Endocrine Disorders in Autoimmune Rheumatological Diseases: A Focus on Thyroid Autoimmune Diseases and on the Effects of Chronic Glucocorticoid Treatment
Abstract
1. Introduction
2. Thyroid Diseases
2.1. Genetic Factors
2.2. Environmental Factors
3. Effects of Chronic Glucocorticoid Treatment
3.1. Cushing’s Syndrome
3.2. Adrenal Insufficiency and Corticosteroids’ Withdrawal Syndrome
3.3. Is There a Safe GC Treatment Regimen?
3.4. Follow up of GC Therapy
4. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Goldblatt, F.; O’Neill, S.G. Clinical aspects of autoimmune rheumatic diseases. Lancet 2013, 382, 797–808. [Google Scholar] [CrossRef]
- AMRER. Onlus Malattie reumatiche, primo report sull’incidenza delle esenzioni per malattia. Quotid. Sanità 2013, 1, 3–7. [Google Scholar]
- Safiri, S.; Kolahi, A.A.; Hoy, D.; Smith, E.; Bettampadi, D.; Mansournia, M.A.; Almasi-Hashiani, A.; Ashrafi-Asgarabad, A.; Moradi-Lakeh, M.; Qorbani, M.; et al. Global, regional and national burden of rheumatoid arthritis 1990–2017: A systematic analysis of the Global Burden of Disease study 2017. Ann. Rheum. Dis. 2019, 78, 11. [Google Scholar] [CrossRef] [PubMed]
- WHO.int. Available online: https://www.who.int/chp/topics/rheumatic/en/ (accessed on 11 February 2021).
- Conigliaro, P.; D’Antonio, A.; Pinto, S.; Chimenti, M.S.; Triggianese, P.; Rotondi, M.; Perricone, R. Autoimmune thyroid disorders and rheumatoid arthritis: A bidirectional interplay. Autoimmun. Rev. 2020, 19, 102529. [Google Scholar] [CrossRef]
- Ringold, D.A.; Nicoloff, J.T.; Kesler, M.; Davis, H.; Hamilton, A.; Mack, T. Further evidence for a strong genetic influence on the development of autoimmune thyroid diseases: The California twin study. Thyroid 2002, 12, 647–653. [Google Scholar] [CrossRef]
- Torfs, C.P. Genetic interrelationship between insulin-dependent diabetes mellitus, the autoimmune thyroid diseases and rheumatoid arthritis. Am. J. Hum. Genet. 1986, 38, 170–187. [Google Scholar]
- Anaya, J.M. Common mechanisms of autoimmune diseases (the autoimmune tautology). Autoimmun. Rev. 2012, 11, 781–784. [Google Scholar] [CrossRef]
- Zaletel, K.; Gaberšček, S. Hashimoto’s Thyroiditis: From Genes to the Disease. Curr. Genom. 2011, 12, 576–588. [Google Scholar] [CrossRef]
- McIver, B.; Morris, J.C. The pathogenesis of Graves’ disease. Endocrinol. Metab. Clin. N. Am. 1998, 27, 73–89. [Google Scholar] [CrossRef]
- Cho, J.H.; Feldman, M. Heterogeneity of autoimmune diseases: Pathophysiologic insights from genetics and implications for new therapies. Nat. Med. 2015, 21, 730–738. [Google Scholar] [CrossRef]
- Medlineplus.gov. Available online: https://medlineplus.gov/genetics/gene/hla-drb1/ (accessed on 11 February 2021).
- Mangalam, A.K.; Taneja, V.; David, C.S. HLA Class II Molecules Influence Susceptibility vs Protection in Inflammatory Diseases by Determining the Cytokine Profile. J. Immunol. 2013, 190, 513–519. [Google Scholar] [CrossRef]
- Miyadera, H.; Tokunaga, K. Associations of human leukocyte antigens with autoimmune diseases: Challenges in identifying the mechanism. J. Hum. Genet. 2015, 60, 697–702. [Google Scholar] [CrossRef]
- Li, C.W.; Osman, R.; Menconi, F.; Concepcion, E.S.; Tomer, Y. Flexible peptide recognition by HLA-DR triggers specific autoimmune T-cell responses in autoimmune thyroiditis and diabetes. J. Immun. 2017, 76, 1–9. [Google Scholar] [CrossRef]
- Criswell, L.A.; Pfeiffer, K.A.; Lum, R.F.; Gonzales, B.; Novitzke, J.; Kern, M.; Moser, K.L.; Begovich, A.B.; Carlton, V.E.; Li, W.; et al. Analysis of families in the Multiple Autoimmune Disease Genetic Consortium (MADGC) Collection: The PTPN22 620W Allele Associates with Multiple Autoimmune Phenothypes. Am. J. Hum. Gen. 2005, 76, 561–571. [Google Scholar] [CrossRef]
- Zhebrun, D.; Kudryashova, Y.; Babenko, A.; Maslyansky, A.; Kunitskaya, N.; Popcova, D.; Klushina, A.; Grineva, E.; Kostareva, A.; Shlyakhto, E. Association of PTPN22 1858T/T genotype with type 1 diabetes, Graves’ disease but not with rheumatoid arthritis in Russian population. Aging 2011, 3, 368–373. [Google Scholar] [CrossRef]
- Mori, M.; Yamada, R.; Kobayashi, K.; Kawaida, R.; Yamamoto, K. Ethnic differences in allele frequency of autoimmune-disease-associated SNPs. J. Hum. Genet. 2005, 50, 264–266. [Google Scholar] [CrossRef]
