Skip to main content

Advertisement

Log in

Environmental Basis of Autoimmunity

  • Published:
Clinical Reviews in Allergy & Immunology Aims and scope Submit manuscript

Abstract

The three common themes that underlie the induction and perpetuation of autoimmunity are genetic predisposition, environmental factors, and immune regulation. Environmental factors have gained much attention for their role in triggering autoimmunity, with increasing evidence of their influence as demonstrated by epidemiological studies, laboratory research, and animal studies. Environmental factors known to trigger and perpetuate autoimmunity include infections, gut microbiota, as well as physical and environmental agents. To address these issues, we will review major potential mechanisms that underlie autoimmunity including molecular mimicry, epitope spreading, bystander activation, polyclonal activation of B and T cells, infections, and autoinflammatory activation of innate immunity. The association of the gut microbiota on autoimmunity will be particularly highlighted by their interaction with pharmaceutical agents that may lead to organ-specific autoimmunity. Nonetheless, and we will emphasize this point, the precise mechanism of environmental influence on disease pathogenesis remains elusive.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Abbreviations

AIDs:

Autoinflammatory diseases

AIH:

Autoimmune hepatitis

APCs:

Antigen-presenting cells

ASGPR:

Asialo glycoprotein receptor

DIA:

Drug-induced autoimmunity

EBV:

Epstein-Barr Virus

HAV:

Hepatitis A virus

HCV:

Hepatitis C Virus

HLA:

Human leukocyte antigen

IFN:

Interferon

IUL:

Interleukin

LSE:

Systemic lupus erythematosus

NLRs:

Nucleotide-binding oligomerization domain-like receptors

NLRC4:

NLR family CARD domain-containing protein 4

Oss:

Organic solvents

PAMPs:

Pathogen-associated molecular patterns

PBC:

Primary biliary cirrhosis

PDC:

Pyruvate dehydrogenase complex

PRRs:

Pattern recognition receptor

RA:

Rheumatoid arthritis

TACE:

TNF-a-converting enzyme

TLRs:

Toll-like receptors

TNF:

Tumor necrosis factor

References

  1. Ermann J, Fathman CG (2001) Autoimmune diseases: genes, bugs and failed regulation. Nat Immunol 2:759–761

    Article  CAS  PubMed  Google Scholar 

  2. Cortes JR, Sanchez-Diaz R, Bovolenta ER et al (2014) Maintenance of immune tolerance by Foxp3+ regulatory T cells requires CD69 expression. J Autoimmun 55:51–62

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Berrih-Aknin S (2014) Myasthenia gravis: paradox versus paradigm in autoimmunity. J Autoimmun 52:1–28

    Article  CAS  PubMed  Google Scholar 

  4. Bao Y, Cao X (2014) The immune potential and immunopathology of cytokine-producing B cell subsets: a comprehensive review. J Autoimmun 55:10–23

    Article  CAS  PubMed  Google Scholar 

  5. Vojdani A, Pollard KM, Campbell AW (2014) Environmental triggers and autoimmunity. Autoimmun Dis 2014:798029

    Google Scholar 

  6. Pollard KM (2015) Environment, autoantibodies, and autoimmunity. Front Immunol 6:60

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Berrih-Aknin S, Frenkian-Cuvelier M, Eymard B (2014) Diagnostic and clinical classification of autoimmune myasthenia gravis. J Autoimmun 48–49:143–148

    Article  PubMed  CAS  Google Scholar 

  8. Karussis D (2014) The diagnosis of multiple sclerosis and the various related demyelinating syndromes: a critical review. J Autoimmun 48–49:134–142

    Article  PubMed  CAS  Google Scholar 

  9. Mosca M, Tani C, Vagnani S, Carli L, Bombardieri S (2014) The diagnosis and classification of undifferentiated connective tissue diseases. J Autoimmun 48–49:50–52

    Article  PubMed  Google Scholar 

  10. Mouthon L, Dunogue B, Guillevin L (2014) Diagnosis and classification of eosinophilic granulomatosis with polyangiitis (formerly named Churg-Strauss syndrome). J Autoimmun 48–49:99–103

    Article  PubMed  CAS  Google Scholar 

  11. Hajj-Ali RA, Calabrese LH (2014) Diagnosis and classification of central nervous system vasculitis. J Autoimmun 48–49:149–152

    Article  PubMed  Google Scholar 

  12. Kivity S, Arango MT, Ehrenfeld M et al (2014) Infection and autoimmunity in Sjogren’s syndrome: a clinical study and comprehensive review. J Autoimmun 51:17–22

    Article  CAS  PubMed  Google Scholar 

  13. Selmi C, Leung PS, Sherr DH et al (2012) Mechanisms of environmental influence on human autoimmunity: a National Institute of Environmental Health Sciences expert panel workshop. J Autoimmun 39:272–284

    Article  PubMed  Google Scholar 

  14. Brouqui P, Raoult D, Conte-Devolx B (1991) Coxsackie thyroiditis. Ann Intern Med 114:1063–1064

    Article  CAS  PubMed  Google Scholar 

  15. Fernandez-Soto L, Gonzalez A, Escobar-Jimenez F et al (1998) Increased risk of autoimmune thyroid disease in hepatitis C vs hepatitis B before, during, and after discontinuing interferon therapy. Arch Intern Med 158:1445–1448

    Article  CAS  PubMed  Google Scholar 

  16. Hwang YC, Jeong IK, Chon S et al (2010) Fulminant type 1 diabetes mellitus associated with acute hepatitis A. Diabet Med 27:366–367

    Article  PubMed  Google Scholar 

  17. Kawai H, Inui T, Kashiwagi S et al (1992) HTLV-I infection in patients with autoimmune thyroiditis (Hashimoto’s thyroiditis). J Med Virol 38:138–141

    Article  CAS  PubMed  Google Scholar 

  18. Kerr JR, Cartron JP, Curran MD, Moore JE, Elliott JR, Mollan RA (1995) A study of the role of parvovirus B19 in rheumatoid arthritis. Br J Rheumatol 34:809–813

    Article  CAS  PubMed  Google Scholar 

  19. Leite JL, Bufalo NE, Santos RB, Romaldini JH, Ward LS (2010) Herpesvirus type 7 infection may play an important role in individuals with a genetic profile of susceptibility to Graves’ disease. Eur J Endocrinol 162:315–321

    Article  CAS  PubMed  Google Scholar 

  20. Mason AL (2011) The evidence supports a viral aetiology for primary biliary cirrhosis. J Hepatol 54:1312–1314

    Article  PubMed  Google Scholar 

  21. Mori K, Munakata Y, Saito T et al (2007) Intrathyroidal persistence of human parvovirus B19 DNA in a patient with Hashimoto’s thyroiditis. J Infect 55:e29–e31

    Article  PubMed  Google Scholar 

  22. Parmar RC, Bavdekar SB, Sahu DR, Warke S, Kamat JR (2001) Thyroiditis as a presenting feature of mumps. Pediatr Infect Dis J 20:637–638

