Environmental Exposures and Autoimmune Diseases: Contribution of Gut Microbiome

Environmental agents have been gaining more attention in recent years for their role in the pathogenesis of autoimmune diseases (ADs). Increasing evidence has linked environmental exposures, including trichloroethene (TCE), silica, mercury, pristane, pesticides, and smoking to higher risk for ADs. However, potential mechanisms by which these environmental agents contribute to the disease pathogenesis remains largely unknown. Dysbiosis of the gut microbiome is another important environmental factor that has been linked to the onset of different ADs. Altered microbiota composition is associated with impaired intestinal barrier function and dysregulation of mucosal immune system, but it is unclear if gut dysbiosis is a causal factor or an outcome of ADs. In this review article, we first describe the recent epidemiological and mechanistic evidences linking environmental/occupational exposures with various ADs (especially SLE). Secondly, we discuss how changes in the gut microbiome composition (dysbiosis) could contribute to the disease pathogenesis, especially in response to exposure to environmental chemicals.

[1]  D. Kono,et al.  Mercury-induced inflammation and autoimmunity. , 2019, Biochimica et biophysica acta. General subjects.

[2]  P. Boor,et al.  Redox regulation of hepatic NLRP3 inflammasome activation and immune dysregulation in trichloroethene-mediated autoimmunity. , 2019, Free radical biology & medicine.

[3]  A. Alekseyenko,et al.  A Link Between Plasma Microbial Translocation, Microbiome, and Autoantibody Development in First‐Degree Relatives of Systemic Lupus Erythematosus Patients , 2019, Arthritis & rheumatology.

[4]  Mengtao Li,et al.  Gut microbiota promote the inflammatory response in the pathogenesis of systemic lupus erythematosus , 2019, Molecular Medicine.

[5]  S. Berndt,et al.  Lifetime Pesticide Use and Antinuclear Antibodies in Male Farmers From the Agricultural Health Study , 2019, Front. Immunol..

[6]  Yasser B. Ruiz-Blanco,et al.  Control of TLR7-mediated type I IFN signaling in pDCs through CXCR4 engagement—A new target for lupus treatment , 2019, Science Advances.

[7]  D. Ye,et al.  Emerging role of air pollution in autoimmune diseases. , 2019, Autoimmunity reviews.

[8]  Bin Gao,et al.  Fructose Promotes Leaky Gut, Endotoxemia and Liver Fibrosis through CYP2E1‐Mediated Oxidative and Nitrative Stress , 2019 .

[9]  L. Kelly,et al.  Bringing microbiome-drug interaction research into the clinic , 2019, EBioMedicine.

[10]  G. Fahey,et al.  Perspective: Physiologic Importance of Short-Chain Fatty Acids from Nondigestible Carbohydrate Fermentation , 2019, Advances in nutrition.

[11]  C. Dehner,et al.  The microbiome in systemic autoimmune disease: mechanistic insights from recent studies , 2019, Current opinion in rheumatology.

[12]  I. Barshack,et al.  Helminth-Based Product and the Microbiome of Mice with Lupus , 2019, mSystems.

[13]  B. Rovin,et al.  Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal , 2019, Annals of the rheumatic diseases.

[14]  E. Gianchecchi,et al.  Recent Advances on Microbiota Involvement in the Pathogenesis of Autoimmunity , 2019, International journal of molecular sciences.

[15]  Y. Shoenfeld,et al.  The microbiome in autoimmune diseases , 2019, Clinical and experimental immunology.

[16]  M. Mahmoudi,et al.  Amelioration of regulatory T cells by Lactobacillus delbrueckii and Lactobacillus rhamnosus in pristane‐induced lupus mice model , 2018, Journal of cellular physiology.

[17]  K. Gokulan,et al.  Irreversible effects of trichloroethylene on the gut microbial community and gut‐associated immune responses in autoimmune‐prone mice , 2018, Journal of applied toxicology : JAT.

