Skip to main content

Advertisement

Log in

The Therapeutic Potential of Epigenetics in Autoimmune Diseases

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

Abstract

Autoimmune diseases now include over 100 conditions and are estimated to affect over 20 million people in the United States or 5% of the world population with numerous geographical differences coined as geoepidemiology. Further, concordance rates in monozygotic twins are significantly higher compared to dizygotic sets while being significantly below 50% for most autoimmune diseases. These lines of evidence suggest that additional mechanisms are needed to link the individual susceptibility with the proposed chemical and infectious factors in the environment. Epigenetics may well constitute this missing link to include DNA methylation, histone changes, and microRNA which contribute to the epigenome characterizing specific diseases. Importantly, these epigenetic changes may be ideal targets for new personalized treatments as suggested by data in cancer. A number of chemical and physical factors, along with proposed infectious agents or aging, are involved in the etiopathogenesis of autoimmune diseases through epigenetic changes. The most prominent evidence on the association between environment and autoimmunity has been reported in systemic lupus erythematosus, but similar mechanisms were proposed in rheumatoid arthritis, systemic sclerosis, and type 1 diabetes.

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.

Similar content being viewed by others

References

  1. Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, McCarthy MI, Ramos EM, Cardon LR, Chakravarti A, Cho JH, Guttmacher AE, Kong A, Kruglyak L, Mardis E, Rotimi CN, Slatkin M, Valle D, Whittemore AS, Boehnke M, Clark AG, Eichler EE, Gibson G, Haines JL, Mackay TF, McCarroll SA, Visscher PM (2009) Finding the missing heritability of complex diseases. Nature 461:747–753

    PubMed  CAS  Google Scholar 

  2. Eichler EE, Flint J, Gibson G, Kong A, Leal SM, Moore JH, Nadeau JH (2010) Missing heritability and strategies for finding the underlying causes of complex disease. Nat Rev Genet 11:446–450

    PubMed  CAS  Google Scholar 

  3. Naidoo N, Chia KS (2009) Discovering gene-environment interactions in the post-genomic era. J Prev Med Public Health 42:356–359

    PubMed  Google Scholar 

  4. Vermeire S (2011) To what extent are genetics clinically useful? Best Pract Res Clin Gastroenterol 25(Suppl 1):S8–14

    PubMed  Google Scholar 

  5. Asimit J, Zeggini E (2011) Testing for rare variant associations in complex diseases. Genome Med 3:24

    PubMed  Google Scholar 

  6. Renaudineau Y, Youinou P (2011) Epigenetics and autoimmunity, with special emphasis on methylation. Keio J Med 60:10–16

    PubMed  CAS  Google Scholar 

  7. Meda F, Folci M, Baccarelli A, Selmi C (2011) The epigenetics of autoimmunity. Cell Mol Immunol 8:226–236

    PubMed  CAS  Google Scholar 

  8. Brooks WH, Le Dantec C, Pers JO, Youinou P, Renaudineau Y (2010) Epigenetics and autoimmunity. J Autoimmun 34:J207–219

    PubMed  CAS  Google Scholar 

  9. Dieker J, Muller S (2010) Epigenetic histone code and autoimmunity. Clin Rev Allergy Immunol 39:78–84

    PubMed  CAS  Google Scholar 

  10. Dai Y, Zhang L, Hu C, Zhang Y (2010) Genome-wide analysis of histone h3 lysine 4 trimethylation by chip-chip in peripheral blood mononuclear cells of systemic lupus erythematosus patients. Clin Exp Rheumatol 28:158–168

    PubMed  CAS  Google Scholar 

  11. Lashine YA, Seoudi AM, Salah S, Abdelaziz AI (2011) Expression signature of microrna-181-a reveals its crucial role in the pathogenesis of paediatric systemic lupus erythematosus. Clin Exp Rheumatol 29:351–357

    PubMed  CAS  Google Scholar 

  12. Faulk C, Dolinoy DC (2011) Timing is everything: The when and how of environmentally induced changes in the epigenome of animals. Epigenetics 6:791–797

    PubMed  Google Scholar 

  13. Skinner MK (2011) Environmental epigenomics and disease susceptibility. EMBO Rep 12:620–622

    PubMed  CAS  Google Scholar 

  14. Ballestar E (2010) Epigenetics lessons from twins: Prospects for autoimmune disease. Clin Rev Allergy Immunol 39:30–41

    PubMed  CAS  Google Scholar 

  15. Masatlioglu S, Seyahi E, Tahir Turanli E, Fresko I, Gogus F, Senates E, Oguz Savran F, Yazici H (2010) A twin study in behcet's syndrome. Clin Exp Rheumatol 28:S62–66

    PubMed  Google Scholar 

  16. Condon J, Shaw JE, Luciano M, Kyvik KO, Martin NG, Duffy DL (2008) A study of diabetes mellitus within a large sample of australian twins. Twin Res Hum Genet 11:28–40

    PubMed  Google Scholar 

  17. Hyttinen V, Kaprio J, Kinnunen L, Koskenvuo M, Tuomilehto J (2003) Genetic liability of type 1 diabetes and the onset age among 22,650 young finnish twin pairs: A nationwide follow-up study. Diabetes 52:1052–1055

    PubMed  CAS  Google Scholar 

  18. Kyvik KO, Green A, Beck-Nielsen H (1995) Concordance rates of insulin dependent diabetes mellitus: A population based study of young danish twins. Bmj 311:913–917

    PubMed  CAS  Google Scholar 

  19. Lawrence JS (1970) Heberden oration, 1969. Rheumatoid arthritis--nature or nurture? Ann Rheum Dis 29:357–379

    PubMed  CAS  Google Scholar 

  20. Bellamy N, Duffy D, Martin N, Mathews J (1992) Rheumatoid arthritis in twins: A study of aetiopathogenesis based on the australian twin registry. Ann Rheum Dis 51:588–593

    PubMed  CAS  Google Scholar 

  21. Aho K, Koskenvuo M, Tuominen J, Kaprio J (1986) Occurrence of rheumatoid arthritis in a nationwide series of twins. J Rheumatol 13:899–902

    PubMed  CAS  Google Scholar 

  22. Silman AJ, MacGregor AJ, Thomson W, Holligan S, Carthy D, Farhan A, Ollier WE (1993) Twin concordance rates for rheumatoid arthritis: Results from a nationwide study. Br J Rheumatol 32:903–907