    Article  CAS  PubMed  Google Scholar 

  23. Pratesi F, Tommasi C, Anzilotti C, Chimenti D, Migliorini P (2006) Deiminated Epstein-Barr virus nuclear antigen 1 is a target of anti-citrullinated protein antibodies in rheumatoid arthritis. Arthritis Rheum 54:733–741

    Article  CAS  PubMed  Google Scholar 

  24. Shimon I, Pariente C, Shlomo-David J, Grossman Z, Sack J (2003) Transient elevation of triiodothyronine caused by triiodothyronine autoantibody associated with acute Epstein-Barr-virus infection. Thyroid 13:211–215

    Article  CAS  PubMed  Google Scholar 

  25. Vento S, Cainelli F (2004) Is there a role for viruses in triggering autoimmune hepatitis? Autoimmun Rev 3:61–69

    Article  PubMed  Google Scholar 

  26. Vento S, Guella L, Mirandola F et al (1995) Epstein-Barr virus as a trigger for autoimmune hepatitis in susceptible individuals. Lancet 346:608–609

    Article  CAS  PubMed  Google Scholar 

  27. Ziring PR, Gallo G, Finegold M, Buimovici-Klein E, Ogra P (1977) Chronic lymphocytic thyroiditis: identification of rubella virus antigen in the thyroid of a child with congenital rubella. J Pediatr 90:419–420

    Article  CAS  PubMed  Google Scholar 

  28. Farquharson D, Butcher JP, Culshaw S (2012) Periodontitis, porphyromonas, and the pathogenesis of rheumatoid arthritis. Mucosal Immunol 5:112–120

    Article  CAS  PubMed  Google Scholar 

  29. Wu HJ, Ivanov II, Darce J et al (2010) Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32:815–827

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bech K, Clemmensen O, Larsen JH, Thyme S, Bendixen G (1978) Cell-mediated immunity of Yersinia enterocolitica serotype 3 in patients with thyroid diseases. Allergy 33:82–88

    Article  CAS  PubMed  Google Scholar 

  31. Hill Gaston JS, Lillicrap MS (2003) Arthritis associated with enteric infection. Best Pract Res Clin Rheumatol 17:219–239

    Article  CAS  PubMed  Google Scholar 

  32. McColl GJ, Diviney MB, Holdsworth RF et al (2000) HLA-B27 expression and reactive arthritis susceptibility in two patient cohorts infected with Salmonella Typhimurium. Aust N Z J Med 30:28–32

    Article  CAS  PubMed  Google Scholar 

  33. Hannu T, Mattila L, Siitonen A, Leirisalo-Repo M (2005) Reactive arthritis attributable to Shigella infection: a clinical and epidemiological nationwide study. Ann Rheum Dis 64:594–598

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ebringer A, Rashid T (2006) Rheumatoid arthritis is an autoimmune disease triggered by Proteus urinary tract infection. Clin Dev Immunol 13:41–48

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Pope JE, Krizova A, Garg AX, Thiessen-Philbrook H, Ouimet JM (2007) Campylobacter reactive arthritis: a systematic review. Semin Arthritis Rheum 37:48–55

    Article  PubMed  PubMed Central  Google Scholar 

  36. Dominguez-Lopez ML, Cancino-Diaz ME, Jimenez-Zamudio L, Granados-Arreola J, Burgos-Vargas R, Garcia-Latorre E (2000) Cellular immune response to Klebsiella pneumoniae antigens in patients with HLA-B27+ ankylosing spondylitis. J Rheumatol 27:1453–1460

    CAS  PubMed  Google Scholar 

  37. Cope A, Anderson J, Wilkins E (1992) Clostridium difficile toxin-induced reactive arthritis in a patient with chronic Reiter’s syndrome. Eur J Clin Microbiol Infect Dis 11:40–43

    Article  CAS  PubMed  Google Scholar 

  38. Liu ZQ, Deng GM, Foster S, Tarkowski A (2001) Staphylococcal peptidoglycans induce arthritis. Arthritis Res 3:375–380

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Fae KC, da Silva DD, Oshiro SE et al (2006) Mimicry in recognition of cardiac myosin peptides by heart-intralesional T cell clones from rheumatic heart disease. J Immunol 176:5662–5670

    Article  CAS  PubMed  Google Scholar 

  40. Sutliff WD, Shepard R, Dunham WB (1953) Acute Leptospira pomona arthritis and myocarditis. Ann Intern Med 39:134–140

    Article  CAS  PubMed  Google Scholar 

  41. Carter JD, Espinoza LR, Inman RD et al (2010) Combination antibiotics as a treatment for chronic Chlamydia-induced reactive arthritis: a double-blind, placebo-controlled, prospective trial. Arthritis Rheum 62:1298–1307

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kim HR, Kim EY, Cerny J, Moudgil KD (2006) Antibody responses to mycobacterial and self heat shock protein 65 in autoimmune arthritis: epitope specificity and implication in pathogenesis. J Immunol 177:6634–6641

    Article  CAS  PubMed  Google Scholar 

  43. Imai D, Holden K, Velazquez EM, Feng S, Hodzic E, Barthold SW (2013) Influence of arthritis-related protein (BBF01) on infectivity of Borrelia burgdorferi B31. BMC Microbiol 13:100

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Smyk DS, Bogdanos DP, Kriese S, Billinis C, Burroughs AK, Rigopoulou EI (2012) Urinary tract infection as a risk factor for autoimmune liver disease: from bench to bedside. Clin Res Hepatol Gastroenterol 36:110–121

    Article  PubMed  Google Scholar 

  45. Hermann E, Mayet WJ, Klein O et al (1991) Candida arthritis: cellular immune responses of synovial fluid and peripheral blood lymphocytes to Candida albicans. Ann Rheum Dis 50:697–701

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kivity S, Agmon-Levin N, Blank M, Shoenfeld Y (2009) Infections and autoimmunity—friends or foes? Trends Immunol 30:409–414

    Article  CAS  PubMed  Google Scholar 

  47. Cusick MF, Libbey JE, Fujinami RS (2012) Molecular mimicry as a mechanism of autoimmune disease. Clin Rev Allergy Immunol 42:102–111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Vojdani A (2014) A potential link between environmental triggers and autoimmunity. Autoimmun Dis 2014:437231

    Google Scholar 

  49. Burroughs AK, Butler P, Sternberg MJ, Baum H (1992) Molecular mimicry in liver disease. Nature 358:377–378

    Article  CAS  PubMed  Google Scholar 

  50. Van de Water J, Ishibashi H, Coppel RL, Gershwin ME (2001) Molecular mimicry and primary biliary cirrhosis: premises not promises. Hepatology 33:771–775