- Tai, X.; Van Laethem, F.; Pobezinsky, L.; Guinter, T.; Sharrow, S.O.; Adams, A.; Granger, L.; Kruhlak, M.; Lindsten, T.; Thompson, C.B. Basis of CTLA-4 function in regulatory and conventional CD4+ T cells. Blood 2012, 119, 5155–5163. [Google Scholar] [CrossRef]
- Ueda, H.; Howson, J.M.; Esposito, L.; Heward, J.; Chamberlain, G.; Rainbow, D.B.; Hunter, K.M.; Smith, A.N.; Di Genova, G.; Herr, M.H.; et al. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 2003, 423, 506–511. [Google Scholar] [CrossRef]
- Cooper, J.D.; Simmonds, M.J.; Walker, N.M.; Burren, O.; Brand, O.J.; Guo, H.; Wallace, C.; Stevens, H.; Coleman, G. Seven newly identified loci for autoimmune thyroid disease. Hum. Mol. Genet. 2012, 21, 5202–5208. [Google Scholar] [CrossRef]
- Okada, Y.; Wu, D.; Trynka, G.; Raj, T.; Terao, C.; Ikari, K.; Kochi, Y.; Ohmura, K.; Suzuki, A.; Yoshida, S. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 2014, 506, 376–381. [Google Scholar] [CrossRef]
- Girotra, M.; Hansen, A.; Farooki, A.; Byun, D.J.; Min, L.; Creelan, B.C.; Callahan, M.K.; Atkins, M.B.; Sharon, E.; Antonia, S.J.; et al. The current understanding of the endocrine effects from immune checkpoint inhibitors and recommendations for management. JNCI Cancer Spectr. 2018, 2, 3. [Google Scholar] [CrossRef]
- Cappelli, L.C.; Gutierrez, A.K.; Baer, A.N.; Albayda, J.; Manno, R.L.; Haque, U.; Lipson, E.J.; Bleich, K.B.; Shah, A.A.; Naidoo, J.; et al. Inflammatory arthritis and sicca syndrome induced by nivolumab and ipilimumab. Ann. Rheum. Dis. 2017, 76, 43–50. [Google Scholar] [CrossRef]
- Maniu, C.; Kobe, C.; Schlaak, M.; Mauch, C.; Eming, S.A. Polymyalgia rheumatica occurring during treatment with ipilimumab. Eur. J. Dermatol. 2016, 26, 513–514. [Google Scholar] [CrossRef]
- Jaberg-Bentele, N.F.; Kunz, M.; Abuhammad, S.; Dummer, R. Flare up of rheumatoid arthritis by anti CTLA-4 antibody but not with anti PD-1 therapy in a patient with metastatic melanoma. Case Rep. Dermatol. 2017, 9, 65–68. [Google Scholar] [CrossRef]
- Kuehn, H.S.; Ouyang, W.; Lo, B.; Deenick, E.K.; Niemela, J.E.; Avery, D.T.; Schickel, J.N.; Tran, D.Q.; Stoddard, J.; Zhang, Y.; et al. Immune dysregulation in human subjects with heterozygous germline mutation in CTLA4. Science 2014, 345, 1623–1627. [Google Scholar] [CrossRef]
- Zeissig, S.; Petersen, B.S.; Tomczak, M.; Melum, E.; Huc-Claustre, E.; Dougan, S.K.; Laerdahl, J.K.; Stade, B.; Forster, M.; Schreiber, S.; et al. Early-Onset Crohn’s disease and autoimmunity associated with a variant in CTLA-4. Gut 2015, 64, 1889–1897. [Google Scholar] [CrossRef]
- Sun, D.; Heimall, J. Disorders of CTLA-4 expression, how they lead to CVID and dysregulated immune responses. Curr. Opin. Allergy Clin. Immunol. 2019, 19, 578–585. [Google Scholar] [CrossRef]
- Buckner, J.H. Mechanisms of impaired regulation by CD4+CD25+FOXP3+ regulatory T cells in human autoimmune diseases. Nat. Rev. Immunol. 2010, 10, 849–859. [Google Scholar] [CrossRef] [PubMed]
- Theofilopoulos, A.N.; Kono, D.H.; Baccala, R. The multiple pathways to autoimmunity. Nat. Immunol. 2017, 18, 716–724. [Google Scholar] [CrossRef] [PubMed]
- Inoue, Y. Aploinsufficiency of A20 causes autoinflammatory and autoimmune disorders. J. Allergy Clin. Immunol. 2018, 141, 1485–1488. [Google Scholar]
- Chen, J.; Li, Y.; Tian, Y.; Huang, C.; Li, D.; Zhong, Q.; Ma, X. Interaction between microbes and host intestinal health: Modulation by dietary nutrients and gut-brain-endocrine-immune axis. Curr. Protein Pept. Sci. 2015, 16, 592–603. [Google Scholar] [CrossRef] [PubMed]
- Cavaglieri, C.R.; Nishiyama, A.; Fernandes, L.C.; Curi, R.; Miles, E.A.; Calder, P.C. Differential effects of short-chain fatty acids on proliferation and production of pro- and anti-inflammatory cytokines by cultured lymphocytes. Life Sci. 2003, 73, 1683–1690. [Google Scholar] [CrossRef]
- Johnson-Henry, K.C.; Pinnell, L.J.; Waskow, A.M.; Irrazabal, T.; Martin, A.; Hausner, M.; Sherman, P.M. Short-Chain fructo-oligosaccharide and inulin modulate inflammatory responses and microbial communities in Caco2-bbe cells and in a mouse model of intestinal injury. J. Nutr. 2014, 144, 1725–1733. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Liu, M.; Cao, J.; Li, X.; Fan, D.; Xia, Y.; Lu, X.; Li, J.; Ju, D.; Zhao, H. The dynamic interplay between the gut microbiota and autoimmune diseases. J. Immunol. Res. 2019, 2019, 7546047. [Google Scholar] [CrossRef]