    PubMed  CAS  Google Scholar 

  23. Svendsen AJ, Holm NV, Kyvik K, Petersen PH, Junker P (2002) Relative importance of genetic effects in rheumatoid arthritis: Historical cohort study of danish nationwide twin population. Bmj 324:264–266

    PubMed  Google Scholar 

  24. Block SR (2006) A brief history of twins. Lupus 15:61–64

    PubMed  CAS  Google Scholar 

  25. Block SR, Winfield JB, Lockshin MD, D'Angelo WA, Christian CL (1975) Studies of twins with systemic lupus erythematosus. A review of the literature and presentation of 12 additional sets. Am J Med 59:533–552

    PubMed  CAS  Google Scholar 

  26. Grennan DM, Parfitt A, Manolios N, Huang Q, Hyland V, Dunckley H, Doran T, Gatenby P, Badcock C (1997) Family and twin studies in systemic lupus erythematosus. Dis Markers 13:93–98

    PubMed  CAS  Google Scholar 

  27. Willer CJ, Dyment DA, Risch NJ, Sadovnick AD, Ebers GC (2003) Twin concordance and sibling recurrence rates in multiple sclerosis. Proc Natl Acad Sci U S A 100:12877–12882

    PubMed  CAS  Google Scholar 

  28. Ristori G, Cannoni S, Stazi MA, Vanacore N, Cotichini R, Alfo M, Pugliatti M, Sotgiu S, Solaro C, Bomprezzi R, Di Giovanni S, Figa Talamanca L, Nistico L, Fagnani C, Neale MC, Cascino I, Giorgi G, Battaglia MA, Buttinelli C, Tosi R, Salvetti M (2006) Multiple sclerosis in twins from continental italy and sardinia: A nationwide study. Ann Neurol 59:27–34

    PubMed  Google Scholar 

  29. Hansen T, Skytthe A, Stenager E, Petersen HC, Bronnum-Hansen H, Kyvik KO (2005) Concordance for multiple sclerosis in danish twins: An update of a nationwide study. Mult Scler 11:504–510

    PubMed  CAS  Google Scholar 

  30. (1992) Multiple sclerosis in 54 twinships: Concordance rate is independent of zygosity. French research group on multiple sclerosis. Ann Neurol 32,724-727.

  31. Feghali-Bostwick C, Medsger TA Jr, Wright TM (2003) Analysis of systemic sclerosis in twins reveals low concordance for disease and high concordance for the presence of antinuclear antibodies. Arthritis Rheum 48:1956–1963

    PubMed  Google Scholar 

  32. Nistico L, Fagnani C, Coto I, Percopo S, Cotichini R, Limongelli MG, Paparo F, D'Alfonso S, Giordano M, Sferlazzas C, Magazzu G, Momigliano-Richiardi P, Greco L, Stazi MA (2006) Concordance, disease progression, and heritability of coeliac disease in italian twins. Gut 55:803–808

    PubMed  CAS  Google Scholar 

  33. Greco L, Romino R, Coto I, Di Cosmo N, Percopo S, Maglio M, Paparo F, Gasperi V, Limongelli MG, Cotichini R, D'Agate C, Tinto N, Sacchetti L, Tosi R, Stazi MA (2002) The first large population based twin study of coeliac disease. Gut 50:624–628

    PubMed  CAS  Google Scholar 

  34. Selmi C, Mayo MJ, Bach N, Ishibashi H, Invernizzi P, Gish RG, Gordon SC, Wright HI, Zweiban B, Podda M, Gershwin ME (2004) Primary biliary cirrhosis in monozygotic and dizygotic twins: Genetics, epigenetics, and environment. Gastroenterology 127:485–492

    PubMed  Google Scholar 

  35. Szyf M (2010) Epigenetic therapeutics in autoimmune disease. Clin Rev Allergy Immunol 39:62–77

    PubMed  CAS  Google Scholar 

  36. Richardson B, Kahn L, Lovett EJ, Hudson J (1986) Effect of an inhibitor of DNA methylation on t cells. I. 5-azacytidine induces t4 expression on t8+ t cells. J Immunol 137:35–39

    PubMed  CAS  Google Scholar 

  37. Lu Q, Kaplan M, Ray D, Zacharek S, Gutsch D, Richardson B (2002) Demethylation of itgal (cd11a) regulatory sequences in systemic lupus erythematosus. Arthritis Rheum 46:1282–1291

    PubMed  CAS  Google Scholar 

  38. Zhao M, Sun Y, Gao F, Wu X, Tang J, Yin H, Luo Y, Richardson B, Lu Q (2010) Epigenetics and sle: Rfx1 downregulation causes cd11a and cd70 overexpression by altering epigenetic modifications in lupus cd4+ t cells. J Autoimmun 35:58–69

    PubMed  Google Scholar 

  39. Zhao S, Long H, Lu Q (2010) Epigenetic perspectives in systemic lupus erythematosus: Pathogenesis, biomarkers, and therapeutic potentials. Clin Rev Allergy Immunol 39:3–9

    PubMed  Google Scholar 

  40. Gorelik G, Fang JY, Wu A, Sawalha AH, Richardson B (2007) Impaired t cell protein kinase c delta activation decreases erk pathway signaling in idiopathic and hydralazine-induced lupus. J Immunol 179:5553–5563

    PubMed  CAS  Google Scholar 

  41. 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

    PubMed  CAS  Google Scholar 

  42. Biggioggero M, Meroni PL (2010) The geoepidemiology of the antiphospholipid antibody syndrome. Autoimmun Rev 9:A299–304

    PubMed  Google Scholar 

  43. Borchers AT, Naguwa SM, Shoenfeld Y, Gershwin ME (2010) The geoepidemiology of systemic lupus erythematosus. Autoimmun Rev 9:A277–287

    PubMed  CAS  Google Scholar 

  44. 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

    PubMed  Google Scholar 

  45. Park SK, O'Neill MS, Vokonas PS, Sparrow D, Spiro A 3rd, Tucker KL, Suh H, Hu H, Schwartz J (2008) Traffic-related particles are associated with elevated homocysteine: The va normative aging study. Am J Respir Crit Care Med 178:283–289

    PubMed  Google Scholar 

  46. Rai K, Huggins IJ, James SR, Karpf AR, Jones DA, Cairns BR (2008) DNA demethylation in zebrafish involves the coupling of a deaminase, a glycosylase, and gadd45. Cell 135:1201–1212

    PubMed  CAS  Google Scholar 

  47. Cornelius LA, Sepp N, Li LJ, Degitz K, Swerlick RA, Lawley TJ, Caughman SW (1994) Selective upregulation of intercellular adhesion molecule (icam-1) by ultraviolet b in human dermal microvascular endothelial cells. J Invest Dermatol 103:23–28