    Article  PubMed  Google Scholar 

  51. Wang J, Yang GX, Tsuneyama K, Gershwin ME, Ridgway WM, Leung PS (2014) Animal models of primary biliary cirrhosis. Semin Liver Dis 34:285–296

    Article  CAS  PubMed  Google Scholar 

  52. Wang JJ, Yang GX, Zhang WC et al (2014) Escherichia coli infection induces autoimmune cholangitis and anti-mitochondrial antibodies in non-obese diabetic (NOD).B6 (Idd10/Idd18) mice. Clin Exp Immunol 175:192–201

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kita H, Matsumura S, He XS et al (2002) Quantitative and functional analysis of PDC-E2-specific autoreactive cytotoxic T lymphocytes in primary biliary cirrhosis. J Clin Invest 109:1231–1240

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Matsumura S, Kita H, He XS et al (2002) Comprehensive mapping of HLA-A0201-restricted CD8 T-cell epitopes on PDC-E2 in primary biliary cirrhosis. Hepatology 36:1125–1134

    Article  CAS  PubMed  Google Scholar 

  55. Shigematsu H, Shimoda S, Nakamura M et al (2000) Fine specificity of T cells reactive to human PDC-E2 163–176 peptide, the immunodominant autoantigen in primary biliary cirrhosis: implications for molecular mimicry and cross-recognition among mitochondrial autoantigens. Hepatology 32:901–909

    Article  CAS  PubMed  Google Scholar 

  56. Shimoda S, Nakamura M, Shigematsu H et al (2000) Mimicry peptides of human PDC-E2 163–176 peptide, the immunodominant T-cell epitope of primary biliary cirrhosis. Hepatology 31:1212–1216

    Article  CAS  PubMed  Google Scholar 

  57. Vanderlugt CJ, Miller SD (1996) Epitope spreading. Curr Opin Immunol 8:831–836

    Article  CAS  PubMed  Google Scholar 

  58. Cornaby C, Gibbons L, Mayhew V, Sloan CS, Welling A, Poole BD (2015) B cell epitope spreading: mechanisms and contribution to autoimmune diseases. Immunol Lett 163:56–68

    Article  CAS  PubMed  Google Scholar 

  59. Prasad S, Kohm AP, McMahon JS, Luo X, Miller SD (2012) Pathogenesis of NOD diabetes is initiated by reactivity to the insulin B chain 9–23 epitope and involves functional epitope spreading. J Autoimmun 39:347–353

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Vanderlugt CL, Miller SD (2002) Epitope spreading in immune-mediated diseases: implications for immunotherapy. Nat Rev Immunol 2:85–95

    Article  CAS  PubMed  Google Scholar 

  61. Dhirapong A, Lleo A, Yang GX et al (2011) B cell depletion therapy exacerbates murine primary biliary cirrhosis. Hepatology 53:527–535

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Dhirapong A, Yang GX, Nadler S et al (2013) Therapeutic effect of cytotoxic T lymphocyte antigen 4/immunoglobulin on a murine model of primary biliary cirrhosis. Hepatology 57:708–715

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Getts DR, Chastain EM, Terry RL, Miller SD (2013) Virus infection, antiviral immunity, and autoimmunity. Immunol Rev 255:197–209

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Pane JA, Webster NL, Coulson BS (2014) Rotavirus activates lymphocytes from non-obese diabetic mice by triggering toll-like receptor 7 signaling and interferon production in plasmacytoid dendritic cells. PLoS Pathog 10:e1003998

    Article  PubMed  PubMed Central  Google Scholar 

  65. Nakamoto N, Kanai T (2014) Role of toll-like receptors in immune activation and tolerance in the liver. Front Immunol 5:221

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Agmon-Levin N, Ram M, Barzilai O et al (2009) Prevalence of hepatitis C serum antibody in autoimmune diseases. J Autoimmun 32:261–266

    Article  CAS  PubMed  Google Scholar 

  67. Zen M, Gatto M, Domeneghetti M et al (2013) Clinical guidelines and definitions of autoinflammatory diseases: contrasts and comparisons with autoimmunity—a comprehensive review. Clin Rev Allergy Immunol 45:227–235

    Article  CAS  PubMed  Google Scholar 

  68. Martinon F, Mayor A, Tschopp J (2009) The inflammasomes: guardians of the body. Annu Rev Immunol 27:229–265

    Article  CAS  PubMed  Google Scholar 

  69. Kahlenberg JM, Kaplan MJ (2014) The inflammasome and lupus: another innate immune mechanism contributing to disease pathogenesis? Curr Opin Rheumatol 26:475–481

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Shoenfeld Y, Agmon-Levin N (2011) ‘ASIA’—autoimmune/inflammatory syndrome induced by adjuvants. J Autoimmun 36:4–8

    Article  CAS  PubMed  Google Scholar 

  71. Vieira SM, Pagovich OE, Kriegel MA (2014) Diet, microbiota and autoimmune diseases. Lupus 23:518–526

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Van Praet JT, Donovan E, Vanassche I et al (2015) Commensal microbiota influence systemic autoimmune responses. EMBO J 34:466–474

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Sargent J (2015) Autoimmunity: T1DM and the gut microbiome. Nat Rev Endocrinol 11:193

    PubMed  Google Scholar 

  74. Farrokhi V, Nemati R, Nichols FC et al (2013) Bacterial lipodipeptide, lipid 654, is a microbiome-associated biomarker for multiple sclerosis. Clin Transl Immunol 2:e8

    Article  CAS  Google Scholar 

  75. Brusca SB, Abramson SB, Scher JU (2014) Microbiome and mucosal inflammation as extra-articular triggers for rheumatoid arthritis and autoimmunity. Curr Opin Rheumatol 26:101–107

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Foxman EF, Iwasaki A (2011) Genome-virome interactions: examining the role of common viral infections in complex disease. Nat Rev Microbiol 9:254–264

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Moon C, Stappenbeck TS (2012) Viral interactions with the host and microbiota in the intestine. Curr Opin Immunol 24:405–410

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Velasquez-Manoff M (2015) Gut microbiome: the peacekeepers. Nature 518:S3–S11

    Article  CAS  PubMed  Google Scholar 

  79. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI (2011) Human nutrition, the gut microbiome and the immune system. Nature 474:327–336

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Greer RL, Morgun A, Shulzhenko N (2013) Bridging immunity and lipid metabolism by gut microbiota. J Allergy Clin Immunol 132:253–262, quiz 263

    Article  CAS  PubMed  Google Scholar 

  81. Greiner TU, Hyotylainen T, Knip M, Backhed F, Oresic M (2014) The gut microbiota modulates glycaemic control and serum metabolite profiles in non-obese diabetic mice. PLoS One 9:e110359

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Rogier R, Koenders MI, Abdollahi-Roodsaz S (2015) Toll-like receptor mediated modulation of T cell response by commensal intestinal microbiota as a trigger for autoimmune arthritis. J Immunol Res 2015:527696