- Liang, B.; Mamula, M.J. Molecular mimicry and the role of B lymphocytes in the processing of autoantigens. Cell. Mol. Life Sci. 2000, 57, 561–568. [Google Scholar] [CrossRef]
- Lanzavecchia, A. How can cryptic epitopes trigger autoimmunity? J. Exp. Med. 1995, 181, 1945–1948. [Google Scholar] [CrossRef]
- Shor, D.B.; Orbach, H.; Boaz, M.; Altman, A.; Anaya, J.M.; Bizzaro, N.; Tincani, A.; Cervera, R.; Espinosa, G.; Stojanovich, L.; et al. Gastrointestinal-Associated autoantibodies in different autoimmune diseases. Am. J. Clin. Exp. Immunol. 2012, 1, 49–55. [Google Scholar]
- Zamani, B.; Golkar, H.R.; Farshbaf, S.; Emadi-Baygi, M.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akhavan, R.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z. Clinical and metabolic response to probiotic supplementation in patients with rheumatoid arthritis: A randomized, double-blind, placebo-controlled trial. Int. J. Rheum. Dis. 2016, 19, 869–879. [Google Scholar] [CrossRef]
- López, P.; De Paz, B.; Rodríguez-Carrio, J.; Hevia, A.; Sánchez, B.; Margolles, A.; Suárez, A. Th17 responses and natural IgM antibodies are related to gut microbiota composition in systemic lupus erythematosus patients. Sci. Rep. 2016, 6, 24072. [Google Scholar] [CrossRef]
- Zeng, J.; Peng, L.; Zheng, W.; Huang, F.; Zhang, N.; Wu, D.; Yang, Y. Fecal microbiota transplantation for rheumatoid arthritis: A case report. Clin. Case Rep. 2020, 9, 1–4. [Google Scholar]
- Mele, C.; Caputo, M.; Bisceglia, A.; Samà, M.T.; Zavattaro, M.; Aimaretti, G.; Pagano, L.; Prodam, F.; Marzullo, P. Immunomodulatory Effects of Vitamin D in Thyroid Diseases. Nutrients 2020, 12, 1444. [Google Scholar] [CrossRef]
- Muscogiuri, G.; Mitri, J.; Mathieu, C.; Badenhoop, K.; Tamer, G.; Orio, F.; Mezza, T.; Vieth, R.; Colao, A.; Pittas, A. Mechanisms in endocrinology: Vitamin D as a potential contributor in endocrine health and disease. Eur. J. Endocrinol. 2014, 171, R101–R110. [Google Scholar] [CrossRef]
- Kim, D. The Role of Vitamin D in Thyroid Diseases. Int. J. Mol. Sci. 2017, 18, 1949. [Google Scholar] [CrossRef]
- Adorini, L.; Penna, G. Control of autoimmune diseases by the vitamin D endocrine system. Nat. Clin. Pract. Rheumatol. 2008, 4, 404–412. [Google Scholar] [CrossRef]
- Cutolo, M.; Plebani, M.; Shoenfeld, Y.; Adorini, L.; Tincani, A. Vitamin D endocrine system and the immune response in rheumatic diseases. Vitam. Horm. 2011, 86, 327–351. [Google Scholar]
- Jin, D.; Wu, S.; Zhang, Y.G.; Lu, R.; Xia, Y.; Dong, H.; Sun, J. Lack of Vitamin D Receptor Causes Dysbiosis and Changes the Functions of the Murine Intestinal Microbiome. Clin. Ther. 2015, 37, 996–1009. [Google Scholar] [CrossRef]
- Wu, S.; Liao, A.P.; Xia, Y.; Li, Y.C.; Li, J.D.; Sartor, R.B.; Sun, J. Vitamin D receptor negatively regulates bacterial-stimulated NF-kappaB activity in intestine. Am. J. Pathol. 2010, 177, 686–697. [Google Scholar] [CrossRef]
- Jahani, R.; Fielding, K.A.; Chen, J.; Villa, C.R.; Castelli, L.M.; Ward, W.E.; Comelli, E.M. Low vitamin D status throughout life results in an inflammatory prone status but does not alter bone mineral or strength in healthy 3-month-old CD-1 male mice. Mol. Nutr. Food Res. 2014, 58, 1491–1501. [Google Scholar] [CrossRef]
- Naderpoor, N.; Mousa, A.; Fernanda Gomez Arango, L.; Barrett, H.L.; Dekker Nitert, M.; de Courten, B. Effect of Vitamin D Supplementation on Faecal Microbiota: A Randomised Clinical Trial. Nutrients 2019, 11, 2888. [Google Scholar] [CrossRef]
- Chassaing, B.; Vijay-Kumar, M.; Gewirtz, A.T. How diet can impact gut microbiota to promote or endanger health. Curr. Opin. Gastroenterol. 2017, 33, 417–421. [Google Scholar] [CrossRef]
- Suez, J.; Korem, T.; Zeevi, D.; Zilberman-Schapira, G.; Thaiss, C.A.; Maza, O.; Israeli, D.; Zmora, N.; Gilad, S.; Weinberger, A.; et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 2014, 514, 181–186. [Google Scholar] [CrossRef]
- Chassaing, B.; Koren, O.; Goodrich, J.K.; Poole, A.C.; Srinivasan, S.; Ley, R.E.; Gewirtz, A.T. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature 2015, 519, 92–96. [Google Scholar] [CrossRef]