    PubMed  CAS  Google Scholar 

  48. Li Y, Zhao M, Yin H, Gao F, Wu X, Luo Y, Zhao S, Zhang X, Su Y, Hu N, Long H, Richardson B, Lu Q (2010) Overexpression of the growth arrest and DNA damage-induced 45alpha gene contributes to autoimmunity by promoting DNA demethylation in lupus t cells. Arthritis Rheum 62:1438–1447

    PubMed  CAS  Google Scholar 

  49. Salvador JM, Hollander MC, Nguyen AT, Kopp JB, Barisoni L, Moore JK, Ashwell JD, Fornace AJ Jr (2002) Mice lacking the p53-effector gene gadd45a develop a lupus-like syndrome. Immunity 16:499–508

    PubMed  CAS  Google Scholar 

  50. Grolleau-Julius A, Ray D, Yung RL (2010) The role of epigenetics in aging and autoimmunity. Clin Rev Allergy Immunol 39:42–50

    PubMed  CAS  Google Scholar 

  51. Huck S, Deveaud E, Namane A, Zouali M (1999) Abnormal DNA methylation and deoxycytosine-deoxyguanine content in nucleosomes from lymphocytes undergoing apoptosis. Faseb J 13:1415–1422

    PubMed  CAS  Google Scholar 

  52. Huck S, Zouali M (1996) DNA methylation: A potential pathway to abnormal autoreactive lupus b cells. Clin Immunol Immunopathol 80:1–8

    PubMed  CAS  Google Scholar 

  53. 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

    PubMed  Google Scholar 

  54. Makrygiannakis D, Hermansson M, Ulfgren AK, Nicholas AP, Zendman AJ, Eklund A, Grunewald J, Skold CM, Klareskog L, Catrina AI (2008) Smoking increases peptidylarginine deiminase 2 enzyme expression in human lungs and increases citrullination in bal cells. Ann Rheum Dis 67:1488–1492

    PubMed  CAS  Google Scholar 

  55. Moscarello MA, Mastronardi FG, Wood DD (2007) The role of citrullinated proteins suggests a novel mechanism in the pathogenesis of multiple sclerosis. Neurochem Res 32:251–256

    PubMed  CAS  Google Scholar 

  56. Hov JR, Lleo A, Selmi C, Woldseth B, Fabris L, Strazzabosco M, Karlsen TH, Invernizzi P (2010) Genetic associations in italian primary sclerosing cholangitis: Heterogeneity across europe defines a critical role for hla-c. J Hepatol 52:712–717

    PubMed  CAS  Google Scholar 

  57. Gorlova O, Martin JE, Rueda B, Koeleman BP, Ying J, Teruel M, Diaz-Gallo LM, Broen JC, Vonk MC, Simeon CP, Alizadeh BZ, Coenen MJ, Voskuyl AE, Schuerwegh AJ, van Riel PL, Vanthuyne M, van’t Slot R, Italiaander A, Ophoff RA, Hunzelmann N, Fonollosa V, Ortego-Centeno N, Gonzalez-Gay MA, Garcia-Hernandez FJ, Gonzalez-Escribano MF, Airo P, van Laar J, Worthington J, Hesselstrand R, Smith V, de Keyser F, Houssiau F, Chee MM, Madhok R, Shiels PG, Westhovens R, Kreuter A, de Baere E, Witte T, Padyukov L, Nordin A, Scorza R, Lunardi C, Lie BA, Hoffmann-Vold AM, Palm O, Garcia de la Pena P, Carreira P, Varga J, Hinchcliff M, Lee AT, Gourh P, Amos CI, Wigley FM, Hummers LK, Nelson JL, Riemekasten G, Herrick A, Beretta L, Fonseca C, Denton CP, Gregersen PK, Agarwal S, Assassi S, Tan FK, Arnett FC, Radstake TR, Mayes MD, Martin J (2011) Correction: Identification of novel genetic markers associated with clinical phenotypes of systemic sclerosis through a genome-wide association strategy. PLoS Genet 7

  58. Elo LL, Mykkanen J, Nikula T, Jarvenpaa H, Simell S, Aittokallio T, Hyoty H, Ilonen J, Veijola R, Simell T, Knip M, Simell O, Lahesmaa R (2010) Early suppression of immune response pathways characterizes children with prediabetes in genome-wide gene expression profiling. J Autoimmun 35:70–76

    PubMed  CAS  Google Scholar 

  59. Hasham A, Tomer Y (2011) The recent rise in the frequency of type 1 diabetes: Who pulled the trigger? J Autoimmun 37:1–2

    PubMed  Google Scholar 

  60. Scott GS, Fishman S, Khai Siew L, Margalit A, Chapman S, Chervonsky AV, Wen L, Gross G, Wong FS (2010) Immunotargeting of insulin reactive cd8 t cells to prevent diabetes. J Autoimmun 35:390–397

    PubMed  CAS  Google Scholar 

  61. Van Belle TL, Juntti T, Liao J, von Herrath MG (2010) Pre-existing autoimmunity determines type 1 diabetes outcome after flt3-ligand treatment. J Autoimmun 34:445–452

    PubMed  Google Scholar 

  62. Ziegler AG, Pflueger M, Winkler C, Achenbach P, Akolkar B, Krischer JP, Bonifacio E (2011) Accelerated progression from islet autoimmunity to diabetes is causing the escalating incidence of type 1 diabetes in young children. J Autoimmun 37:3–7

    PubMed  Google Scholar 

  63. Lempainen J, Vaarala O, Makela M, Veijola R, Simell O, Knip M, Hermann R, Ilonen J (2009) Interplay between ptpn22 c1858t polymorphism and cow's milk formula exposure in type 1 diabetes. J Autoimmun 33:155–164

    PubMed  CAS  Google Scholar 

  64. Chamson-Reig A, Arany EJ, Summers K, Hill DJ (2009) A low protein diet in early life delays the onset of diabetes in the non-obese diabetic mouse. J Endocrinol 201:231–239

    PubMed  CAS  Google Scholar 

  65. Boujendar S, Arany E, Hill D, Remacle C, Reusens B (2003) Taurine supplementation of a low protein diet fed to rat dams normalizes the vascularization of the fetal endocrine pancreas. J Nutr 133:2820–2825

    PubMed  CAS  Google Scholar 

  66. Arany E, Strutt B, Romanus P, Remacle C, Reusens B, Hill DJ (2004) Taurine supplement in early life altered islet morphology, decreased insulitis and delayed the onset of diabetes in non-obese diabetic mice. Diabetologia 47:1831–1837