[18]  Eric P. Skaar,et al.  Nonsteroidal Anti-inflammatory Drugs Alter the Microbiota and Exacerbate Clostridium difficile Colitis while Dysregulating the Inflammatory Response , 2018, mBio.

[19]  M. Bastami,et al.  The role of microbiota in the pathogenesis of lupus: Dose it impact lupus nephritis? , 2019, Pharmacological research.

[20]  Gangduo Wang,et al.  Contribution of poly(ADP‐ribose)polymerase‐1 activation and apoptosis in trichloroethene‐mediated autoimmunity , 2019, Toxicology and applied pharmacology.

[21]  N. Starobinas,et al.  miRNA Expression and Interaction with Genes Involved in Susceptibility to Pristane-Induced Arthritis , 2018, Journal of immunology research.

[22]  L. Mollet,et al.  STING-dependent sensing of self-DNA drives silica-induced lung inflammation , 2018, Nature Communications.

[23]  Ç. Tükel,et al.  Curli-Containing Enteric Biofilms Inside and Out: Matrix Composition, Immune Recognition, and Disease Implications , 2018, Microbiology and Molecular Biology Reviews.

[24]  Trevor O. Kirby,et al.  The Gut Microbiome in Multiple Sclerosis: A Potential Therapeutic Avenue , 2018, Medical sciences.

[25]  M. Mahmoudi,et al.  In vivo study: Th1–Th17 reduction in pristane‐induced systemic lupus erythematosus mice after treatment with tolerogenic Lactobacillus probiotics , 2018, Journal of cellular physiology.

[26]  Xin M. Luo,et al.  Retinoic Acid, Leaky Gut, and Autoimmune Diseases , 2018, Nutrients.

[27]  P. Söderkvist,et al.  Bank1 and NF-kappaB as key regulators in anti-nucleolar antibody development , 2018, PloS one.

[28]  E. Volkmann,et al.  Update on the Gastrointestinal Microbiome in Systemic Sclerosis , 2018, Current Rheumatology Reports.

[29]  K. Costenbader,et al.  Cigarette smoking and the pathogenesis of systemic lupus erythematosus , 2018, Expert review of clinical immunology.

[30]  Xiong Ma,et al.  The microbiome and autoimmunity: a paradigm from the gut–liver axis , 2018, Cellular & Molecular Immunology.

[31]  D. Kono,et al.  Silicosis and Silica-Induced Autoimmunity in the Diversity Outbred Mouse , 2018, Front. Immunol..

[32]  Z. Savin,et al.  Smoking and the intestinal microbiome , 2018, Archives of Microbiology.

[33]  V. Tripathi,et al.  Smoking under hypoxic conditions: a potent environmental risk factor for inflammatory and autoimmune diseases , 2018, Military Medical Research.

[34]  D. Jain,et al.  Translocation of a gut pathobiont drives autoimmunity in mice and humans , 2018, Science.

[35]  Nicholas J. Carruthers,et al.  Low level Hg2+ exposure modulates the B-cell cytoskeletal phosphoproteome. , 2018, Journal of proteomics.

[36]  Q. Lu,et al.  IL-6/STAT3 pathway induced deficiency of RFX1 contributes to Th17-dependent autoimmune diseases via epigenetic regulation , 2018, Nature Communications.

[37]  G. Radford-Smith,et al.  Chronic cigarette smoke exposure induces systemic hypoxia that drives intestinal dysfunction. , 2018, JCI insight.

[38]  Gangduo Wang,et al.  Environmental Agents, Oxidative Stress and Autoimmunity. , 2018, Current opinion in toxicology.

[39]  J. Clemente,et al.  The role of the gut microbiome in systemic inflammatory disease , 2018, British Medical Journal.

[40]  Rabi Yacoub,et al.  Lupus: The microbiome angle. , 2017, Immunobiology.