- Go, W.Y.; Liu, X.; Roti, M.A.; Liu, F.; Ho, S.N. NFAT5/TonEBP mutant mice define osmotic stress as a critical feature of the lymphoid microenvironment. Proc. Natl. Acad. Sci. USA 2004, 101, 10673–10678. [Google Scholar] [CrossRef]
- Shapiro, L.; Dinarello, C.A. Osmotic regulation of cytokine synthesis in vitro. Proc. Natl. Acad. Sci. USA 1995, 92, 12230–12234. [Google Scholar] [CrossRef]
- Wu, C.; Yosef, N.; Thalhamer, T.; Zhu, C.; Xiao, S.; Kishi, Y.; Regev, A.; Kuchroo, V.K. Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1. Nature 2013, 496, 513–517. [Google Scholar] [CrossRef]
- Salgado, E.; Bes-Rastrollo, M.; de Irala, J.; Carmona, L.; Gomez-Reino, J.J. High sodium intake is associated with self-reported rheumatoid arthritis: A cross sectional and case control analysis within the SUN cohort. Medicine 2015, 94, e924. [Google Scholar] [CrossRef]
- Marouen, S.; du Cailar, G.; Audo, R.; Lukas, C.; Vial, G.; Tournadre, A.; Barrat, E.; Ribstein, J.; Combe, B.; Morel, J.; et al. Sodium excretion is higher in patients with rheumatoid arthritis than in matched controls. PLoS ONE 2017, 12, e0186157. [Google Scholar] [CrossRef]
- Sundstrom, B.; Johansson, I.; Rantapää-Dahlqvist, S. Interaction between dietay sodium and smoking increases the risk for rheumatoid arthritis: Results from a nested case-control study. Rheumatology 2015, 54, 487–493. [Google Scholar] [CrossRef]
- Pfau, J.C.; Sentissi, J.J.; Li, S.A.; Calderon-Garciduenas, L.; Brown, J.M.; Blake, D.J. Asbestos-induced autoimmunity in C57BL/6 mice. J. Immunotoxicol. 2008, 5, 129–137. [Google Scholar] [CrossRef]
- Brown, J.M.; Archer, A.J.; Pfau, J.C.; Holian, A. Silica accelerated systemic autoimmune disease in lupus-prone New Zealand mixed mice. Clin. Exp. Immunol. 2003, 131, 415–421. [Google Scholar] [CrossRef]
- Pfau, J.C.; Brown, J.M.; Holian, A. Silica-Exposed mice generate autoantibodies to apoptotic cells. Toxicology 2004, 195, 167–176. [Google Scholar] [CrossRef] [PubMed]
- Hamilton, R.F.; Thakur, S.A.; Holian, A. Silica binding and toxicity in alveolar macrophages. Free Radical Biol. Med. 2008, 44, 1246–1258. [Google Scholar] [CrossRef] [PubMed]
- Rasooly, L.; Burek, C.L.; Rose, N.R. Iodine-Induced autoimmune thyroiditis in NOD-H-2h4 mice. Clin. Immunol. Immunopathol. 1996, 81, 287–292. [Google Scholar] [CrossRef] [PubMed]
- Bournaud, C.; Orgiazzi, J.J. Iodine excess and thyroid autoimmunity. J. Endocrinol. Invest. 2003, 26, 49–56. [Google Scholar] [PubMed]
- Zois, C.; Stavrou, I.; Svarna, E.; Seferiadis, K.; Tsatsoulis, A. Natural course of autoimmune thyroiditis after elimination of iodine deficiency in northwestern Greece. Thyroid 2006, 16, 289–293. [Google Scholar] [CrossRef]
- Benvenga, S.; Vigo, M.T.; Metro, D.; Granese, R.; Vita, R.; Le Donne, M. Type of fish consumed and thyroid autoimmunity in pregnancy and postpartum. Endocrine 2016, 52, 120–129. [Google Scholar] [CrossRef]
- Tajtakova, M.; Semanová, Z.; Tomková, Z.; Szökeová, E.; Majoroš, J.; Rádiková, Ž.; Šeböková, E.; Klimeš, I.; Langer, P. Increased thyroid volume and frequency of thyroid disorders signs in schoolchildren from nitrate polluted area. Chemosphere 2006, 62, 559–564. [Google Scholar] [CrossRef]
- Elter, E.; Wagner, M.; Buchenauer, L.; Bauer, M.; Polte, T. Phthalate exposure during the prenatal and lactation period increases the susceptibility to rheumatoid arthritis in mice. Front. Immunol. 2020, 11, 550. [Google Scholar] [CrossRef]
- Ghassabian, A.; Salmon, J.; Karthikraj, R.; Kahn, L.; Mehta-Lee, S.; Buyon, J.; Trasande, L. Phthalate exposure in pregnant women with systemic lupus erythematosus (SLE). Environ. Epidemiol. 2019, 3, 134. [Google Scholar]
- Pryor, W.A.; Stone, K.; Zang, L.Y.; Bermúdez, E. Fractionation of aqueous cigarette tar extracts: Fractions that contain the tar radical cause DNA damage. Chem. Res. Toxicol. 1998, 11, 441–448. [Google Scholar] [CrossRef]
- Robbins, C.S.; Dawe, D.E.; Goncharova, S.I.; Pouladi, M.A.; Drannik, A.G.; Swirski, F.K.; Cox, G.; Stampfli, M.R. Cigarette smoke decreases pulmonary dendritic cells and impacts antiviral immune responsiveness. Am. J. Respir. Cell Mol. Biol. 2004, 30, 202–211. [Google Scholar] [CrossRef]
- Moszczynski, P.; Żabiński, Z.; Moszczyński, P., Jr.; Rutowski, J.; Słowiński, S.; Tabarowski, Z. Immunological findings in cigarette smokers. Toxicol. Lett. 2001, 118, 121–127. [Google Scholar] [CrossRef]