    PubMed  CAS  Google Scholar 

  67. 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

    PubMed  Google Scholar 

  68. Selmi C, Gershwin ME (2009) The role of environmental factors in primary biliary cirrhosis. Trends Immunol 30:415–420

    PubMed  CAS  Google Scholar 

  69. Katz U, Zandman-Goddard G (2010) Drug-induced lupus: An update. Autoimmun Rev 10:46–50

    PubMed  CAS  Google Scholar 

  70. Renaudineau Y, Garaud S, Le Dantec C, Alonso-Ramirez R, Daridon C, Youinou P (2010) Autoreactive b cells and epigenetics. Clin Rev Allergy Immunol 39:85–94

    PubMed  CAS  Google Scholar 

  71. Korganow AS, Knapp AM, Nehme-Schuster H, Soulas-Sprauel P, Poindron V, Pasquali JL, Martin T (2010) Peripheral b cell abnormalities in patients with systemic lupus erythematosus in quiescent phase: Decreased memory b cells and membrane cd19 expression. J Autoimmun 34:426–434

    PubMed  CAS  Google Scholar 

  72. Scheinecker C, Bonelli M, Smolen JS (2010) Pathogenetic aspects of systemic lupus erythematosus with an emphasis on regulatory t cells. J Autoimmun 35:269–275

    PubMed  CAS  Google Scholar 

  73. Santiago-Raber ML, Dunand-Sauthier I, Wu T, Li QZ, Uematsu S, Akira S, Reith W, Mohan C, Kotzin BL, Izui S (2010) Critical role of tlr7 in the acceleration of systemic lupus erythematosus in tlr9-deficient mice. J Autoimmun 34:339–348

    PubMed  CAS  Google Scholar 

  74. Kontaki E, Boumpas DT (2010) Innate immunity in systemic lupus erythematosus: Sensing endogenous nucleic acids. J Autoimmun 35:206–211

    PubMed  CAS  Google Scholar 

  75. Adams LE, Mongey AB (1994) Role of genetic factors in drug-related autoimmunity. Lupus 3:443–447

    PubMed  CAS  Google Scholar 

  76. Reidenberg MM, Drayer DE, Lorenzo B, Strom BL, West SL, Snyder ES, Freundlich B, Stolley PD (1993) Acetylation phenotypes and environmental chemical exposure of people with idiopathic systemic lupus erythematosus. Arthritis Rheum 36:971–973

    PubMed  CAS  Google Scholar 

  77. Hsieh CC, Lin BF (2011) Dietary factors regulate cytokines in murine models of systemic lupus erythematosus. Autoimmun Rev

  78. Fu SM, Deshmukh US, Gaskin F (2011) Pathogenesis of systemic lupus erythematosus revisited 2011: End organ resistance to damage, autoantibody initiation and diversification, and hla-dr. J Autoimmun 37:104–112

    PubMed  CAS  Google Scholar 

  79. Mazari L, Ouarzane M, Zouali M (2007) Subversion of b lymphocyte tolerance by hydralazine, a potential mechanism for drug-induced lupus. Proc Natl Acad Sci U S A 104:6317–6322

    PubMed  CAS  Google Scholar 

  80. Selmi C, Papini AM, Pugliese P, Arcaro MC, Gershwin ME (2011) Environmental pathways to autoimmune diseases: The cases of primary biliary cirrhosis and multiple sclerosis. Arch Med Sci in press

  81. Gorelik G, Richardson B (2009) Aberrant t cell erk pathway signaling and chromatin structure in lupus. Autoimmun Rev 8:196–198

    PubMed  CAS  Google Scholar 

  82. Borchers AT, Uibo R, Gershwin ME (2010) The geoepidemiology of type 1 diabetes. Autoimmun Rev 9:A355–365

    PubMed  Google Scholar 

  83. Chandran V, Raychaudhuri SP (2010) Geoepidemiology and environmental factors of psoriasis and psoriatic arthritis. J Autoimmun 34:J314–321

    PubMed  CAS  Google Scholar 

  84. Chen M, Kallenberg CG (2010) The environment, geoepidemiology and anca-associated vasculitides. Autoimmun Rev 9:A293–298

    PubMed  CAS  Google Scholar 

  85. Deane S, Teuber SS, Gershwin ME (2010) The geoepidemiology of immune thrombocytopenic purpura. Autoimmun Rev 9:A342–349

    PubMed  Google Scholar 

  86. Ehrenfeld M (2010) Geoepidemiology: The environment and spondyloarthropathies. Autoimmun Rev 9:A325–329

    PubMed  Google Scholar 

  87. Invernizzi P (2010) Geoepidemiology of autoimmune liver diseases. J Autoimmun 34:J300–306

    PubMed  CAS  Google Scholar 

  88. Lambert JF, Nydegger UE (2010) Geoepidemiology of autoimmune hemolytic anemia. Autoimmun Rev 9:A350–354

    PubMed  CAS  Google Scholar 

  89. Logan I, Bowlus CL (2010) The geoepidemiology of autoimmune intestinal diseases. Autoimmun Rev 9:A372–378

    PubMed  Google Scholar 

  90. Mavragani CP, Moutsopoulos HM (2010) The geoepidemiology of sjogren's syndrome. Autoimmun Rev 9:A305–310

    PubMed  Google Scholar 

  91. Meyer A, Levy Y (2010) Chapter 33: Geoepidemiology of myasthenia gravis. Autoimmun Rev 9:A383–386

    PubMed  Google Scholar 

  92. Milo R, Kahana E (2010) Multiple sclerosis: Geoepidemiology, genetics and the environment. Autoimmun Rev 9:A387–394

    PubMed  CAS  Google Scholar 

  93. Prieto S, Grau JM (2010) The geoepidemiology of autoimmune muscle disease. Autoimmun Rev 9:A330–334

    PubMed  Google Scholar 

  94. Ranque B, Mouthon L (2010) Geoepidemiology of systemic sclerosis. Autoimmun Rev 9:A311–318

    PubMed  Google Scholar 

  95. Sands J, Tuscano JM (2010) Geoepidemiology and autoimmune manifestations of lymphoproliferative disorders. Autoimmun Rev 9:A335–341