[41]  B. Beutler,et al.  Induction of Systemic Autoimmunity by a Xenobiotic Requires Endosomal TLR Trafficking and Signaling from the Late Endosome and Endolysosome but Not Type I IFN , 2017, The Journal of Immunology.

[42]  Yang Yu,et al.  Gut Microbiota in Human Systemic Lupus Erythematosus and a Mouse Model of Lupus , 2017, Applied and Environmental Microbiology.

[43]  C. Rosenfeld Gut Dysbiosis in Animals Due to Environmental Chemical Exposures , 2017, Front. Cell. Infect. Microbiol..

[44]  Aline Zazeri Leite,et al.  Intestinal dysbiosis and probiotic applications in autoimmune diseases , 2017, Immunology.

[45]  J. Irudayaraj,et al.  Toxicological effects of trichloroethylene exposure on immune disorders , 2017, Immunopharmacology and immunotoxicology.

[46]  G. Katona,et al.  Survivin in autoimmune diseases. , 2017, Autoimmunity reviews.

[47]  Nicholas J. Carruthers,et al.  Mercury alters endogenous phosphorylation profiles of SYK in murine B cells , 2017, BMC Immunology.

[48]  Xiaofeng Liao,et al.  Control of lupus nephritis by changes of gut microbiota , 2017, Microbiome.

[49]  Vaishali R. Moulton,et al.  Pathogenesis of Human Systemic Lupus Erythematosus: A Cellular Perspective. , 2017, Trends in molecular medicine.

[50]  Chunbao Guo,et al.  Fecal microbiota transplantation (FMT) could reverse the severity of experimental necrotizing enterocolitis (NEC) via oxidative stress modulation , 2017, Free radical biology & medicine.

[51]  S. Bhattacharyya,et al.  Trichloroethylene-induced alterations in DNA methylation were enriched in polycomb protein binding sites in effector/memory CD4+ T cells , 2017, Environmental epigenetics.

[52]  James L. Richards,et al.  Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes , 2017, Nature Immunology.

[53]  T. Otsuki,et al.  Silicosis and autoimmunity , 2017, Current opinion in allergy and clinical immunology.

[54]  A. Neish,et al.  Redox signaling mediated by the gut microbiota. , 2017, Free radical biology & medicine.

[55]  Ernest Y Lee,et al.  Bacterial amyloid curli acts as a carrier for DNA to elicit an autoimmune response via TLR2 and TLR9 , 2017, PLoS pathogens.

[56]  A. Xu,et al.  Low-dose penicillin exposure in early life decreases Th17 and the susceptibility to DSS colitis in mice through gut microbiota modification , 2017, Scientific Reports.

[57]  A. Bossios,et al.  Smoking activates cytotoxic CD8+ T cells and causes survivin release in rheumatoid arthritis. , 2017, Journal of autoimmunity.

[58]  Alexa R. Weingarden,et al.  Intestinal microbiota, fecal microbiota transplantation, and inflammatory bowel disease , 2017, Gut microbes.

[59]  S. Paira,et al.  Different end-points to assess effects in systemic lupus erythematosus patients exposed to pesticide mixtures. , 2017, Toxicology.

[60]  J. Strain,et al.  Mercury as an environmental stimulus in the development of autoimmunity - A systematic review. , 2017, Autoimmunity reviews.

[61]  Y. Saeki,et al.  Dysbiosis Contributes to Arthritis Development via Activation of Autoreactive T Cells in the Intestine , 2016, Arthritis & rheumatology.

[62]  P. Jafari,et al.  Clinical and metabolic response to probiotic supplementation in patients with rheumatoid arthritis: a randomized, double‐blind, placebo‐controlled trial , 2016, International journal of rheumatic diseases.

[63]  A. Margolles,et al.  Th17 responses and natural IgM antibodies are related to gut microbiota composition in systemic lupus erythematosus patients , 2016, Scientific Reports.

[64]  Y. Shoenfeld,et al.  Smoke and autoimmunity: The fire behind the disease. , 2016, Autoimmunity reviews.