    Article  PubMed  PubMed Central  Google Scholar 

  83. Sargent J (2014) Diabetes. Altered gut microbial networks linked to islet cell autoimmunity. Nat Rev Endocrinol 10:313

    Article  PubMed  Google Scholar 

  84. McLean MH, Dieguez D Jr, Miller LM, Young HA (2015) Does the microbiota play a role in the pathogenesis of autoimmune diseases? Gut 64:332–341

    Article  CAS  PubMed  Google Scholar 

  85. Barbhaiya M, Costenbader KH (2014) Ultraviolet radiation and systemic lupus erythematosus. Lupus 23:588–595

    Article  CAS  PubMed  Google Scholar 

  86. Caricchio R, McPhie L, Cohen PL (2003) Ultraviolet B radiation-induced cell death: critical role of ultraviolet dose in inflammation and lupus autoantigen redistribution. J Immunol 171:5778–5786

    Article  CAS  PubMed  Google Scholar 

  87. Coleman DJ, Garcia G, Hyter S et al (2014) Retinoid-X-receptors (alpha/beta) in melanocytes modulate innate immune responses and differentially regulate cell survival following UV irradiation. PLoS Genet 10:e1004321

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Metwally D, Sayed K, Abdel Hay R and Rashed L (2014) Reduction in tissue plasmin: a new mechanism of action of narrowband ultraviolet B in psoriasis. Clin Exp Dermatol

  89. Yang CY, Leung PS, Adamopoulos IE, Gershwin ME (2013) The implication of vitamin D and autoimmunity: a comprehensive review. Clin Rev Allergy Immunol 45:217–226

    Article  CAS  PubMed  Google Scholar 

  90. Grober U, Spitz J, Reichrath J, Kisters K, Holick MF (2013) Vitamin D: update 2013: from rickets prophylaxis to general preventive healthcare. Dermatoendocrinol 5:331–347

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Gorman S, Hart PH (2012) The current state of play of rodent models to study the role of vitamin D in UV-induced immunomodulation. Photochem Photobiol Sci 11:1788–1796

    Article  CAS  PubMed  Google Scholar 

  92. D’Aurizio F, Villalta D, Metus P, Doretto P, Tozzoli R (2015) Is vitamin D a player or not in the pathophysiology of autoimmune thyroid diseases? Autoimmun Rev 14:363–369

    Article  PubMed  CAS  Google Scholar 

  93. Lin WY, Wan L, Tsai CH, Chen RH, Lee CC, Tsai FJ (2006) Vitamin D receptor gene polymorphisms are associated with risk of Hashimoto’s thyroiditis in Chinese patients in Taiwan. J Clin Lab Anal 20:109–112

    Article  CAS  PubMed  Google Scholar 

  94. Stefanic M, Papic S, Suver M, Glavas-Obrovac L, Karner I (2008) Association of vitamin D receptor gene 3′-variants with Hashimoto’s thyroiditis in the Croatian population. Int J Immunogenet 35:125–131

    Article  CAS  PubMed  Google Scholar 

  95. Yazici D, Yavuz D, Tarcin O, Sancak S, Deyneli O, Akalin S (2013) Vitamin D receptor gene ApaI, TaqI, FokI and BsmI polymorphisms in a group of Turkish patients with Hashimoto’s thyroiditis. Minerva Endocrinol 38:195–201

    CAS  PubMed  Google Scholar 

  96. Stefanic M, Karner I, Glavas-Obrovac L et al (2005) Association of vitamin D receptor gene polymorphism with susceptibility to Graves’ disease in Eastern Croatian population: case-control study. Croat Med J 46:639–646

    PubMed  Google Scholar 

  97. Horst-Sikorska W, Ignaszak-Szczepaniak M, Marcinkowska M, Kaczmarek M, Stajgis M, Slomski R (2008) Association analysis of vitamin D receptor gene polymorphisms with bone mineral density in young women with Graves’ disease. Acta Biochim Pol 55:371–380

    CAS  PubMed  Google Scholar 

  98. Inoue N, Watanabe M, Ishido N et al (2014) The functional polymorphisms of VDR, GC and CYP2R1 are involved in the pathogenesis of autoimmune thyroid diseases. Clin Exp Immunol 178:262–269

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Pani MA, Regulla K, Segni M et al (2002) A polymorphism within the vitamin D-binding protein gene is associated with Graves’ disease but not with Hashimoto’s thyroiditis. J Clin Endocrinol Metab 87:2564–2567

    CAS  PubMed  Google Scholar 

  100. Kurylowicz A, Ramos-Lopez E, Bednarczuk T, Badenhoop K (2006) Vitamin D-binding protein (DBP) gene polymorphism is associated with Graves’ disease and the vitamin D status in a Polish population study. Exp Clin Endocrinol Diabetes 114:329–335

    Article  CAS  PubMed  Google Scholar 

  101. Collins JE, Heward JM, Nithiyananthan R et al (2004) Lack of association of the vitamin D receptor gene with Graves’ disease in UK Caucasians. Clin Endocrinol (Oxf) 60:618–624

    Article  CAS  Google Scholar 

  102. Maalej A, Petit-Teixeira E, Chabchoub G et al (2008) Lack of association of VDR gene polymorphisms with thyroid autoimmune disorders: familial and case/control studies. J Clin Immunol 28:21–25

    Article  CAS  PubMed  Google Scholar 

  103. Pani MA, Regulla K, Segni M et al (2002) Vitamin D 1alpha-hydroxylase (CYP1alpha) polymorphism in Graves’ disease, Hashimoto’s thyroiditis and type 1 diabetes mellitus. Eur J Endocrinol 146:777–781

    Article  CAS  PubMed  Google Scholar 

  104. Jaakkola JJ, Gissler M (2005) Maternal smoking in pregnancy as a determinant of rheumatoid arthritis and other inflammatory polyarthropathies during the first 7 years of life. Int J Epidemiol 34:664–671

    Article  PubMed  Google Scholar 

  105. van der Helm-van Mil AH, Verpoort KN, le Cessie S, Huizinga TW, de Vries RR, Toes RE (2007) The HLA-DRB1 shared epitope alleles differ in the interaction with smoking and predisposition to antibodies to cyclic citrullinated peptide. Arthritis Rheum 56:425–432

    Article  PubMed  CAS  Google Scholar 

  106. Sugiyama D, Nishimura K, Tamaki K et al (2010) Impact of smoking as a risk factor for developing rheumatoid arthritis: a meta-analysis of observational studies. Ann Rheum Dis 69:70–81

    Article  CAS  PubMed  Google Scholar 

  107. Arnson Y, Shoenfeld Y, Amital H (2010) Effects of tobacco smoke on immunity, inflammation and autoimmunity. J Autoimmun 34:J258–J265

    Article  CAS  PubMed  Google Scholar 

  108. Conrad K, Mehlhorn J, Luthke K, Dorner T, Frank KH (1996) Systemic lupus erythematosus after heavy exposure to quartz dust in uranium mines: clinical and serological characteristics. Lupus 5:62–69