    PubMed  Google Scholar 

  96. Selmi C (2010) The worldwide gradient of autoimmune conditions. Autoimmun Rev 9:A247–250

    PubMed  Google Scholar 

  97. Selmi C, Tsuneyama K (2010) Nutrition, geoepidemiology, and autoimmunity. Autoimmun Rev 9:A267–270

    PubMed  CAS  Google Scholar 

  98. Shapira Y, Agmon-Levin N, Shoenfeld Y (2010) Defining and analyzing geoepidemiology and human autoimmunity. J Autoimmun 34:J168–177

    PubMed  CAS  Google Scholar 

  99. Shapira Y, Poratkatz BS, Gilburd B, Barzilai O, Ram M, Blank M, Lindeberg S, Frostegard J, Anaya JM, Bizzaro N, Jara LJ, Damoiseaux J, Shoenfeld Y, Agmon Levin N (2011) Geographical differences in autoantibodies and anti-infectious agents antibodies among healthy adults. Clin Rev Allergy Immunol

  100. Youinou P, Pers JO, Gershwin ME, Shoenfeld Y (2010) Geo-epidemiology and autoimmunity. J Autoimmun 34:J163–167

    PubMed  CAS  Google Scholar 

  101. Lessard CJ, Adrianto I, Kelly JA, Kaufman KM, Grundahl KM, Adler A, Williams AH, Gallant CJ, Anaya JM, Bae SC, Boackle SA, Brown EE, Chang DM, Criswell LA, Edberg JC, Freedman BI, Gregersen PK, Gilkeson GS, Jacob CO, James JA, Kamen DL, Kimberly RP, Martin J, Merrill JT, Niewold TB, Park SY, Petri MA, Pons-Estel BA, Ramsey-Goldman R, Reveille JD, Song YW, Stevens AM, Tsao BP, Vila LM, Vyse TJ, Yu CY, Guthridge JM, Bruner GR, Langefeld CD, Montgomery C, Harley JB, Scofield RH, Gaffney PM, Moser KL (2011) Identification of a systemic lupus erythematosus susceptibility locus at 11p13 between pdhx and cd44 in a multiethnic study. Am J Hum Genet 88:83–91

    PubMed  CAS  Google Scholar 

  102. Sheng YJ, Gao JP, Li J, Han JW, Xu Q, Hu WL, Pan TM, Cheng YL, Yu ZY, Ni C, Yao S, He CF, Liu YS, Li Y, Ge HM, Xiao FL, Sun LD, Yang S, Zhang XJ (2011) Follow-up study identifies two novel susceptibility loci prkcb and 8p11.21 for systemic lupus erythematosus. Rheumatology (Oxford) 50:682–688

    CAS  Google Scholar 

  103. Han JW, Zheng HF, Cui Y, Sun LD, Ye DQ, Hu Z, Xu JH, Cai ZM, Huang W, Zhao GP, Xie HF, Fang H, Lu QJ, Li XP, Pan YF, Deng DQ, Zeng FQ, Ye ZZ, Zhang XY, Wang QW, Hao F, Ma L, Zuo XB, Zhou FS, Du WH, Cheng YL, Yang JQ, Shen SK, Li J, Sheng YJ, Zuo XX, Zhu WF, Gao F, Zhang PL, Guo Q, Li B, Gao M, Xiao FL, Quan C, Zhang C, Zhang Z, Zhu KJ, Li Y, Hu DY, Lu WS, Huang JL, Liu SX, Li H, Ren YQ, Wang ZX, Yang CJ, Wang PG, Zhou WM, Lv YM, Zhang AP, Zhang SQ, Lin D, Low HQ, Shen M, Zhai ZF, Wang Y, Zhang FY, Yang S, Liu JJ, Zhang XJ (2009) Genome-wide association study in a chinese han population identifies nine new susceptibility loci for systemic lupus erythematosus. Nat Genet 41:1234–1237

    PubMed  CAS  Google Scholar 

  104. Cunninghame Graham DS (2009) Genome-wide association studies in systemic lupus erythematosus: A perspective. Arthritis Res Ther 11:119

    PubMed  Google Scholar 

  105. Gorlova O, Martin JE, Rueda B, Koeleman BP, Ying J, Teruel M, Diaz-Gallo LM, Broen JC, Vonk MC, Simeon CP, Alizadeh BZ, Coenen MJ, Voskuyl AE, Schuerwegh AJ, van Riel PL, Vanthuyne M, Slot Rvan't, Italiaander A, Ophoff RA, Hunzelmann N, Fonollosa V, Ortego-Centeno N, Gonzalez-Gay MA, Garcia-Hernandez FJ, Gonzalez-Escribano MF, Airo P, van Laar J, Worthington J, Hesselstrand R, Smith V, de Keyser F, Houssiau F, Chee MM, Madhok R, Shiels PG, Westhovens R, Kreuter A, de Baere E, Witte T, Padyukov L, Nordin A, Scorza R, Lunardi C, Lie BA, Hoffmann-Vold AM, Palm O, Garcia de la Pena P, Carreira P, Varga J, Hinchcliff M, Lee AT, Gourh P, Amos CI, Wigley FM, Hummers LK, Hummers J, Nelson JL, Riemekasten G, Herrick A, Beretta L, Fonseca C, Denton CP, Gregersen PK, Agarwal S, Assassi S, Tan FK, Arnett FC, Radstake TR, Mayes MD, Martin J (2011) Identification of novel genetic markers associated with clinical phenotypes of systemic sclerosis through a genome-wide association strategy. PLoS Genet 7:e1002178

    PubMed  CAS  Google Scholar 

  106. Allanore Y, Saad M, Dieude P, Avouac J, Distler JH, Amouyel P, Matucci-Cerinic M, Riemekasten G, Airo P, Melchers I, Hachulla E, Cusi D, Wichmann HE, Wipff J, Lambert JC, Hunzelmann N, Tiev K, Caramaschi P, Diot E, Kowal-Bielecka O, Valentini G, Mouthon L, Czirjak L, Damjanov N, Salvi E, Conti C, Muller M, Muller-Ladner U, Riccieri V, Ruiz B, Cracowski JL, Letenneur L, Dupuy AM, Meyer O, Kahan A, Munnich A, Boileau C, Martinez M (2011) Genome-wide scan identifies tnip1, psors1c1, and rhob as novel risk loci for systemic sclerosis. PLoS Genet 7:e1002091

    PubMed  CAS  Google Scholar 

  107. Hu X, Kim H, Stahl E, Plenge R, Daly M, Raychaudhuri S (2011) Integrating autoimmune risk loci with gene-expression data identifies specific pathogenic immune cell subsets. Am J Hum Genet