[65]  K. Pollard Silica, Silicosis, and Autoimmunity , 2016, Front. Immunol..

[66]  Olli Simell,et al.  Association of Early Exposure of Probiotics and Islet Autoimmunity in the TEDDY Study. , 2016, JAMA pediatrics.

[67]  G. Ansari,et al.  iNOS null MRL+/+ mice show attenuation of trichloroethene-mediated autoimmunity: contribution of reactive nitrogen species and lipid-derived reactive aldehydes. , 2015, Free radical biology & medicine.

[68]  Husen Zhang,et al.  SLE: Another Autoimmune Disorder Influenced by Microbes and Diet? , 2015, Front. Immunol..

[69]  R. Gold,et al.  Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine. , 2015, Immunity.

[70]  Christopher D. Scharer,et al.  Lactobacilli Modulate Epithelial Cytoprotection through the Nrf2 Pathway. , 2015, Cell reports.

[71]  M. Goulian,et al.  Amyloid-DNA Composites of Bacterial Biofilms Stimulate Autoimmunity. , 2015, Immunity.

[72]  Y. Shoenfeld,et al.  Unraveling the Hygiene Hypothesis of helminthes and autoimmunity: origins, pathophysiology, and clinical applications , 2015, BMC Medicine.

[73]  K. Pollard,et al.  Environmental Triggers and Autoimmunity , 2014, Autoimmune diseases.

[74]  F. Turroni,et al.  Intestinal Dysbiosis Associated with Systemic Lupus Erythematosus , 2014, mBio.

[75]  Xiaofeng Liao,et al.  Dynamics of Gut Microbiota in Autoimmune Lupus , 2014, Applied and Environmental Microbiology.

[76]  Todd J. Zurlinden,et al.  Modeling toxicodynamic effects of trichloroethylene on liver in mouse model of autoimmune hepatitis. , 2014, Toxicology and applied pharmacology.

[77]  Ling Lin,et al.  Effects of Pristane Alone or Combined with Chloroquine on Macrophage Activation, Oxidative Stress, and Th1/Th2 Skewness , 2014, Journal of immunology research.

[78]  A. Gomes,et al.  Gut microbiota, probiotics and diabetes , 2014, Nutrition Journal.

[79]  G. Ansari,et al.  Nitrosative Stress and Nitrated Proteins in Trichloroethene-Mediated Autoimmunity , 2014, PloS one.

[80]  S. Holdsworth,et al.  Endogenous interleukin (IL)‐17A promotes pristane‐induced systemic autoimmunity and lupus nephritis induced by pristane , 2014, Clinical and experimental immunology.

[81]  Angela M. Yu,et al.  The SLE Transcriptome Exhibits Evidence of Chronic Endotoxin Exposure and Has Widespread Dysregulation of Non-Coding and Coding RNAs , 2014, PloS one.

[82]  A. Vojdani A Potential Link between Environmental Triggers and Autoimmunity , 2014, Autoimmune diseases.

[83]  J. Lambeth,et al.  Nox enzymes and new thinking on reactive oxygen: a double-edged sword revisited. , 2014, Annual review of pathology.

[84]  A. Neish,et al.  Redox signaling mediates symbiosis between the gut microbiota and the intestine , 2014, Gut microbes.

[85]  Wenhua Zhu,et al.  MicroRNA-26a negatively regulates toll-like receptor 3 expression of rat macrophages and ameliorates pristane induced arthritis in rats , 2014, Arthritis Research & Therapy.

[86]  R. Medzhitov,et al.  The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition , 2014, Proceedings of the National Academy of Sciences.

[87]  J. Lambeth,et al.  Symbiotic lactobacilli stimulate gut epithelial proliferation via Nox‐mediated generation of reactive oxygen species , 2013, The EMBO journal.

[88]  G. Ansari,et al.  N-Acetylcysteine protects against trichloroethene-mediated autoimmunity by attenuating oxidative stress. , 2013, Toxicology and applied pharmacology.