    CAS  PubMed  Google Scholar 

  109. Costenbader KH, Chang SC, De Vivo I, Plenge R, Karlson EW (2008) Genetic polymorphisms in PTPN22, PADI-4, and CTLA-4 and risk for rheumatoid arthritis in two longitudinal cohort studies: evidence of gene-environment interactions with heavy cigarette smoking. Arthritis Res Ther 10:R52

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Costenbader KH, Kim DJ, Peerzada J et al (2004) Cigarette smoking and the risk of systemic lupus erythematosus: a meta-analysis. Arthritis Rheum 50:849–857

    Article  PubMed  Google Scholar 

  111. Ghaussy NO, Sibbitt W Jr, Bankhurst AD, Qualls CR (2003) Cigarette smoking and disease activity in systemic lupus erythematosus. J Rheumatol 30:1215–1221

    PubMed  Google Scholar 

  112. Hatron PY, Plouvier B, Francois M, Baclet JL, Deconninck P, Devulder B (1982) Association of lupus erythematosus and silicosis. Rev Med Interne 3:245–246

    Article  CAS  PubMed  Google Scholar 

  113. Jones RN, Turner-Warwick M, Ziskind M, Weill H (1976) High prevalence of antinuclear antibodies in sandblasters’ silicosis. Am Rev Respir Dis 113:393–395

    CAS  PubMed  Google Scholar 

  114. Karlson EW, Chang SC, Cui J et al (2010) Gene-environment interaction between HLA-DRB1 shared epitope and heavy cigarette smoking in predicting incident rheumatoid arthritis. Ann Rheum Dis 69:54–60

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Kiyohara C, Washio M, Horiuchi T et al (2009) Cigarette smoking, N-acetyltransferase 2 polymorphisms and systemic lupus erythematosus in a Japanese population. Lupus 18:630–638

    Article  CAS  PubMed  Google Scholar 

  116. Klareskog L, Padyukov L, Alfredsson L (2007) Smoking as a trigger for inflammatory rheumatic diseases. Curr Opin Rheumatol 19:49–54

    Article  CAS  PubMed  Google Scholar 

  117. Klockars M, Koskela RS, Jarvinen E, Kolari PJ, Rossi A (1987) Silica exposure and rheumatoid arthritis: a follow up study of granite workers 1940–81. Br Med J (Clin Res Ed) 294:997–1000

    Article  CAS  Google Scholar 

  118. Kobayashi S, Okamoto H, Iwamoto T et al (2008) A role for the aryl hydrocarbon receptor and the dioxin TCDD in rheumatoid arthritis. Rheumatology (Oxford) 47:1317–1322

    Article  CAS  Google Scholar 

  119. Lee DM, Phillips R, Hagan EM, Chibnik LB, Costenbader KH, Schur PH (2009) Quantifying anti-cyclic citrullinated peptide titres: clinical utility and association with tobacco exposure in patients with rheumatoid arthritis. Ann Rheum Dis 68:201–208

    Article  CAS  PubMed  Google Scholar 

  120. Olsson AR, Skogh T, Axelson O, Wingren G (2004) Occupations and exposures in the work environment as determinants for rheumatoid arthritis. Occup Environ Med 61:233–238

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Rosenman KD, Moore-Fuller M, Reilly MJ (1999) Connective tissue disease and silicosis. Am J Ind Med 35:375–381

    Article  CAS  PubMed  Google Scholar 

  122. Sanchez-Roman J, Wichmann I, Salaberri J, Varela JM, Nunez-Roldan A (1993) Multiple clinical and biological autoimmune manifestations in 50 workers after occupational exposure to silica. Ann Rheum Dis 52:534–538

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Silman AJ, Newman J, MacGregor AJ (1996) Cigarette smoking increases the risk of rheumatoid arthritis. Results from a nationwide study of disease-discordant twins. Arthritis Rheum 39:732–735

    Article  CAS  PubMed  Google Scholar 

  124. Simard JF, Costenbader KH, Liang MH, Karlson EW, Mittleman MA (2009) Exposure to maternal smoking and incident SLE in a prospective cohort study. Lupus 18:431–435

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Smyk D, Mytilinaiou MG, Rigopoulou EI, Bogdanos DP (2010) PBC triggers in water reservoirs, coal mining areas and waste disposal sites: from Newcastle to New York. Dis Markers 29:337–344

    Article  PubMed  PubMed Central  Google Scholar 

  126. Steenland K, Brown D (1995) Mortality study of gold miners exposed to silica and nonasbestiform amphibole minerals: an update with 14 more years of follow-up. Am J Ind Med 27:217–229

    Article  CAS  PubMed  Google Scholar 

  127. Steenland K, Sanderson W, Calvert GM (2001) Kidney disease and arthritis in a cohort study of workers exposed to silica. Epidemiology 12:405–412

    Article  CAS  PubMed  Google Scholar 

  128. Stolt P, Kallberg H, Lundberg I et al (2005) Silica exposure is associated with increased risk of developing rheumatoid arthritis: results from the Swedish EIRA study. Ann Rheum Dis 64:582–586

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Turner S, Cherry N (2000) Rheumatoid arthritis in workers exposed to silica in the pottery industry. Occup Environ Med 57:443–447

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Freemer MM, King TE Jr, Criswell LA (2006) Association of smoking with dsDNA autoantibody production in systemic lupus erythematosus. Ann Rheum Dis 65:581–584

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Selmi C (2012) Cutting-edge issues in autoimmunity and allergy of the digestive system. Clin Rev Allergy Immunol 42:265–268

    Article  CAS  PubMed  Google Scholar 

  132. Somers EC, Richardson BC (2014) Environmental exposures, epigenetic changes and the risk of lupus. Lupus 23:568–576

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Germolec D, Kono DH, Pfau JC, Pollard KM (2012) Animal models used to examine the role of the environment in the development of autoimmune disease: findings from an NIEHS Expert Panel Workshop. J Autoimmun 39:285–293

    Article  PubMed  PubMed Central  Google Scholar 

  134. Gilbert KM, Reisfeld B, Zurlinden TJ, Kreps MN, Erickson SW, Blossom SJ (2014) Modeling toxicodynamic effects of trichloroethylene on liver in mouse model of autoimmune hepatitis. Toxicol Appl Pharmacol 279:284–293

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Lerner A, Matthias T (2015) Changes in intestinal tight junction permeability associated with industrial food additives explain the rising incidence of autoimmune disease. Autoimmun Rev 14:479–489

    Article  CAS  PubMed  Google Scholar 

  136. Gourley M, Miller FW (2007) Mechanisms of disease: environmental factors in the pathogenesis of rheumatic disease. Nat Clin Pract Rheumatol 3:172–180