- Hutchinson, D.; Shepstone, L.; Moots, R.; Lear, J.T.; Lynch, M.P. Heavy cigarette smoking is strongly associated with rheumatoid arthritis (RA), particularly in patients without a family history of RA. Ann. Rheum. Dis. 2001, 60, 223–227. [Google Scholar] [CrossRef]
- Chang, K.; Yang, S.M.; Kim, S.H.; Han, K.H.; Park, S.J.; Shin, J.I. Smoking and rheumatoid arthritis. Int. J. Mol. Sci. 2014, 15, 22279–22295. [Google Scholar] [CrossRef]
- Criswell, L.A.; Merlino, L.A.; Cerhan, J.R.; Mikuls, T.R.; Mudano, A.S.; Burma, M.; Folsom, A.R.; Saag, K.G. Cigarette smoking and the risk of rheumatoid arthritis among postmenopausal women: Results from the Iowa Women’s Health Study. Am. J. Med. 2002, 112, 465–471. [Google Scholar] [CrossRef]
- Majka, D.S.; Kim, D.J.; Peerzada, J.; Lockman, S.; Nobles-Knight, D.; Petri, M.; Karlson, E.W. Cigarette smoking and the risk of systemic lupus erythematosus and rheumatoid arthritis. Ann. Rheum. Dis. 2006, 65, 561–563. [Google Scholar] [CrossRef]
- Barbhaiya, M.; Tedeschi, S.K.; Lu, B.; Malspeis, S.; Kreps, D.; Sparks, J.A.; Karlson, E.W.; Costenbader, K.H. Cigarette smoking and the risk of systemic lupus erythematosus, overall and by anti-double stranded DNA antibody subtype, in the Nurses’ Health Study cohorts. Ann. Rheum. Dis. 2018, 77, 196–202. [Google Scholar] [CrossRef]
- Yan Chua, M.H.; Ng, I.A.T.; Mike, W.C.; Mak, A. Association between cigarette smoking and systemic lupus erythematosus- an updated multivariate Bayesian metaanalysis. J. Rheumatol. 2019, 47, 1514–1521. [Google Scholar] [CrossRef]
- Holm, I.A.; Manson, J.E.; Michels, K.B.; Alexander, E.K.; Willett, W.C.; Utiger, R.D. Smoking and Other Lifestyle Factors and the Risk of Graves’ Hyperthyroidism. Arch. Intern. Med. 2005, 165, 1606–1611. [Google Scholar] [CrossRef]
- Hegediüs, L.; Brix, T.H.; Vestergaard, P. Relationship between cigarette smoking and Graves’ ophthalmopathy. J. Endocrinol. Invest. 2004, 27, 265–271. [Google Scholar] [CrossRef]
- Yuksel, N.; Yaman, D.; Pasaoglu, O.T.; Pasaoglu, H. The Effect of Smoking on Mitochondrial Biogenesis in Patients with Graves Ophthalmopathy. Ophthalmic. Plast. Reconstr. Surg. 2020, 36, 172–177. [Google Scholar] [CrossRef] [PubMed]
- Barzilai, O.; Sherer, Y.; Ram, M.; Izhaky, D.; Anaya, J.M.; Shoenfeld, Y. Epstein–Barr Virus and Cytomegalovirus in Autoimmune Diseases. Ann. N. Y. Acad. Sci. 2007, 1108, 567–577. [Google Scholar] [CrossRef] [PubMed]
- Maya, R.; Anaya, J.M.; Barzilai, O.; Izhaky, D.; Katz, B.S.P.; Blank, M.; Shoenfeld, Y. The putative protective role of hepatitis B virus (HBV) infection from autoimmune disorders. Autoimmun. Rev. 2008, 7, 621–625. [Google Scholar]
- Zahiri Yeganeh, S. Bacteriological and Molecular Assessment of Staphylococcal Enterotoxin E in the Blood of Patients with Rheumatoid Arthritis. Jundishapur J. Microbiol. 2015, 8, e59811. [Google Scholar] [CrossRef] [PubMed]
- Carter, J.K.; Smith, R.E. Rapid induction of hypothyroidism by an avian leukosis virus. Infect. Immun. 1983, 40, 795–805. [Google Scholar] [CrossRef] [PubMed]
- Davies, T.F. Infection and Autoimmune Thyroid Disease. J. Clin. Endocrinol. Metab. 2008, 93, 674–676. [Google Scholar] [CrossRef]
- Kivity, S.; Agmon-Levin, N.; Blank, M.; Shoenfeld, Y. Infections and autoimmunity–friends or foes? Trends Immunol. 2009, 30, 409–414. [Google Scholar] [CrossRef]
- Rioux, J.D. Paths to understanding the genetic basis of autoimmune disease. Nature 2005, 435, 584–589. [Google Scholar] [CrossRef]
- Guilherme, L.; Kalil, J.; Cunningham, M. Molecular mimicry in the autoimmune pathogenesis of rheumatic heart disease. Autoimmunity 2006, 39, 31–39. [Google Scholar] [CrossRef]
- Scheinman, R.I.; Cogswell, P.C.; Lofquist, A.K.; Baldwin, A.S. Role of transcriptional activation of I kappa B alpha in mediation of immunosuppression by glucocorticoids. Science 1995, 270, 283. [Google Scholar] [CrossRef]
- Auphan, N.; DiDonato, J.A.; Rosette, C.; Helmberg, A.; Karin, M. Immunosuppression by glucocorticoids: Inhibition of NF-kappa B activity through induction of I kappa B synthesis. Science 1995, 270, 286. [Google Scholar] [CrossRef]
- Rhen, T.; Cidlowski, J.A. Antiinflammatory action of glucocorticoids—New mechanisms for old drugs. N. Engl. J. Med. 2005, 353, 1711. [Google Scholar] [CrossRef]