[89]  Curtis Huttenhower,et al.  Biodiversity and functional genomics in the human microbiome. , 2013, Trends in genetics : TIG.

[90]  Q. Lu,et al.  Heritability versus the role of the environment in autoimmunity. , 2012, Journal of autoimmunity.

[91]  G. Ansari,et al.  Protein adducts of malondialdehyde and 4-hydroxynonenal contribute to trichloroethene-mediated autoimmunity via activating Th17 cells: dose- and time-response studies in female MRL+/+ mice. , 2012, Toxicology.

[92]  R. Fuleihan,et al.  The hygiene hypothesis revisited: does exposure to infectious agents protect us from allergy? , 2012, Current opinion in pediatrics.

[93]  B. Diamond,et al.  Current and novel therapeutics in the treatment of systemic lupus erythematosus , 2011 .

[94]  G. Zandman-Goddard,et al.  Drug-induced lupus: an update. , 2010, Autoimmunity reviews.

[95]  D. Kono,et al.  Toxicology of autoimmune diseases. , 2010, Chemical research in toxicology.

[96]  A. Perl,et al.  Infection in systemic lupus erythematosus: friend or foe? , 2010, International journal of clinical rheumatology.

[97]  M. Satoh,et al.  Induction of autoimmunity by pristane and other naturally occurring hydrocarbons. , 2009, Trends in immunology.

[98]  Gangduo Wang,et al.  Increased nitration and carbonylation of proteins in MRL+/+ mice exposed to trichloroethene: potential role of protein oxidation in autoimmunity. , 2009, Toxicology and applied pharmacology.

[99]  J. Fuscoe,et al.  Delineating liver events in trichloroethylene-induced autoimmune hepatitis. , 2009, Chemical research in toxicology.

[100]  S. Makris,et al.  Evidence of Autoimmune-Related Effects of Trichloroethylene Exposure from Studies in Mice and Humans , 2009, Environmental health perspectives.

[101]  S. Roberts,et al.  Comparison of chlordecone and estradiol effects on splenic T-cells in (NZBxNZW)F(1) mice. , 2008, Toxicology letters.

[102]  Richard A. Flavell,et al.  The Nalp3 inflammasome is essential for the development of silicosis , 2008, Proceedings of the National Academy of Sciences.

[103]  E. Ogier-Denis,et al.  NOX enzymes and Toll-like receptor signaling , 2008, Seminars in Immunopathology.

[104]  J. Tschopp,et al.  Innate Immune Activation Through Nalp3 Inflammasome Sensing of Asbestos and Silica , 2008, Science.

[105]  G. Ansari,et al.  Chronic exposure to trichloroethene causes early onset of SLE-like disease in female MRL +/+ mice. , 2008, Toxicology and applied pharmacology.

[106]  G. Ansari,et al.  Involvement of Lipid Peroxidation-Derived Aldehyde–protein Adducts in Autoimmunity Mediated by Trichloroethene , 2007, Journal of toxicology and environmental health. Part A.

[107]  G. Ansari,et al.  Autoimmune response in MRL+/+ mice following treatment with dichloroacetyl chloride or dichloroacetic anhydride. , 2006, Toxicology and applied pharmacology.

[108]  H. Sakaguchi,et al.  Reduced Function of CD4+25+ Regulatory T Cell Fraction in Silicosis Patients , 2006, International journal of immunopathology and pharmacology.

[109]  L. Criswell,et al.  Association of smoking with dsDNA autoantibody production in systemic lupus erythematosus , 2005, Annals of the rheumatic diseases.

[110]  J. Gaughan,et al.  A Role for B Cell-Activating Factor of the TNF Family in Chemically Induced Autoimmunity1 , 2005, The Journal of Immunology.

[111]  F. Silvestris,et al.  Induction of Apoptosis by the Hydrocarbon Oil Pristane: Implications for Pristane-Induced Lupus1 , 2005, The Journal of Immunology.