    Article  CAS  PubMed  Google Scholar 

  137. Barragan-Martinez C, Speck-Hernandez CA, Montoya-Ortiz G, Mantilla RD, Anaya JM, Rojas-Villarraga A (2012) Organic solvents as risk factor for autoimmune diseases: a systematic review and meta-analysis. PLoS One 7:e51506

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Hardy CJ, Palmer BP, Muir KR, Sutton AJ, Powell RJ (1998) Smoking history, alcohol consumption, and systemic lupus erythematosus: a case-control study. Ann Rheum Dis 57:451–455

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Bengtsson AA, Rylander L, Hagmar L, Nived O, Sturfelt G (2002) Risk factors for developing systemic lupus erythematosus: a case-control study in southern Sweden. Rheumatology (Oxford) 41:563–571

    Article  CAS  Google Scholar 

  140. Washio M, Horiuchi T, Kiyohara C et al (2006) Smoking, drinking, sleeping habits, and other lifestyle factors and the risk of systemic lupus erythematosus in Japanese females: findings from the KYSS study. Mod Rheumatol 16:143–150

    Article  PubMed  Google Scholar 

  141. Nagata C, Fujita S, Iwata H et al (1995) Systemic lupus erythematosus: a case-control epidemiologic study in Japan. Int J Dermatol 34:333–337

    Article  CAS  PubMed  Google Scholar 

  142. Ghaussy NO, Sibbitt WL Jr, Qualls CR (2001) Cigarette smoking, alcohol consumption, and the risk of systemic lupus erythematosus: a case-control study. J Rheumatol 28:2449–2453

    CAS  PubMed  Google Scholar 

  143. Formica MK, Palmer JR, Rosenberg L, McAlindon TE (2003) Smoking, alcohol consumption, and risk of systemic lupus erythematosus in the Black Women’s Health Study. J Rheumatol 30:1222–1226

    PubMed  Google Scholar 

  144. Takvorian SU, Merola JF, Costenbader KH (2014) Cigarette smoking, alcohol consumption and risk of systemic lupus erythematosus. Lupus 23:537–544

    Article  CAS  PubMed  Google Scholar 

  145. Wang J, Pan HF, Ye DQ, Su H, Li XP (2008) Moderate alcohol drinking might be protective for systemic lupus erythematosus: a systematic review and meta-analysis. Clin Rheumatol 27:1557–1563

    Article  PubMed  Google Scholar 

  146. Scott IC, Tan R, Stahl D, Steer S, Lewis CM, Cope AP (2013) The protective effect of alcohol on developing rheumatoid arthritis: a systematic review and meta-analysis. Rheumatology (Oxford) 52:856–867

    Article  CAS  Google Scholar 

  147. Adamzik K, McAleer MA, Kirby B (2013) Alcohol and psoriasis: sobering thoughts. Clin Exp Dermatol 38:819–822

    Article  CAS  PubMed  Google Scholar 

  148. Poikolainen K, Reunala T, Karvonen J, Lauharanta J, Karkkainen P (1990) Alcohol intake: a risk factor for psoriasis in young and middle aged men? BMJ 300:780–783

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Qureshi AA, Dominguez PL, Choi HK, Han J, Curhan G (2010) Alcohol intake and risk of incident psoriasis in US women: a prospective study. Arch Dermatol 146:1364–1369

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Poikolainen K, Karvonen J, Pukkala E (1999) Excess mortality related to alcohol and smoking among hospital-treated patients with psoriasis. Arch Dermatol 135:1490–1493

    Article  CAS  PubMed  Google Scholar 

  151. Farkas A, Kemeny L (2010) Psoriasis and alcohol: is cutaneous ethanol one of the missing links? Br J Dermatol 162:711–716

    Article  CAS  PubMed  Google Scholar 

  152. Nath B, Szabo G (2009) Alcohol-induced modulation of signaling pathways in liver parenchymal and nonparenchymal cells: implications for immunity. Semin Liver Dis 29:166–177

    Article  CAS  PubMed  Google Scholar 

  153. Xiao X, Chang C (2014) Diagnosis and classification of drug-induced autoimmunity (DIA). J Autoimmun 48–49:66–72

    Article  PubMed  CAS  Google Scholar 

  154. Rubin RL (2005) Drug-induced lupus. Toxicology 209:135–147

    Article  CAS  PubMed  Google Scholar 

  155. Yokogawa N, Vivino FB (2009) Hydralazine-induced autoimmune disease: comparison to idiopathic lupus and ANCA-positive vasculitis. Mod Rheumatol 19:338–347

    Article  PubMed  Google Scholar 

  156. Scheinbart LS, Johnson MA, Gross LA, Edelstein SR, Richardson BC (1991) Procainamide inhibits DNA methyltransferase in a human T cell line. J Rheumatol 18:530–534

    CAS  PubMed  Google Scholar 

  157. Lu Q, Wu A, Richardson BC (2005) Demethylation of the same promoter sequence increases CD70 expression in lupus T cells and T cells treated with lupus-inducing drugs. J Immunol 174:6212–6219

    Article  CAS  PubMed  Google Scholar 

  158. Kinch MS and Merkel J (2015) An analysis of FDA-approved drugs for inflammation and autoimmune diseases. Drug Discov Today

  159. Sada PR, Isenberg D, Ciurtin C (2015) Biologic treatment in Sjogren’s syndrome. Rheumatology (Oxford) 54:219–230

    Article  Google Scholar 

  160. Lisnevskaia L, Murphy G, Isenberg D (2014) Systemic lupus erythematosus. Lancet 384:1878–1888

    Article  PubMed  Google Scholar 

  161. Liberal R, Mieli-Vergani G, Vergani D (2013) Clinical significance of autoantibodies in autoimmune hepatitis. J Autoimmun 46:17–24

    Article  CAS  PubMed  Google Scholar 

  162. Invernizzi P (2013) Liver auto-immunology: the paradox of autoimmunity in a tolerogenic organ. J Autoimmun 46:1–6

    Article  CAS  PubMed  Google Scholar 

  163. Czaja AJ (2011) Drug-induced autoimmune-like hepatitis. Dig Dis Sci 56:958–976

    Article  CAS  PubMed  Google Scholar 

  164. Lawrenson RA, Seaman HE, Sundstrom A, Williams TJ, Farmer RD (2000) Liver damage associated with minocycline use in acne: a systematic review of the published literature and pharmacovigilance data. Drug Saf 23:333–349

    Article  CAS  PubMed  Google Scholar 

  165. Elkayam O, Yaron M, Caspi D (1999) Minocycline-induced autoimmune syndromes: an overview. Semin Arthritis Rheum 28:392–397

    Article  CAS  PubMed  Google Scholar 

  166. Amit G, Cohen P, Ackerman Z (2002) Nitrofurantoin-induced chronic active hepatitis. Isr Med Assoc J 4:184–186

    PubMed  Google Scholar 

  167. Hatoff DE, Cohen M, Schweigert BF, Talbert WM (1979) Nitrofurantoin: another cause of drug-induced chronic active hepatitis? A report of a patient with HLA-B8 antigen. Am J Med 67:117–121