- Van Staa, T.P.; Leufkens, H.G.; Abenhaim, L.; Begaud, B.; Zhang, B.; Cooper, C. Use of oral corticosteroids in the United Kingdom. QJM 2000, 93, 105–111. [Google Scholar] [CrossRef]
- Zhang, X.W.; Li, Y.; Wang, Z.L.; Li, P. Glucocorticoid receptor subunit gene expression in thyroid gland and adenomas. Acta Oncol. 2006, 45, 1073–1078. [Google Scholar] [CrossRef]
- Kühn, E.R.; Geris, K.L.; van der Geyten, S.; Mol, K.A.; Darras, V.M. Inhibition and activation of the thyroidal axis by the adrenal axis in vertebrates. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 1998, 120, 169–174. [Google Scholar] [CrossRef]
- Williams, D.E.; Chopra, I.J.; Orgiazzi, J.; Solomon, D.H. Acute effects of corticosteroids on thyroid activity in Graves’ disease. J. Clin. Endocrinol. Metab. 1975, 41, 354–361. [Google Scholar] [CrossRef]
- Carroll, R.; Matfin, G. Endocrine and metabolic emergencies: Thyroid storm. Ther. Adv. Endocrinol. Metab. 2010, 1, 139–145. [Google Scholar] [CrossRef]
- Hopkins, R.L.; Leinung, M.C. Exogenous Cushing’s syndrome and glucocorticoid withdrawal. Endocrinol. Metab. Clin. N. Am. 2005, 34, 371–384. [Google Scholar] [CrossRef]
- Paragliola, R.M.; Papi, G.; Pontecorvi, A.; Corsello, S.M. Treatment with Synthetic Glucocorticoids and the Hypothalamus-Pituitary-Adrenal Axis. Int. J. Mol. Sci. 2017, 18, 2201. [Google Scholar] [CrossRef]
- Ferraù, F.; Korbonits, M. Metabolic comorbidities in Cushing’s syndrome. Eur. J. Endocrinol. 2015, 173, 133–157. [Google Scholar] [CrossRef]
- Ferris, H.A.; Kahn, C.R. New mechanisms of glucocorticoid-induced insulin resistance: Make no bones about it. J. Clin. Invest. 2012, 122, 3854–3857. [Google Scholar] [CrossRef]
- Arnaldi, G.; Mancini, T.; Polenta, B.; Boscaro, M. Cardiovascular risk in Cushing’s syndrome. Pituitary 2004, 7, 253–256. [Google Scholar] [CrossRef]
- Dal Bo Zanon, R.; Fornasiero, L.; Boscaro, M.; Cappellato, G.; Fabris, F.; Girolami, A. Increased factor VIII associated activities in Cushing’s syndrome: A probable hypercoagulable state. Thromb. Haemostas. 1982, 47, 116–117. [Google Scholar]
- Canalis, E. Mechanisms of glucocorticoid-induced osteoporosis. Curr. Opin. Rheumatol. 2003, 15, 454–457. [Google Scholar] [CrossRef] [PubMed]
- Chaudhry, H.S.; Singh, G. Cushing Syndrome. Available online: https://www.ncbi.nlm.nih.gov/books/NBK470218/ (accessed on 29 June 2021).
- Schakman, O.; Kalista, S.; Barbé, C.; Loumaye, A.; Thissen, J.P. Glucocorticoid-induced skeletal muscle atrophy. Int. J. Biochem. Cell Biol. 2013, 45, 2163–2172. [Google Scholar] [CrossRef] [PubMed]
- Conn, H.O.; Poynard, T. Corticosteroids and peptic ulcer: Meta-Analysis of adverse events during steroid therapy. J. Intern. Med. 1994, 236, 619–632. [Google Scholar] [CrossRef] [PubMed]
- Conn, H.O.; Blitzer, B.L. Nonassociation of adrenocorticosteroid therapy and peptic ulcer. N. Engl. J. Med. 1976, 294, 473–479. [Google Scholar] [CrossRef] [PubMed]
- Guslandi, M. Steroid ulcers: Any news? World J. Gastrointest. Pharmacol. Ther. 2013, 4, 39–40. [Google Scholar] [CrossRef][Green Version]
- Dorlo, T.P.; Jager, N.G.; Beijnen, J.H.; Schellens, J.H. Concomitant use of proton pump inhibitors and systemic corticosteroids. Ned. Tijdschr. Geneeskd. 2013, 157, A5540. [Google Scholar]
- Black, R.L.; Oglesby, R.B.; von Sallmann, L.; Bunim, J.J. Posterior subcapsular cataracts induced by corticosteroids in patients with rheumatoid arthritis. JAMA 1960, 174, 150–155. [Google Scholar] [CrossRef]
- Van Venrooj, W.J.; Groeneveld, A.A.; Bloemendal, H.; Benedetti, E.L. Cultured calf lens epithelium. The effect of dexamethasone. Exp. Eye Res. 1974, 18, 527–536. [Google Scholar] [CrossRef]
- Alward, W.L. The genetics of open-angle glaucoma: The story of GLC1A and myocilin. Eye 2000, 14, 429–436. [Google Scholar] [CrossRef]
- Polansky, J.R.; Nguyen, T.D. The TIGR gene, pathogenic mechanisms, and other recent advances in glaucoma genetics. Curr. Opin. Ophthalmol. 1998, 9, 15–23. [Google Scholar] [CrossRef]
- Lo, W.R.; Rowlette, L.L.; Caballero, M.; Yang, P.; Hernandez, M.R.; Borrás, T. Tissue differential microarray analysis of dexamethasone induction reveals potential mechanisms of steroid glaucoma. Invest. Ophthalmol. Vis. Sci. 2003, 44, 473–485. [Google Scholar] [CrossRef]