  108. Deshmukh H, Kim-Howard X, Nath SK (2009) Replication of recently identified associated single-nucleotide polymorphisms from six autoimmune diseases in genetic analysis workshop 16 rheumatoid arthritis data. BMC Proc 7(3):S31

    Google Scholar 

  109. Danska JS, Poussier P (2009) After the gwas rush: Nuggets of insight into the pathogenesis of autoimmune disease. Semin Immunol 21:313–317

    PubMed  Google Scholar 

  110. Lettre G, Rioux JD (2008) Autoimmune diseases: Insights from genome-wide association studies. Hum Mol Genet 17:R116–121

    PubMed  CAS  Google Scholar 

  111. Jirtle RL, Skinner MK (2007) Environmental epigenomics and disease susceptibility. Nat Rev Genet 8:253–262

    PubMed  CAS  Google Scholar 

  112. Lucchinetti E, Feng J, Silva R, Tolstonog GV, Schaub MC, Schumann GG, Zaugg M (2006) Inhibition of line-1 expression in the heart decreases ischemic damage by activation of akt/pkb signaling. Physiol Genomics 25:314–324

    PubMed  CAS  Google Scholar 

  113. Belinsky SA, Snow SS, Nikula KJ, Finch GL, Tellez CS, Palmisano WA (2002) Aberrant cpg island methylation of the p16(ink4a) and estrogen receptor genes in rat lung tumors induced by particulate carcinogens. Carcinogenesis 23:335–339

    PubMed  CAS  Google Scholar 

  114. Wright RO, Baccarelli A (2007) Metals and neurotoxicology. J Nutr 137:2809–2813

    PubMed  CAS  Google Scholar 

  115. Arora-Singh RK, Assassi S, del Junco DJ, Arnett FC, Perry M, Irfan U, Sharif R, Mattar T, Mayes MD (2010) Autoimmune diseases and autoantibodies in the first degree relatives of patients with systemic sclerosis. J Autoimmun 35:52–57

    PubMed  CAS  Google Scholar 

  116. Gourh P, Agarwal SK, Martin E, Divecha D, Rueda B, Bunting H, Assassi S, Paz G, Shete S, McNearney T, Draeger H, Reveille JD, Radstake TR, Simeon CP, Rodriguez L, Vicente E, Gonzalez-Gay MA, Mayes MD, Tan FK, Martin J, Arnett FC (2010) Association of the c8orf13-blk region with systemic sclerosis in north-american and european populations. J Autoimmun 34:155–162

    PubMed  CAS  Google Scholar 

  117. Takagi K, Kawaguchi Y, Kawamoto M, Ota Y, Tochimoto A, Gono T, Katsumata Y, Takagi M, Hara M, Yamanaka H (2011) Activation of the activin a-alk-smad pathway in systemic sclerosis. J Autoimmun 36:181–188

    PubMed  CAS  Google Scholar 

  118. Iobagiu C, Magyar A, Nogueira L, Cornillet M, Sebbag M, Arnaud J, Hudecz F, Serre G (2011) The antigen specificity of the rheumatoid arthritis-associated acpa directed to citrullinated fibrin is very closely restricted. J Autoimmun

  119. Somers K, Geusens P, Elewaut D, De Keyser F, Rummens JL, Coenen M, Blom M, Stinissen P, Somers V (2011) Novel autoantibody markers for early and seronegative rheumatoid arthritis. J Autoimmun 36:33–46

    PubMed  CAS  Google Scholar 

  120. Trenkmann M, Brock M, Ospelt C, Gay S (2010) Epigenetics in rheumatoid arthritis. Clin Rev Allergy Immunol 39:10–19

    PubMed  CAS  Google Scholar 

  121. Ospelt C, Reedquist KA, Gay S, Tak PP (2011) Inflammatory memories: Is epigenetics the missing link to persistent stromal cell activation in rheumatoid arthritis? Autoimmun Rev 10:519–524

    PubMed  Google Scholar 

  122. Gonzalez S, Aguilera S, Urzua U, Quest AF, Molina C, Alliende C, Hermoso M, Gonzalez MJ (2011) Mechanotransduction and epigenetic control in autoimmune diseases. Autoimmun Rev 10:175–179

    PubMed  Google Scholar 

  123. Lei W, Luo Y, Yan K, Zhao S, Li Y, Qiu X, Zhou Y, Long H, Zhao M, Liang Y, Su Y, Lu Q (2009) Abnormal DNA methylation in cd4+ t cells from patients with systemic lupus erythematosus, systemic sclerosis, and dermatomyositis. Scand J Rheumatol 38:369–374

    PubMed  CAS  Google Scholar 

  124. Karouzakis E, Gay RE, Michel BA, Gay S, Neidhart M (2009) DNA hypomethylation in rheumatoid arthritis synovial fibroblasts. Arthritis Rheum 60:3613–3622

    PubMed  CAS  Google Scholar 

  125. Maxwell DB, Grotendorst CA, Grotendorst GR, LeRoy EC (1987) Fibroblast heterogeneity in scleroderma: Clq studies. J Rheumatol 14:756–759

    PubMed  CAS  Google Scholar 

  126. Wang Y, Fan PS, Kahaleh B (2006) Association between enhanced type i collagen expression and epigenetic repression of the fli1 gene in scleroderma fibroblasts. Arthritis Rheum 54:2271–2279

    PubMed  CAS  Google Scholar 

  127. Kuchen S, Seemayer CA, Rethage J, von Knoch R, Kuenzler P, Beat AM, Gay RE, Gay S, Neidhart M (2004) The l1 retroelement-related p40 protein induces p38delta map kinase. Autoimmunity 37:57–65

    PubMed  CAS  Google Scholar 

  128. Richardson B, Scheinbart L, Strahler J, Gross L, Hanash S, Johnson M (1990) Evidence for impaired t cell DNA methylation in systemic lupus erythematosus and rheumatoid arthritis. Arthritis Rheum 33:1665–1673

    PubMed  CAS  Google Scholar 

  129. Daneman D (2006) Type 1 diabetes. Lancet 367:847–858

    PubMed  CAS  Google Scholar 

  130. Liu X, Invernizzi P, Lu Y, Kosoy R, Lu Y, Bianchi I, Podda M, Xu C, Xie G, Macciardi F, Selmi C, Lupoli S, Shigeta R, Ransom M, Lleo A, Lee AT, Mason AL, Myers RP, Peltekian KM, Ghent CN, Bernuzzi F, Zuin M, I.C.f.P.B.C.G. ICPBCG, Gregersen PK, Heathcote EJ, Hirschfield GM, Siminovitch KA, Amos CI, Gershwin ME, Seldin MF (2010) Genome-wide meta-analyses identifies three loci associated with primary biliary cirrhosis. Nat Genet in press