    Article  CAS  PubMed  Google Scholar 

  168. Verthelyi D (2001) Sex hormones as immunomodulators in health and disease. Int Immunopharmacol 1:983–993

    Article  CAS  PubMed  Google Scholar 

  169. Gourdy P, Araujo LM, Zhu R et al (2005) Relevance of sexual dimorphism to regulatory T cells: estradiol promotes IFN-gamma production by invariant natural killer T cells. Blood 105:2415–2420

    Article  CAS  PubMed  Google Scholar 

  170. Ostensen M, Villiger PM, Forger F (2012) Interaction of pregnancy and autoimmune rheumatic disease. Autoimmun Rev 11:A437–A446

    Article  CAS  PubMed  Google Scholar 

  171. Parikh-Patel A, Gold E, Utts J, Gershwin ME (2002) The association between gravidity and primary biliary cirrhosis. Ann Epidemiol 12:264–272

    Article  PubMed  PubMed Central  Google Scholar 

  172. Cockrill T, del Junco DJ, Arnett FC et al (2010) Separate influences of birth order and gravidity/parity on the development of systemic sclerosis. Arthritis Care Res (Hoboken) 62:418–424

    Article  Google Scholar 

  173. Floreani A, Infantolino C, Franceschet I et al (2015) Pregnancy and primary biliary cirrhosis: a case-control study. Clin Rev Allergy Immunol 48:236–242

    Article  PubMed  Google Scholar 

  174. Lansink M, de Boer A, Dijkmans BA, Vandenbroucke JP, Hazes JM (1993) The onset of rheumatoid arthritis in relation to pregnancy and childbirth. Clin Exp Rheumatol 11:171–174

    CAS  PubMed  Google Scholar 

  175. Nelson JL, Ostensen M (1997) Pregnancy and rheumatoid arthritis. Rheum Dis Clin North Am 23:195–212

    Article  CAS  PubMed  Google Scholar 

  176. Costenbader KH, Feskanich D, Stampfer MJ, Karlson EW (2007) Reproductive and menopausal factors and risk of systemic lupus erythematosus in women. Arthritis Rheum 56:1251–1262

    Article  PubMed  Google Scholar 

  177. Hughes GC (2012) Progesterone and autoimmune disease. Autoimmun Rev 11:A502–A514

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Talal N (1978) Natural history of murine lupus. Modulation by sex hormones. Arthritis Rheum 21:S58–S63

    Article  CAS  PubMed  Google Scholar 

  179. Ganesan K, Balachandran C, Manohar BM, Puvanakrishnan R (2012) Effects of testosterone, estrogen and progesterone on TNF-alpha mediated cellular damage in rat arthritic synovial fibroblasts. Rheumatol Int 32:3181–3188

    Article  CAS  PubMed  Google Scholar 

  180. Ganesan K, Tiwari M, Balachandran C, Manohar BM, Puvanakrishnan R (2008) Estrogen and testosterone attenuate extracellular matrix loss in collagen-induced arthritis in rats. Calcif Tissue Int 83:354–364

    Article  CAS  PubMed  Google Scholar 

  181. Jochems C, Islander U, Erlandsson M et al (2011) Effects of oestradiol and raloxifene on the induction and effector phases of experimental postmenopausal arthritis and secondary osteoporosis. Clin Exp Immunol 165:121–129

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Chighizola C, Meroni PL (2012) The role of environmental estrogens and autoimmunity. Autoimmun Rev 11:A493–A501

    Article  CAS  PubMed  Google Scholar 

  183. Petri M, Allbritton J (1992) Hair product use in systemic lupus erythematosus. A case-control study. Arthritis Rheum 35:625–629

    Article  CAS  PubMed  Google Scholar 

  184. Sanchez-Guerrero J, Karlson EW, Colditz GA, Hunter DJ, Speizer FE, Liang MH (1996) Hair dye use and the risk of developing systemic lupus erythematosus. Arthritis Rheum 39:657–662

    Article  CAS  PubMed  Google Scholar 

  185. Gershwin ME, Selmi C, Worman HJ et al (2005) Risk factors and comorbidities in primary biliary cirrhosis: a controlled interview-based study of 1032 patients. Hepatology 42:1194–1202

    Article  PubMed  PubMed Central  Google Scholar 

  186. Cooper GS, Wither J, Bernatsky S et al (2010) Occupational and environmental exposures and risk of systemic lupus erythematosus: silica, sunlight, solvents. Rheumatology (Oxford) 49:2172–2180

    Article  CAS  Google Scholar 

  187. Long SA, Quan C, Van de Water J et al (2001) Immunoreactivity of organic mimeotopes of the E2 component of pyruvate dehydrogenase: connecting xenobiotics with primary biliary cirrhosis. J Immunol 167:2956–2963

    Article  CAS  PubMed  Google Scholar 

  188. Leung PS, Quan C, Park O et al (2003) Immunization with a xenobiotic 6-bromohexanoate bovine serum albumin conjugate induces antimitochondrial antibodies. J Immunol 170:5326–5332

    Article  CAS  PubMed  Google Scholar 

  189. Morikawa F, Fukuda M, Naganuma M, Nakayama Y (1976) Phototoxic reaction to xanthene dyes induced by visible light. J Dermatol 3:59–67

    Article  CAS  PubMed  Google Scholar 

  190. Lim SY, Ghosh SK (2003) Autoreactive responses to an environmental factor: 1. Phthalate induces antibodies exhibiting anti-DNA specificity. Immunology 110:482–492

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Lim SY, Ghosh SK (2004) Autoreactive responses to an environmental factor. 2. Phthalate-induced anti-DNA specificity is downregulated by autoreactive cytotoxic T cells. Immunology 112:94–104

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Lim SY, Ghosh SK (2005) Autoreactive responses to environmental factors: 3. Mouse strain-specific differences in induction and regulation of anti-DNA antibody responses due to phthalate-isomers. J Autoimmun 25:33–45

    Article  CAS  PubMed  Google Scholar 

  193. Amano K, Leung PS, Rieger R et al (2005) Chemical xenobiotics and mitochondrial autoantigens in primary biliary cirrhosis: identification of antibodies against a common environmental, cosmetic, and food additive, 2-octynoic acid. J Immunol 174:5874–5883

    Article  CAS  PubMed  Google Scholar 

  194. Chen RC, Naiyanetr P, Shu SA et al (2013) Antimitochondrial antibody heterogeneity and the xenobiotic etiology of primary biliary cirrhosis. Hepatology 57:1498–1508

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Leung PS, Wang J, Naiyanetr P et al (2013) Environment and primary biliary cirrhosis: electrophilic drugs and the induction of AMA. J Autoimmun 41:79–86