- Davidovici, B.B.; Orion, E.; Wolf, R. Cutaneous manifestations of pituitary gland diseases. Clin Dermatol. 2008, 26, 288–295. [Google Scholar] [CrossRef]
- Lewis, D.A.; Smith, R.E. Steroid-induced psychiatric syndromes: A report of 14 cases and a review of the literature. J. Affect. Disord. 1983, 5, 319–332. [Google Scholar] [CrossRef]
- Warrington, T.P.; Bostwick, J.M. Psychiatric adverse effect of corticosteroids. Mayo Clin. Proc. 2006, 81, 1361–1367. [Google Scholar] [CrossRef]
- Nguyen, D.M.; Yassa, M.A.; Tustison, N.J.; Roberts, J.M.; Kulikova, A.; Nakamura, A.; Ivleva, E.I.; Van Enkevort, E.; Brown, E.S. The Relationship between Cumulative Exogenous Corticosteroid Exposure and Volumes of Hippocampal Subfields and Surrounding Structures. J. Clin. Psychopharmacol. 2019, 39, 653–657. [Google Scholar] [CrossRef]
- Fareau, G.G.; Vassilopoulou-Sellin, R. Hypercortisolemia and infection. Infect. Dis. Clin. N. Am. 2007, 21, 639–657. [Google Scholar] [CrossRef]
- Bakker, R.C.; Gallas, P.R.J.; Romijn, J.A.; Wiersinga, W.M. Cushing’s syndrome complicated by multiple opportunistic infections. J. Endocrinol. Invest. 1998, 21, 329–333. [Google Scholar] [CrossRef]
- Broersen, L.H.; Pereira, A.M.; Jørgensen, J.O.L.; Dekkers, O.M. Adrenal insufficiency in corticosteroids use: Systematic review and meta-analysis. J. Clin. Endocrinol. Metab. 2015, 100, 2171–2180. [Google Scholar] [CrossRef] [PubMed]
- Mebrahtu, T.F.; Morgan, A.W.; Keeley, A.; Baxter, P.D.; Stewart, P.M.; Pujades-Rodriguez, M. Dose dependency of iatrogenic glucocorticoid excess and adrenal insufficiency and mortality: A cohort study in England. J. Clin. Endocrinol. Metab. 2019, 104, 3757–3767. [Google Scholar] [CrossRef] [PubMed]
- Amatruda, T.T.; Hurst, A.M.; D’Esopo, N.D. Certain Endocrine and Metabolic Facets of the Steroid Withdrawal Syndrome. J. Clin. Endocrinol. Metab. 1965, 25, 1207–1217. [Google Scholar] [CrossRef] [PubMed]
- Amatruda, T.T.; Hollingsworth, D.R.; D’esopo, N.D.; Upton, G.V.; Bondy, P.K. A study of the mechanism of the steroid withdrawal syndrome. Evidence for integrity of the hypothalamic-pituitary-adrenal system. J. Clin. Endocrinol. Metab. 1960, 20, 339–354. [Google Scholar] [CrossRef] [PubMed]
- Hochberg, Z.E.; Pacak, K.; Chrousos, G.P. Endocrine withdrawal syndromes. Endocr. Rev. 2003, 24, 523–538. [Google Scholar] [CrossRef] [PubMed]
- Dorn, L.D.; Burgess, E.S.; Friedman, T.C.; Dubbert, B.; Gold, P.W.; Chrousos, G.P. The longitudinal course of psychopathology in Cushing’s syndrome after correction of hypercortisolism. J. Clin. Endocrinol. Metab. 1997, 82, 912–919. [Google Scholar] [PubMed]
- Giordano, R.; Guaraldi, F.; Mazzoli, M.; Ghigo, E. Do glucocorticoids induce addiction in humans? J. Endocrinol. Invest. 2017, 40, 881–883. [Google Scholar] [CrossRef]
- Vittecoq, O.; Desouches, S.; Kozyreff, M.; Nicolau, J.; Pouplin, S.; Rottenberg, P.; Sens, N.; Lequerre, T.; Avenel, G. Relapse in rheumatoid arthritis patients undergoing dose reduction and withdrawal of biologics: Are predictable factors more relevant than predictive parameters? An observational prospective real-life study. BMJ Open 2019, 9, e031467. [Google Scholar] [CrossRef]
- Strehl, C.; Bijlsma, J.W.; de Wit, M.; Boers, M.; Caeyers, N.; Cutolo, M.; Dasgupta, B.; Dixon, W.G.; Geenen, R.; Huizinga, T.W.; et al. Defining conditions where long-term glucocorticoid treatment has an acceptably low level of harm to facilitate implementation of existing recommendations: Viewpoints from an EULAR task force. Ann. Rheum. Dis. 2016, 75, 952–957. [Google Scholar] [CrossRef]
- Thiele, K.; Buttgereit, F.; Huscher, D.; Zink, A.; German Collaborative Arthritis Centres. Current use of glucocorticoids in patients with rheumatoid arthritis in Germany. Arthritis Care Res. 2005, 53, 740–747. [Google Scholar] [CrossRef]
- Mazzantini, M.; Talarico, R.; Doveri, M.; Consensi, A.; Cazzato, M.; Bazzichi, L.; Bombardieri, S. Incident comorbidity among patients with rheumatoid arthritis treated or not with low-dose glucocorticoids: A retrospective study. J. Rheumatol. 2010, 37, 2232–2236. [Google Scholar] [CrossRef]
- Van Staa, T.P.; Leufkens, H.G.; Abenhaim, L.; Zhang, B.; Cooper, C. Use of oral corticosteroids and risk of fractures. J. Bone Miner. Res. 2000, 15, 993–1000. [Google Scholar] [CrossRef]