[112]  W. Fujimoto,et al.  Detection, epitope‐mapping and function of anti‐Fas autoantibody in patients with silicosis , 2005, Immunology.

[113]  B. Croker,et al.  Acceleration of Autoimmunity by Organochlorine Pesticides in (NZB × NZW)F1 Mice , 2004, Environmental health perspectives.

[114]  N. Pumford,et al.  Activation and attenuation of apoptosis of CD4+ T cells following in vivo exposure to two common environmental toxicants, trichloroacetaldehyde hydrate and trichloroacetic acid. , 2004, Journal of autoimmunity.

[115]  J. Lambeth NOX enzymes and the biology of reactive oxygen , 2004, Nature Reviews Immunology.

[116]  A. Holian,et al.  Silica-exposed mice generate autoantibodies to apoptotic cells. , 2004, Toxicology.

[117]  Thomas R. Riley,et al.  A Randomized Double-blind Placebo-controlled Trial , 2004 .

[118]  A. Holian,et al.  Silica accelerated systemic autoimmune disease in lupus‐prone New Zealand mixed mice , 2003, Clinical and experimental immunology.

[119]  I. Shapiro,et al.  Mercury-induced apoptosis in human lymphocytes: caspase activation is linked to redox status. , 2002, Antioxidants & redox signaling.

[120]  H. Lührs,et al.  Butyrate Inhibits NF-κB Activation in Lamina Propria Macrophages of Patients with Ulcerative Colitis , 2002, Scandinavian journal of gastroenterology.

[121]  B. Croker,et al.  Interferon- (cid:1) is required for lupus nephritis in mice treated with the hydrocarbon oil pristane , 2010 .

[122]  G. Ansari,et al.  Anti-malondialdehyde antibodies in MRL+/+ mice treated with trichloroethene and dichloroacetyl chloride: possible role of lipid peroxidation in autoimmunity. , 2001, Toxicology and applied pharmacology.

[123]  B. Croker,et al.  Interferon-gamma is required for lupus nephritis in mice treated with the hydrocarbon oil pristane. , 2001, Kidney international.

[124]  N. Pumford,et al.  Trichloroethylene accelerates an autoimmune response by Th1 T cell activation in MRL +/+ mice. , 2000, Immunopharmacology.

[125]  Y. Phillips,et al.  Prevalence of Pulmonary Disorders in Patients with Newly Diagnosed Rheumatoid Arthritis , 2000, Clinical Rheumatology.

[126]  N. Pumford,et al.  Trichloroethylene activates CD4+ T cells: potential role in an autoimmune response. , 1999, Drug metabolism reviews.

[127]  V. Poli,et al.  Interleukin 6 Dependence of Anti-DNA Antibody Production: Evidence for Two Pathways of Autoantibody Formation in Pristane-induced Lupus , 1998, The Journal of experimental medicine.

[128]  I. Shapiro,et al.  Low-level methylmercury exposure causes human T-cells to undergo apoptosis: evidence of mitochondrial dysfunction. , 1998, Environmental research.

[129]  G. Ansari,et al.  Trichloroethene-induced autoimmune response in female MRL +/+ mice. , 1995, Toxicology and applied pharmacology.

[130]  C. Elson,et al.  Susceptibility to pristane-induced arthritis is altered with changes in bowel flora. , 1993, Immunology letters.

[131]  R. Madison,et al.  Immunologic abnormalities in humans exposed to chlorpyrifos: preliminary observations. , 1993, Archives of environmental health.

[132]  K. Kilburn,et al.  Prevalence of symptoms of systemic lupus erythematosus (SLE) and of fluorescent antinuclear antibodies associated with chronic exposure to trichloroethylene and other chemicals in well water. , 1992, Environmental research.

[133]  K. Johnson An Update. , 1984, Journal of food protection.

[134]  Peilin Wu,et al.  Preliminary Observations , 1830, The Medico-chirurgical review.