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Naiyanetr P, Butler JD, Meng L et al (2011) Electrophile-modified lipoic derivatives of PDC-E2 elicits anti-mitochondrial antibody reactivity. J Autoimmun 37:209–216

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Grob K, Vass M, Biedermann M, Neukom HP (2001) Contamination of animal feed and food from animal origin with mineral oil hydrocarbons. Food Addit Contam 18:1–10

    Article  CAS  PubMed  Google Scholar 

  198. Reeves WH, Lee PY, Weinstein JS, Satoh M, Lu L (2009) Induction of autoimmunity by pristane and other naturally occurring hydrocarbons. Trends Immunol 30:455–464

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Minhas U, Das P, Bhatnagar A (2011) Role of reactive intermediates in the immunopathogenesis of the pristane-induced Balb/c model of lupus. Lupus 20:1421–1425

    Article  CAS  PubMed  Google Scholar 

  200. Selmi C, De Santis M, Cavaciocchi F, Gershwin ME (2010) Infectious agents and xenobiotics in the etiology of primary biliary cirrhosis. Dis Markers 29:287–299

    Article  PubMed  PubMed Central  Google Scholar 

  201. Bogdanos DP, Pares A, Baum H et al (2004) Disease-specific cross-reactivity between mimicking peptides of heat shock protein of Mycobacterium gordonae and dominant epitope of E2 subunit of pyruvate dehydrogenase is common in Spanish but not British patients with primary biliary cirrhosis. J Autoimmun 22:353–362

    Article  CAS  PubMed  Google Scholar 

  202. Juran BD, Lazaridis KN (2014) Environmental factors in primary biliary cirrhosis. Semin Liver Dis 34:265–272

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Ortega-Hernandez OD, Levin NA, Altman A, Shoenfeld Y (2010) Infectious agents in the pathogenesis of primary biliary cirrhosis. Dis Markers 29:277–286

    Article  PubMed  PubMed Central  Google Scholar 

  204. Prince MI, Chetwynd A, Diggle P, Jarner M, Metcalf JV, James OF (2001) The geographical distribution of primary biliary cirrhosis in a well-defined cohort. Hepatology 34:1083–1088

    Article  CAS  PubMed  Google Scholar 

  205. Triger DR (1980) Primary biliary cirrhosis: an epidemiological study. Br Med J 281:772–775

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. McNally RJ, Ducker S, James OF (2009) Are transient environmental agents involved in the cause of primary biliary cirrhosis? Evidence from space-time clustering analysis. Hepatology 50:1169–1174

    Article  PubMed  Google Scholar 

  207. McNally RJ, James PW, Ducker S, James OF (2011) Seasonal variation in the patient diagnosis of primary biliary cirrhosis: further evidence for an environmental component to etiology. Hepatology 54:2099–2103

    Article  PubMed  Google Scholar 

  208. Dronamraju D, Odin J, Bach N (2010) Primary biliary cirrhosis: environmental risk factors. Dis Markers 29:323–328

    Article  PubMed  PubMed Central  Google Scholar 

  209. Ala A, Stanca CM, Bu-Ghanim M et al (2006) Increased prevalence of primary biliary cirrhosis near Superfund toxic waste sites. Hepatology 43:525–531

    Article  PubMed  Google Scholar 

  210. Kurth MJ, Yokoi T, Gershwin ME (2014) Halothane-induced hepatitis: paradigm or paradox for drug-induced liver injury. Hepatology 60:1473–1475

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Amano K, Leung PS, Xu Q et al (2004) Xenobiotic-induced loss of tolerance in rabbits to the mitochondrial autoantigen of primary biliary cirrhosis is reversible. J Immunol 172:6444–6452

    Article  CAS  PubMed  Google Scholar 

  212. Gershwin ME, Mackay IR (2008) The causes of primary biliary cirrhosis: convenient and inconvenient truths. Hepatology 47:737–745

    Article  PubMed  Google Scholar 

  213. Butler P, Valle F, Hamilton-Miller JM, Brumfitt W, Baum H, Burroughs AK (1993) M2 mitochondrial antibodies and urinary rough mutant bacteria in patients with primary biliary cirrhosis and in patients with recurrent bacteriuria. J Hepatol 17:408–414

    Article  CAS  PubMed  Google Scholar 

  214. Sluis-Cremer GK, Hessel PA, Hnizdo E, Churchill AR (1986) Relationship between silicosis and rheumatoid arthritis. Thorax 41:596–601

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Finckh A, Cooper GS, Chibnik LB et al (2006) Occupational silica and solvent exposures and risk of systemic lupus erythematosus in urban women. Arthritis Rheum 54:3648–3654

    Article  PubMed  Google Scholar 

  216. Parks CG, Cooper GS, Nylander-French LA et al (2002) Occupational exposure to crystalline silica and risk of systemic lupus erythematosus: a population-based, case-control study in the southeastern United States. Arthritis Rheum 46:1840–1850

    Article  PubMed  Google Scholar 

  217. Parks CG, Conrad K, Cooper GS (1999) Occupational exposure to crystalline silica and autoimmune disease. Environ Health Perspect 107(Suppl 5):793–802

    Article  PubMed  PubMed Central  Google Scholar 

  218. Hart JE, Laden F, Puett RC, Costenbader KH, Karlson EW (2009) Exposure to traffic pollution and increased risk of rheumatoid arthritis. Environ Health Perspect 117:1065–1069

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Zeft AS, Prahalad S, Lefevre S et al (2009) Juvenile idiopathic arthritis and exposure to fine particulate air pollution. Clin Exp Rheumatol 27:877–884

    CAS  PubMed  Google Scholar 

  220. Bernatsky S, Fournier M, Pineau CA, Clarke AE, Vinet E, Smargiassi A (2011) Associations between ambient fine particulate levels and disease activity in patients with systemic lupus erythematosus (SLE). Environ Health Perspect 119:45–49

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Balluz L, Philen R, Ortega L et al (2001) Investigation of systemic lupus erythematosus in Nogales, Arizona. Am J Epidemiol 154:1029–1036

    Article  CAS  PubMed  Google Scholar 

  222. Cooper GS, Parks CG, Treadwell EL, St Clair EW, Gilkeson GS, Dooley MA (2004) Occupational risk factors for the development of systemic lupus erythematosus. J Rheumatol 31:1928–1933

    PubMed  Google Scholar 

  223. Dahlgren J, Takhar H, Anderson-Mahoney P, Kotlerman J, Tarr J, Warshaw R (2007) Cluster of systemic lupus erythematosus (SLE) associated with an oil field waste site: a cross sectional study. Environ Health 6:8

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Annarosa Floreani.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Floreani, A., Leung, P.S.C. & Gershwin, M.E. Environmental Basis of Autoimmunity. Clinic Rev Allerg Immunol 50, 287–300 (2016). https://doi.org/10.1007/s12016-015-8493-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12016-015-8493-8

Keywords

Navigation