37. [PubMed] [Google Scholar]
239. Balluz IM, Glasgow GM, Killen HM, Mabruk MJ, Sheahan BJ, Atkins GJ. Virulent and avirulent strains of Semliki Forest virus show similar cell tropism for the murine central nervous system but differ in the severity and rate of induction of cytolytic damage. Neuropathol Appl Neurobiol. 1993;19:233–9. [PubMed] [Google Scholar]
240. Amor S, Scallan MF, Morris MM, Dyson H, Fazakerley JK. Role of immune responses in protection and pathogenesis during Semliki Forest virus encephalitis. J Gen Virol. 1996;77:281–91. [PubMed] [Google Scholar]
241. Mokhtarian F, Swoveland P. Predisposition to EAE induction in resistant mice by prior infection with Semliki Forest virus. J Immunol. 1987;138:3264–8. [PubMed] [Google Scholar]
242. Fazakerley JK, Amor S, Webb HE. Reconstitution of Semliki forest virus infected mice, induces immune mediated pathological changes in the CNS. Clin Exp Immunol. 1983;52:115–20. [PMC free article] [PubMed] [Google Scholar]
243. Subak-Sharpe I, Dyson H, Fazakerley J. In vivo depletion of CD8+ T cells prevents lesions of demyelination in Semliki Forest virus infection. J Virol. 1993;67:7629–33. [PMC free article] [PubMed] [Google Scholar]
244. Keogh B, Atkins GJ, Mills KH, Sheahan BJ. Avirulent Semliki Forest virus replication and pathology in the central nervous system is enhanced in IL-12-defective and reduced in IL-4-defective mice: a role for Th1 cells in the protective immunity. J Neuroimmunol. 2002;125:15–22. [PubMed] [Google Scholar]
245. Keogh B, Atkins GJ, Mills KH, Sheahan BJ. Role of interferon-gamma and nitric oxide in the neuropathogenesis of avirulent Semliki Forest virus infection. Neuropathol Appl Neurobiol. 2003;29:553–62. [PubMed] [Google Scholar]
246. Mokhtarian F, Shi Y, Zhu PF, Grob D. Immune responses, and autoimmune outcome, during virus infection of the central nervous system. Cell Immunol. 1994;157:195–210. [PubMed] [Google Scholar]
247. Smith-Norowitz TA, Sobel RA, Mokhtarian F. B cells and antibodies in the pathogenesis of myelin injury in Semliki Forest virus encephalomyelitis. Cell Immunol. 2000;200:27–35. [PubMed] [Google Scholar]
248. Mokhtarian F, Zhang Z, Shi Y, Gonzales E, Sobel RA. Molecular mimicry between a viral peptide and a myelin oligodendrocyte glycoprotein peptide induces autoimmune demyelinating disease in mice. J Neuroimmunol. 1999;95:43–54. [PubMed] [Google Scholar]