  131. Invernizzi P, Selmi C, Mackay IR, Podda M, Gershwin ME (2005) From bases to basis: Linking genetics to causation in primary biliary cirrhosis. Clin Gastroenterol Hepatol 3:401–410

    PubMed  CAS  Google Scholar 

  132. Selmi C, Bowlus CL, Gershwin ME, Coppel RL (2011) Primary biliary cirrhosis. Lancet 377:1600–1609

    PubMed  CAS  Google Scholar 

  133. Selmi C, Invernizzi P, Miozzo M, Podda M, Gershwin ME (2004) Primary biliary cirrhosis: Does x mark the spot? Autoimmun Rev 3:493–499

    PubMed  CAS  Google Scholar 

  134. Mitchell MM, Lleo A, Zammataro L, Mayo MJ, Invernizzi P, Bach N, Shimoda S, Gordon SC, Podda M, Gershwin ME, Selmi C, Lasalle JM (2011) Epigenetic investigation of variably x chromosome inactivated genes in monozygotic female twins discordant for primary biliary cirrhosis. Epigenetics 6

  135. Invernizzi P, Miozzo M, Battezzati PM, Bianchi I, Grati FR, Simoni G, Selmi C, Watnik M, Gershwin ME, Podda M (2004) Frequency of monosomy x in women with primary biliary cirrhosis. Lancet 363:533–535

    PubMed  Google Scholar 

  136. Aota N, Shiohara T (2009) Viral connection between drug rashes and autoimmune diseases: How autoimmune responses are generated after resolution of drug rashes. Autoimmun Rev 8:488–494

    PubMed  CAS  Google Scholar 

  137. Poole BD, Templeton AK, Guthridge JM, Brown EJ, Harley JB, James JA (2009) Aberrant epstein-barr viral infection in systemic lupus erythematosus. Autoimmun Rev 8:337–342

    PubMed  CAS  Google Scholar 

  138. Intlekofer AM, Banerjee A, Takemoto N, Gordon SM, Dejong CS, Shin H, Hunter CA, Wherry EJ, Lindsten T, Reiner SL (2008) Anomalous type 17 response to viral infection by cd8+ t cells lacking t-bet and eomesodermin. Science 321:408–411

    PubMed  CAS  Google Scholar 

  139. Kain R, Exner M, Brandes R, Ziebermayr R, Cunningham D, Alderson CA, Davidovits A, Raab I, Jahn R, Ashour O, Spitzauer S, Sunder-Plassmann G, Fukuda M, Klemm P, Rees AJ, Kerjaschki D (2008) Molecular mimicry in pauci-immune focal necrotizing glomerulonephritis. Nat Med 14:1088–1096

    PubMed  CAS  Google Scholar 

  140. Teijaro JR, Njau MN, Verhoeven D, Chandran S, Nadler SG, Hasday J, Farber DL (2009) Costimulation modulation uncouples protection from immunopathology in memory t cell responses to influenza virus. J Immunol 182:6834–6843

    PubMed  CAS  Google Scholar 

  141. Pierer M, Schulz A, Rossol M, Kendzia E, Kyburz D, Haentzschel H, Baerwald C, Wagner U (2009) Herpesvirus entry mediator-ig treatment during immunization aggravates rheumatoid arthritis in the collagen-induced arthritis model. J Immunol 182:3139–3145

    PubMed  CAS  Google Scholar 

  142. Fairweather D, Cihakova D (2009) Alternatively activated macrophages in infection and autoimmunity. J Autoimmun 33:222–230

    Google Scholar 

  143. Frazer IH (2008) Autoimmunity and persistent viral infection: Two sides of the same coin? J Autoimmun 31:131–135

    Google Scholar 

  144. Shi J, Sun X, Zhao Y, Zhao J, Li Z (2008) Prevalence and significance of antibodies to citrullinated human papilloma virus-47 e2(345–362) in rheumatoid arthritis. J Autoimmun

  145. Khan N, Jeffers M, Kumar S, Hackett C, Boldog F, Khramtsov N, Qian X, Mills E, Berghs SC, Carey N, Finn PW, Collins LS, Tumber A, Ritchie JW, Jensen PB, Lichenstein HS, Sehested M (2008) Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors. Biochem J 409:581–589

    PubMed  CAS  Google Scholar 

  146. Dinarello CA, Fossati G, Mascagni P (2011) Histone deacetylase inhibitors for treating a spectrum of diseases not related to cancer. Mol Med 17:333–352

    PubMed  CAS  Google Scholar 

  147. Chen S, Sang N (2011) Histone deacetylase inhibitors: The epigenetic therapeutics that repress hypoxia-inducible factors. J Biomed Biotechnol 2011:197946

    PubMed  Google Scholar 

  148. Leoni F, Fossati G, Lewis EC, Lee JK, Porro G, Pagani P, Modena D, Moras ML, Pozzi P, Reznikov LL, Siegmund B, Fantuzzi G, Dinarello CA, Mascagni P (2005) The histone deacetylase inhibitor itf2357 reduces production of pro-inflammatory cytokines in vitro and systemic inflammation in vivo. Mol Med 11:1–15

    PubMed  CAS  Google Scholar 

  149. Leoni F, Zaliani A, Bertolini G, Porro G, Pagani P, Pozzi P, Dona G, Fossati G, Sozzani S, Azam T, Bufler P, Fantuzzi G, Goncharov I, Kim SH, Pomerantz BJ, Reznikov LL, Siegmund B, Dinarello CA, Mascagni P (2002) The antitumor histone deacetylase inhibitor suberoylanilide hydroxamic acid exhibits antiinflammatory properties via suppression of cytokines. Proc Natl Acad Sci U S A 99:2995–3000

    PubMed  CAS  Google Scholar 

  150. Song W, Tai YT, Tian Z, Hideshima T, Chauhan D, Nanjappa P, Exley MA, Anderson KC, Munshi NC (2011) Hdac inhibition by lbh589 affects the phenotype and function of human myeloid dendritic cells. Leukemia 25:161–168

    PubMed  CAS  Google Scholar 

  151. Bosisio D, Vulcano M, Del Prete A, Sironi M, Salvi V, Salogni L, Riboldi E, Leoni F, Dinarello CA, Girolomoni G, Sozzani S (2008) Blocking th17-polarizing cytokines by histone deacetylase inhibitors in vitro and in vivo. J Leukoc Biol 84:1540–1548