- Buckley, L.; Guyatt, G.; Fink, H.A.; Cannon, M.; Grossman, J.; Hansen, K.E.; Humphrey, M.B.; Lane, N.E.; Magrey, M.; Miller, M.; et al. American College of Rheumatology Guideline for the Prevention and Treatment of Glucocorticoid-Induced Osteoporosis. Arthritis Rheumatol. 2017, 69, 1521–1537. [Google Scholar] [CrossRef]
- Van Staa, T.P.; Geusens, P.; Pols, H.A.; de Laet, C.; Leufkens, H.G.; Cooper, C. A simple score for estimating the long-term risk of fracture in patients using oral glucocorticoids. QJM 2005, 98, 191–198. [Google Scholar] [CrossRef]
- George, M.D.; Baker, J.F.; Winthrop, K.; Hsu, J.Y.; Wu, Q.; Chen, L.; Xie, F.; Yun, H.; Curtis, J.R. Risk for Serious Infection with Low-Dose Glucocorticoids in Patients With Rheumatoid Arthritis: A Cohort Study. Ann. Intern. Med. 2020, 173, 870–878. [Google Scholar] [CrossRef]
- Listing, J.; Kekow, J.; Manger, B.; Burmester, G.R.; Pattloch, D.; Zink, A.; Strangfeld, A. Mortality in rheumatoid arthritis: The impact of disease activity, treatment with glucocorticoids, TNFα inhibitors and rituximab. Ann. Rheum. Dis. 2015, 74, 415–421. [Google Scholar] [CrossRef]
- Del Rincon, I.; Battaforano, D.F.; Restrepo, J.F.; Erikson, J.M.; Escalante, A. A glucocorticoid dose threshold associated with all-cause and cardiovascular mortality in rheumatoid arthritis. Arthritis Rheum. 2014, 66, 264–272. [Google Scholar] [CrossRef]
- Pincus, T. The clinical efficacy of 3 mg/day prednisone in patients with rheumatoid arthritis: Evidence from a randomized, double-blind, placebo-controlled withdrawal clinical trial. Clin. Exp. Rheumatol. 2011, 29, S73–S76. [Google Scholar]
- Teitelbaum, S.L.; Seton, M.P.; Saag, K.G. Should bisphosphonates be used for long-term treatment of glucocorticoid-induced osteoporosis? Arthritis Rheum. 2011, 63, 325–328. [Google Scholar] [CrossRef]
- Suh, S.; Park, M.K. Glucocorticoid-Induced Diabetes Mellitus: An Important but Overlooked Problem. Endocrinol. Metab. 2017, 32, 180–189. [Google Scholar] [CrossRef]
| System/Organ | Associated Diseases and Comorbidities | 
|---|---|
| Metabolic | Diabetes mellitus and dyslipidemia | 
| Endocrine | Hypogonadism, GH-Deficiency and secondary hypothyroidism | 
| Cardiovascular | Hypertension, hypercoagulability | 
| Bone | Osteoporosis, fractures | 
| Kidney | Nephrolithiasis (calcium) | 
| Muscle | Myopathy | 
| Gastro-Intestinal | Gastritis and peptic ulcers | 
| Eye | Glaucoma and Sub-Capsular cataract | 
| Psyche | Mania, psychosis, depression, and delirium | 
| Immunological | Increased infection rate | 
| Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. | 
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Egalini, F.; Parasiliti Caprino, M.; Gaggero, G.; Cappiello, V.; Giannelli, J.; Rossetto Giaccherino, R.; Pagano, L.; Giordano, R. Endocrine Disorders in Autoimmune Rheumatological Diseases: A Focus on Thyroid Autoimmune Diseases and on the Effects of Chronic Glucocorticoid Treatment. Endocrines 2021, 2, 171-184. https://doi.org/10.3390/endocrines2030018
Egalini F, Parasiliti Caprino M, Gaggero G, Cappiello V, Giannelli J, Rossetto Giaccherino R, Pagano L, Giordano R. Endocrine Disorders in Autoimmune Rheumatological Diseases: A Focus on Thyroid Autoimmune Diseases and on the Effects of Chronic Glucocorticoid Treatment. Endocrines. 2021; 2(3):171-184. https://doi.org/10.3390/endocrines2030018
Chicago/Turabian StyleEgalini, Filippo, Mirko Parasiliti Caprino, Giulia Gaggero, Vincenzo Cappiello, Jacopo Giannelli, Ruth Rossetto Giaccherino, Loredana Pagano, and Roberta Giordano. 2021. "Endocrine Disorders in Autoimmune Rheumatological Diseases: A Focus on Thyroid Autoimmune Diseases and on the Effects of Chronic Glucocorticoid Treatment" Endocrines 2, no. 3: 171-184. https://doi.org/10.3390/endocrines2030018
APA StyleEgalini, F., Parasiliti Caprino, M., Gaggero, G., Cappiello, V., Giannelli, J., Rossetto Giaccherino, R., Pagano, L., & Giordano, R. (2021). Endocrine Disorders in Autoimmune Rheumatological Diseases: A Focus on Thyroid Autoimmune Diseases and on the Effects of Chronic Glucocorticoid Treatment. Endocrines, 2(3), 171-184. https://doi.org/10.3390/endocrines2030018
 
         
                                                

 
       