    PubMed  CAS  Google Scholar 

  152. Crazzolara R, Johrer K, Johnstone RW, Greil R, Kofler R, Meister B, Bernhard D (2002) Histone deacetylase inhibitors potently repress cxcr4 chemokine receptor expression and function in acute lymphoblastic leukaemia. Br J Haematol 119:965–969

    PubMed  CAS  Google Scholar 

  153. Chen L, Fischle W, Verdin E, Greene WC (2001) Duration of nuclear nf-kappab action regulated by reversible acetylation. Science 293:1653–1657

    CAS  Google Scholar 

  154. Choi S, Reddy P (2011) Hdac inhibition and graft versus host disease. Mol Med 17:404–416

    PubMed  CAS  Google Scholar 

  155. Grabiec AM, Krausz S, de Jager W, Burakowski T, Groot D, Sanders ME, Prakken BJ, Maslinski W, Eldering E, Tak PP, Reedquist KA (2010) Histone deacetylase inhibitors suppress inflammatory activation of rheumatoid arthritis patient synovial macrophages and tissue. J Immunol 184:2718–2728

    PubMed  CAS  Google Scholar 

  156. Joosten LA, Leoni F, Meghji S, Mascagni P (2011) Inhibition of hdac activity by itf2357 ameliorates joint inflammation and prevents cartilage and bone destruction in experimental arthritis. Mol Med 17:391–396

    PubMed  CAS  Google Scholar 

  157. Vojinovic J, Damjanov N (2011) Hdac inhibition in rheumatoid arthritis and juvenile idiopathic arthritis. Mol Med 17:397–403

    PubMed  CAS  Google Scholar 

  158. Reilly CM, Regna N, Mishra N (2011) Hdac inhibition in lupus models. Mol Med 17:417–425

    PubMed  CAS  Google Scholar 

  159. Larsen L, Tonnesen M, Ronn SG, Storling J, Jorgensen S, Mascagni P, Dinarello CA, Billestrup N, Mandrup-Poulsen T (2007) Inhibition of histone deacetylases prevents cytokine-induced toxicity in beta cells. Diabetologia 50:779–789

    PubMed  CAS  Google Scholar 

  160. Vojinovic J, Damjanov N, D'Urzo C, Furlan A, Susic G, Pasic S, Iagaru N, Stefan M, Dinarello CA (2011) Safety and efficacy of an oral histone deacetylase inhibitor in systemic-onset juvenile idiopathic arthritis. Arthritis Rheum 63:1452–1458

    PubMed  CAS  Google Scholar 

  161. Mizuno S, Yasuo M, Bogaard HJ, Kraskauskas D, Natarajan R, Voelkel NF (2011) Inhibition of histone deacetylase causes emphysema. Am J Physiol Lung Cell Mol Physiol 300:L402–413

    PubMed  CAS  Google Scholar 

  162. Bishton MJ, Harrison SJ, Martin BP, McLaughlin N, James C, Josefsson EC, Henley KJ, Kile BT, Prince HM, Johnstone RW (2011) Deciphering the molecular and biologic processes that mediate histone deacetylase inhibitor-induced thrombocytopenia. Blood 117:3658–3668

    PubMed  CAS  Google Scholar 

  163. Gowher H, Jeltsch A (2004) Mechanism of inhibition of DNA methyltransferases by cytidine analogs in cancer therapy. Cancer Biol Ther 3:1062–1068

    PubMed  CAS  Google Scholar 

  164. Baer-Dubowska W, Majchrzak-Celinska A, Cichocki M (2011) Pharmocoepigenetics: A new approach to predicting individual drug responses and targeting new drugs. Pharmacol Rep 63:293–304

    PubMed  CAS  Google Scholar 

  165. Ingelman-Sundberg M, Sim SC, Gomez A, Rodriguez-Antona C (2007) Influence of cytochrome p450 polymorphisms on drug therapies: Pharmacogenetic, pharmacoepigenetic and clinical aspects. Pharmacol Ther 116:496–526

    PubMed  CAS  Google Scholar 

  166. Kusaba H, Nakayama M, Harada T, Nomoto M, Kohno K, Kuwano M, Wada M (1999) Association of 5' cpg demethylation and altered chromatin structure in the promoter region with transcriptional activation of the multidrug resistance 1 gene in human cancer cells. Eur J Biochem 262:924–932

    PubMed  CAS  Google Scholar 

  167. Park JY, Helm JF, Zheng W, Ly QP, Hodul PJ, Centeno BA, Malafa MP (2008) Silencing of the candidate tumor suppressor gene solute carrier family 5 member 8 (slc5a8) in human pancreatic cancer. Pancreas 36:e32–39

    PubMed  CAS  Google Scholar 

  168. Youssef EM, Lotan D, Issa JP, Wakasa K, Fan YH, Mao L, Hassan K, Feng L, Lee JJ, Lippman SM, Hong WK, Lotan R (2004) Hypermethylation of the retinoic acid receptor-beta(2) gene in head and neck carcinogenesis. Clin Cancer Res 10:1733–1742

    PubMed  CAS  Google Scholar 

  169. Bovenzi V, Momparler RL (2001) Antineoplastic action of 5-aza-2'-deoxycytidine and histone deacetylase inhibitor and their effect on the expression of retinoic acid receptor beta and estrogen receptor alpha genes in breast carcinoma cells. Cancer Chemother Pharmacol 48:71–76

    PubMed  CAS  Google Scholar 

  170. Virgin HW, Todd JA (2011) Metagenomics and personalized medicine. Cell 147:44–56

    PubMed  CAS  Google Scholar 

  171. Yan Q (2010) Immunoinformatics and systems biology methods for personalized medicine. Methods Mol Biol 662:203–220

    PubMed  CAS  Google Scholar 

  172. Esteller M (2008) Epigenetics in cancer. N Engl J Med 358:1148–1159

    PubMed  CAS  Google Scholar 

  173. Odenike O, Thirman MJ, Artz AS, Godley LA, Larson RA, Stock W (2011) Gene mutations, epigenetic dysregulation, and personalized therapy in myeloid neoplasia: Are we there yet? Semin Oncol 38:196–214

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carlo Selmi.

Rights and permissions

Reprints and permissions

About this article

Cite this article

De Santis, M., Selmi, C. The Therapeutic Potential of Epigenetics in Autoimmune Diseases. Clinic Rev Allerg Immunol 42, 92–101 (2012). https://doi.org/10.1007/s12016-011-8293-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12016-011-8293-8

Keywords

Navigation